1
|
Fan Y, Miao W, Yan R, Li C, Wang X. Tannylated lipid nanoparticles for prolonged circulation and PET imaging-guided cancer therapy. BIOMATERIALS ADVANCES 2025; 175:214325. [PMID: 40334338 DOI: 10.1016/j.bioadv.2025.214325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Tannic acid-based nanoplatforms represent a promising approach to overcome current challenges in anticancer drug delivery by enhancing therapeutic efficacy, reducing systemic toxicity, and improving tumor targeting. In this study, we developed a novel nanoparticle system-tannic acid-condensed lipid nanoparticles (TLNPs) through the self-assembly of tannic acid (TA), DSPE-PEG2k, and doxorubicin (DOX). The resulting nanoparticles DOX@TLNPs exhibited a uniform size, satisfactory encapsulation efficiency, excellent stability, and sustained drug release properties. In vitro evaluations demonstrated efficient cellular uptake and comparable cytotoxic activity to free DOX against multiple breast cancer cell lines (SKBR3, MCF-7). In vivo evaluation involved positron emission tomography (PET) imaging of 89Zr-labeled DOX@TLNPs administered to SKBR3 tumor-bearing mice. The nanoparticles showed prolonged circulation and enhanced tumor accumulation, evidenced by a significantly higher area under the curve compared to free DOX, likely due to the enhanced permeability and retention effect. Furthermore, antitumor studies revealed that DOX@TLNPs markedly inhibited tumor growth, improved survival rates, and induced increased apoptosis alongside reduced proliferation within tumor tissues, without eliciting significant histopathological changes in major organs. These findings highlight the significant potential of TA-condensed TLNPs as a safe, robust, and effective nanomedicine platform for targeted cancer treatment.
Collapse
Affiliation(s)
- Yeli Fan
- School of Environmental Science and Engineering, Wuxi University, Wuxi 214105, PR China.
| | - Wujun Miao
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Rusong Yan
- School of Environmental Science and Engineering, Wuxi University, Wuxi 214105, PR China
| | - Changyuan Li
- School of Environmental Science and Engineering, Wuxi University, Wuxi 214105, PR China
| | - Xinyu Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China.
| |
Collapse
|
2
|
Xu L, Wang Y, Hu Y, Dai X, Sun C, Cheng J. ROS-responsive oridonin and dihydroartemisinin hetero-polymeric prodrug NPs for potentiating ferroptosis in gastric cancer by disrupting redox balance. Colloids Surf B Biointerfaces 2025; 252:114637. [PMID: 40132335 DOI: 10.1016/j.colsurfb.2025.114637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Gastric cancer presents a significant global health concern, with conventional therapies often limited in effectiveness. The abnormal redox balance in gastric cancer cells may represent a breakthrough in the treatment of gastric cancer. In this study, we report for the first time the development of reactive oxygen species (ROS)-sensitive hetero-polymeric prodrug nanoparticles (NPs) designed for the co-delivery of the Chinese herbal extract oridonin (ORI) and dihydroartemisinin (DHA) in combination therapy for gastric cancer. This strategy aims to disrupt the intracellular redox balance and ultimately induce ferroptosis in gastric cancer cells. The ROS-responsive ORI and DHA polymeric prodrug were synthesised by conjugating ORI or DHA to poly(ethylene glycol)-block-poly(L-lysine) (PEG-b-PLL) via a ROS-sensitive linker thioketal (TK). The resulting polymeric prodrugs self-assemble in water to form NPs OD-M. After internalization by gastric cancer cells, OD-M released ORI and DHA in response to high ROS conditions within cancer cells. The released ORI reacts with GSH to induce GSH depletion while DHA amplifies intracellular ROS levels, ultimately inducing ferroptosis in gastric cancer cells. Experimental results demonstrate that OD-M acts as both a GSH scavenger and ROS generator, effectively disrupting intracellular redox balance, inducing ferroptosis, and exhibiting effective anticancer efficacy in vitro and in vivo, offering a departure from traditional methods.
Collapse
Affiliation(s)
- Luzhou Xu
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| | - Yan Wang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Yanqin Hu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinyi Dai
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Sun
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun Cheng
- Jiangsu Hongdian Chinese Medicine Industry Research Institute, Nanjing 210042, China
| |
Collapse
|
3
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 PMCID: PMC12067981 DOI: 10.1016/j.addr.2025.115579] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
Huang X, Hou S, Li Y, Xu G, Xia N, Duan Z, Luo K, Tian B. Targeting lipid metabolism via nanomedicine: A prospective strategy for cancer therapy. Biomaterials 2025; 317:123022. [PMID: 39754967 DOI: 10.1016/j.biomaterials.2024.123022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025]
Abstract
Lipid metabolism has been increasingly recognized to play an influencing role in tumor initiation, progression, metastasis, and therapeutic drug resistance. Targeting lipid metabolic reprogramming represents a promising therapeutic strategy. Despite their structural complexity and poor targeting efficacy, lipid-metabolizing drugs, either used alone or in combination with chemotherapeutic agents, have been employed in clinical practice. The advent of nanotechnology offers new approaches to enhancing therapeutic effects, includingthe targeted delivery and integration of lipid metabolic reprogramming with chemotherapy, photodynamic therapy (PDT), and immunotherapy. The integrated nanoformulation, nanomedicine, could significantly advance the field of lipid metabolism therapy. In this review, we will briefly introduce the concept of cancer lipid metabolism reprogramming, then elaborate the latest advances in engineered nanomedicine for targeting lipid metabolism during cancer treatment, and finally provide our insights into future perspectives of nanomedicine for interference with lipid metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
- Xing Huang
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shengzhong Hou
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinggang Li
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Xu
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Ning Xia
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Duan
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Kui Luo
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, Key Laboratory of Transplant Engineering and Immunology, NHC, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China.
| | - Bole Tian
- Division of Pancreatic Surgery, Department of General Surgery, Department of Radiology, Huaxi MR Research Center (HMRRC), Liver Transplant Center, Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Zhu M, Jia J, Tang H, Xie Y, Lv Z, Bao H, Zhang Y, Miao D, Guo X, Chen K, Wang S, Yu L, Pei J. Antitumor Efficacy, Pharmacokinetics, and Toxicity Studies of Novel Estrogen Receptors Targeted PEGylated Liposomes Encapsulating Paclitaxel and Cisplatin in SKOV-3 Tumor-Bearing Nude Mice, ICR Mice, and SD Rats. Mol Pharm 2025. [PMID: 40415642 DOI: 10.1021/acs.molpharmaceut.4c01457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Ovarian malignancies overexpress estrogen receptors (ERs), offering a therapeutic avenue for targeted drug delivery. Here, we developed a novel ER-targeted PEGylated liposome (ES-SSL-PTX/CDDP) coencapsulating paclitaxel (PTX) and cisplatin (CDDP) to enhance therapeutic efficacy and mitigate systemic toxicity. ES-SSL-PTX/CDDP exhibited a spherical shape with a hydrodynamic diameter of around 150 nm, a negative zeta potential of about -20 mV, and high encapsulation efficiencies of 83.7% for PTX and 41.1% for CDDP. ES-SSL-PTX/CDDP showed a sustained release pattern, with release rates of <60% for both drugs within 12 h. Meanwhile, ES-SSL-PTX/CDDP demonstrated excellent storage and serum stability, with the leakage rates less than 30% when stored at either 4 or 25 °C. ES-SSL-PTX/CDDP exhibited a strong antitumor effect in athymic mice with the tumor volume 8.50 times smaller than that in the control group on the 27th day, and the tumor inhibition rate reached 87.3%. Pharmacokinetic studies revealed prolonged circulation of ES-SSL-PTX/CDDP, with elimination half-lives (t1/2β) of 13.84 h (PTX) and 7.18 h (CDDP), which were 8.82- and 1.83-fold higher than those of PTX/CDDP, and clearance rates reduced to 0.01 L/h/kg (PTX) and 0.02 L/h/kg (CDDP), being 18.0- and 4.0-fold lower than those of PTX/CDDP. Acute toxicity results demonstrated a 2.12-fold increase in the LD50 of ES-SSL-PTX/CDDP (27.82 mg/kg for PTX; 19.87 mg/kg for CDDP) versus PTX/CDDP. Long-term toxicity studies demonstrated that ES-SSL-PTX/CDDP attenuated myelosuppression and nephrotoxicity, with no histopathological abnormalities observed across 32 tissues after 16 weeks of administration. This study highlights the potential of ES-SSL-PTX/CDDP to improve the efficacy and reduce the toxicity of platinum-taxane regimens in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Juan Jia
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Huan Tang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhe Lv
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yan Zhang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Kejia Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Shanshan Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Liangping Yu
- Department of Clinical Pharmacy, The First Hospital of Jilin University, Changchun 130061, China
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
6
|
Yang D, Zhong D, Wang R, Hu Q, Wei P, Tong A, Wang Z, He C, Zhang J, Hu H, Zhou M. Manganese-Based Natural Photosensitive Protein Nanocomplex for Image-Guided Multimodal Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29248-29265. [PMID: 40329511 DOI: 10.1021/acsami.5c03372] [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: 05/08/2025]
Abstract
Photodynamic therapy (PDT) is widely utilized in cancer treatment as a noninvasive strategy. Phycocyanin (PC), a natural water-soluble photosensitizer with nontoxic properties, shows promise as a PDT candidate. However, the limitations of PC when used alone in PDT for cancer treatment, such as inadequate tumor delivery and weak efficacy, must be addressed. Herein, an efficient theranostic manganese (Mn)-based PC nanocomplex (PC@Mn) was synthesized through a straightforward one-pot self-assembly reaction for synergistic antitumor therapy. The PC@Mn nanoparticles were found to have a suitable size (∼129 nm) and demonstrated excellent biocompatibility and biosafety. Importantly, these nanoparticles exhibited enhanced biodistribution with improved tumor targeting and retention properties. When combined with 650 nm laser irradiation, PC@Mn showed a significant enhancement in the PDT effect in vivo. Additionally, PC@Mn displayed promising magnetic resonance (MR) imaging capabilities, with a high relaxation rate (r1 = 10.14 mM-1 s-1) and an extended imaging time window (4 h). This feature enables real-time monitoring of the nanoparticles' distribution within tumors, facilitating precise determination of the optimal PDT treatment time. Overall, the study highlights PC@Mn as a simple, safe, and highly efficient strategy for synergistic antitumor therapy. Its ability to combine PDT with MR imaging for real-time guidance represents an efficient approach to cancer treatment, promising improved therapeutic outcomes and potential clinical applications in the future.
Collapse
Affiliation(s)
- Di Yang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
- Department of Radiology, Sir Run Run Shaw Hospital (SRRSH) of School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
- Department of Radiology, Zhejiang Hospital, Hangzhou 310013, China
- Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Zhejiang University, Haining 314400, China
| | - Danni Zhong
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Ruoxi Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Qiuhui Hu
- Department of Radiology, Sir Run Run Shaw Hospital (SRRSH) of School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Peiying Wei
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China
| | - Aiying Tong
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Ziwei Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Chengbin He
- Department of Radiology, Sir Run Run Shaw Hospital (SRRSH) of School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Jianjun Zhang
- Department of Radiology, Zhejiang Hospital, Hangzhou 310013, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital (SRRSH) of School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310000, China
- Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Zhejiang University, Haining 314400, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining 314400, China
- The National Key Laboratory of Biobased Transportation Fuel Technology, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
7
|
Gao S, Zhou JR, Yokomizo K, Fang J. Nano-drug delivery system of natural products for disease prevention and treatment. Expert Opin Drug Deliv 2025:1-11. [PMID: 40366774 DOI: 10.1080/17425247.2025.2506830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/22/2025] [Accepted: 05/12/2025] [Indexed: 05/16/2025]
Abstract
INTRODUCTION Natural products, derived from plants, animals, and microorganisms, offer a wide range of pharmacological activities, including anti-infective, antifungal, anti-tumor, cholesterol-lowering, and anti-inflammatory effects. However, their clinical use is often limited by challenges such as low stability, poor bioavailability, and short half-lives. Thus, developing effective drug delivery systems for these compounds is crucial. AREAS COVERED This review highlights the integration of natural products with nano-drug delivery systems, focusing on recent advancements that utilize the enhanced permeability and retention (EPR) effect to improve their stability, bioavailability, and targeting. By embedding natural compounds into polymeric nanoparticles or similar nanoplatforms, these formulations significantly enhance pharmacokinetic and pharmacodynamic properties, overcoming traditional limitations. EXPERT OPINION Combining natural products with nanoparticle technology shows great potential to expand their therapeutic applications. Although these innovations improve the pharmacological profiles of natural compounds, continued research is essential to optimize clinical use. Advances in nanoparticle design and delivery strategies will be key to maximizing the therapeutic potential of natural products, addressing existing challenges, and enhancing their efficacy in disease treatment.
Collapse
Affiliation(s)
- Shanghui Gao
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Jian-Rong Zhou
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Kazumi Yokomizo
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Jun Fang
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| |
Collapse
|
8
|
Wang X, Xiong X. Mitochondrial Reactive Oxygen Species (mROS) Generation and Cancer: Emerging Nanoparticle Therapeutic Approaches. Int J Nanomedicine 2025; 20:6085-6119. [PMID: 40385494 PMCID: PMC12085131 DOI: 10.2147/ijn.s510972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 04/24/2025] [Indexed: 05/20/2025] Open
Abstract
Mitochondrial reactive oxygen species (mROS) are generated as byproducts of mitochondrial oxidative phosphorylation. Changes in mROS levels are involved in tumorigenesis through their effects on cancer genome instability, sustained cancer cell survival, metabolic reprogramming, and tumor metastasis. Recent advances in nanotechnology offer a promising approach for precise regulation of mROS by either enhancing or depleting mROS generation. This review examines the association between dysregulated mROS levels and key cancer hallmarks. We also discuss the potential applications of mROS-targeted nanoparticles that artificially manipulate ROS levels in the mitochondria to achieve precise delivery of antitumor drugs.
Collapse
Affiliation(s)
- Xinyao Wang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
- Queen Mary School of Nanchang University, Nanchang, People’s Republic of China
| | - Xiangyang Xiong
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
9
|
Dirheimer L, Cortese S, Dolivet G, Merlin JL, Marchal F, Mastronicola R, Bezdetnaya L. Fluorescence Imaging-Assessed Surgical Margin Detection in Head and Neck Oncology by Passive and Active Targeting. Mol Diagn Ther 2025:10.1007/s40291-025-00781-x. [PMID: 40342044 DOI: 10.1007/s40291-025-00781-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/11/2025]
Abstract
Surgery remains the gold standard in the management of head and neck squamous cell carcinoma (HNSCC). However, the anatomical complexity of these cancers, combined with the difficulty in discriminating between healthy and cancerous tissue and the detection of microlesions, complicates tumor resection, resulting in positive surgical margins, which are associated with a poor patient prognosis. Fluorescence-guided surgery (FGS) has emerged as a promising technique in the management of HNSCC, improving tumor resection and margin assessment. FGS strategies can be roughly divided into three approaches; namely, natural tissue autofluorescence, passive delivery of fluorescent contrast agents, and active targeting. This review provides a comprehensive overview of the advances made in FGS of head and neck cancers, particularly aiming to improve surgical margin assessment. Recently, the field has shown promising results by addressing contrast agents targeted to the overexpressed epidermal growth factor receptor (EGFR), both in preclinical and clinical settings. The identification of new targets such as αVβ6 integrin, uPAR, PARP1, and so on, as well as the development of contrast agents, are key steps in the further development of FGS of head and neck cancers, making it an essential tool in precision oncology. Among these, as was demonstrated in preclinical studies, the αVβ6 integrin is emerging as a promising target due to its high and specific expression in tumor and tumor margins.
Collapse
Affiliation(s)
- L Dirheimer
- Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy, France
| | - S Cortese
- Surgical Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy, France
| | - G Dolivet
- Surgical Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy, France
- Université de Lorraine, CNRS, CRAN, 54000, Nancy, France
| | - J L Merlin
- Université de Lorraine, CNRS, CRAN, 54000, Nancy, France
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN-Université de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy Cedex, France
| | - F Marchal
- Surgical Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy, France
- Université de Lorraine, CNRS, CRAN, 54000, Nancy, France
| | - R Mastronicola
- Surgical Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy, France
- Université de Lorraine, CNRS, CRAN, 54000, Nancy, France
| | - L Bezdetnaya
- Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandœuvre-lès-Nancy, France.
- Université de Lorraine, CNRS, CRAN, 54000, Nancy, France.
| |
Collapse
|
10
|
Liu X, Liu C, Wu M, Cao L, Lu C, Liu B. Donor Optimizing to Boost Type I and Type II Photosensitization for Solid Tumor Therapy. Adv Healthc Mater 2025; 14:e2500726. [PMID: 40171747 DOI: 10.1002/adhm.202500726] [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: 02/08/2025] [Revised: 03/12/2025] [Indexed: 04/04/2025]
Abstract
Oxygen-less dependent Type I photosensitizers (PSs) have emerged as a crucial strategy for enhancing photodynamic therapy efficiency in treating hypoxic tumors. However, solid tumors have normoxia regions situated near functional blood vessels and hypoxia regions in their interiors. To maximize the utilization of oxygen within solid tumors, herein a viable donor optimizing approach is developed to enhance both Type I&II reactive oxygen species generation of PSs. At the same mole concentration, one optimized PS (named DE) generated 9 times more 1O2 than commercial Type II PS Chlorin e6 upon white light irradiation for 60 s. Compared to the commercial Type I PS Rose Bengal, •OH generation by DE is 2.9 times more under the hypoxia condition. With its optimized Type I&II pathway under normoxia and hypoxia conditions, DE is proven to be an efficient PS for solid tumor treatment, offering a promising approach for PS development.
Collapse
Affiliation(s)
- Xingang Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Chuang Liu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Min Wu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
| | - Lei Cao
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, 350207, China
| |
Collapse
|
11
|
Yu X, Zhang Q, Wang L, Zhang Y, Zhu L. Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma. Exp Hematol Oncol 2025; 14:62. [PMID: 40307921 PMCID: PMC12044934 DOI: 10.1186/s40164-025-00658-z] [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: 01/19/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Liver cancer, notably hepatocellular carcinoma (HCC), poses a significant global health burden due to its high fatality rates. Conventional antitumor medications face challenges, including poor targeting, high toxicity, and drug resistance, leading to suboptimal clinical outcomes. This review focused on nanoparticle use in diagnosing and delivering medication for HCC, aiming to advance the development of nanomedicines for improved treatment outcomes. As an emerging frontier science and technology, nanotechnology has shown great potential, especially in precision medicine and personalized treatment. The success of nanosystems is attributable to their smaller size, biocompatibility, selective tumor accumulation, and lower toxicity. Nanoparticles, as a central part of nanotechnology innovation, have emerged in the field of medical diagnostics and therapeutics to overcome the various limitations of conventional chemotherapy, thus offering promising applications for improved selectivity, earlier and more precise diagnosis of cancers, personalized treatment, and overcoming drug resistance. Nanoparticles play a crucial role in drug delivery and imaging of HCC, with the body acting as a delivery system to target and deliver drugs or diagnostic reagents to specific organs or tissues, helping to accurately diagnose and target therapies while minimizing damage to healthy tissues. They protect drugs from early degradation and increase their biological half-life.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, No. 10 Qinyun Nan Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qin Zhang
- Department of Postgraduate Students, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Leibo Wang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital Guiyang, Guiyang, 550000, Guizhou, The People's Republic of China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
12
|
Yin X, Zhao X, Shen Y, Xie W, He C, Guo J, Li Z, Xuan F, Zeng S, Zeng X, Fang C. Nanoparticle-mediated dual targeting of stromal and immune components to overcome fibrotic and immunosuppressive barriers in hepatocellular carcinoma. J Control Release 2025; 383:113783. [PMID: 40306574 DOI: 10.1016/j.jconrel.2025.113783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are key drivers of hepatocellular carcinoma (HCC) through their promotion of fibrosis and immune suppression activity. To overcome this stroma-immune barrier, we developed D/F@MRL, a stroma-immune co-targeting nanoplatform that enables the spatiotemporal coordination of CAF reprogramming and immune activation. In D/F@MRL, MMP-2-responsive hybrid liposomes (MRL) was employed to co-load digoxin (Dig) and PD-L1-degrading nanofibers (NFs). Upon encountering the MMP-2-enriched HCC stroma, D/F@MRL undergoes enzymatic cleavage, thereby enabling the targeted release of Dig and NFs within the HCC microenvironment. Mechanistically, Dig inhibits the phosphorylation of SMAD3 in CAFs, while PD-L1 degradation destabilizes the TGFβ receptor, synergistically silencing TGF-β/Smad signaling to reprogram CAFs. This combination not only disrupts the fibrotic barrier but also creates a feed-forward loop that further enhances drug penetration, while reinforcing the immune activation driven by Dig-induced immunogenic cell death (ICD) and PD-L1 degradation. In the humanized immune PDX model, D/F@MRL successfully reprogrammed CAFs and robustly remodeled the stromal and immune microenvironments without causing systemic toxicity, highlighting its promising potential for clinical translation. By integrating CAF reprogramming with ICD and immune checkpoint inhibition, this strategy overcame the limitations of single-target therapies, induced robust immune activation, further provided a clinic-transformative approach for fibrotic malignancies.
Collapse
Affiliation(s)
- Xiangyi Yin
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xingyang Zhao
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiming Shen
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weizhong Xie
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Cheng He
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianan Guo
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zirong Li
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Feichao Xuan
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Silue Zeng
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Zeng
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chihua Fang
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Institute of Digital Intelligent Minimally Invasive Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; South China Institute of National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Guangzhou 510280, China.
| |
Collapse
|
13
|
Sun J, Li HL, Zhou WJ, Ma ZX, Huang XP, Li C. Current status and recent progress of nanomaterials in transcatheter arterial chemoembolization therapy for hepatocellular carcinoma. World J Clin Oncol 2025; 16:104435. [PMID: 40290691 PMCID: PMC12019268 DOI: 10.5306/wjco.v16.i4.104435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Transcatheter arterial chemoembolization has become a common treatment modality for some patients with unresectable advanced HCC. Since the introduction of nanomaterials in 1974, their use in various fields has evolved rapidly. In medical applications, nanomaterials can serve as carriers for the delivery of chemotherapeutic drugs to tumour tissues. Additionally, nanomaterials have potential for in vivo tumour imaging. This article covers the properties and uses of several kinds of nanomaterials, focusing on their use in transcatheter arterial chemoembolization for HCC treatment. This paper also discusses the limitations currently associated with the use of nanomaterials.
Collapse
Affiliation(s)
- Jia Sun
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Hai-Liang Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Wen-Jun Zhou
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Zeng-Xin Ma
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Xiao-Pei Huang
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Cheng Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| |
Collapse
|
14
|
Liu F, Li S, Huang C, Bi Z, Xiang X, Zhang S, Yang R, Zheng L. Self-assembled nanoplatform-mediated co-delivery of brusatol to sensitize sorafenib for hepatocellular carcinoma treatment. RSC Adv 2025; 15:11675-11687. [PMID: 40230634 PMCID: PMC11995455 DOI: 10.1039/d5ra00108k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Sorafenib (Sor), recognized as a frontline multi-kinase inhibitor, constitutes the primary targeted therapy for hepatocellular carcinoma (HCC). Despite its potential, many HCC patients exhibit reduced responsiveness to Sor, thereby undermining its therapeutic efficacy. Recent studies highlight the importance of nuclear factor erythroid-2-related factor 2 (Nrf2) activation in HCC, which contributes to Sor resistance. Brusatol (Bru), a plant-derived Nrf2 inhibitor, counteracts this resistance but faces challenges due to its poor solubility in aqueous media. In this study, we developed a glutathione (GSH)-responsive nanoplatform that effectively dispersed in water for the co-delivery of Bru and Sor (B/S NP). This approach enhanced Bru's therapeutic efficacy and increased Sor sensitivity in HCC. Our nanoplatform significantly reduced Nrf2 expression, thereby increasing Sor sensitivity both in vitro and in vivo, while presenting a favorable biosafety profile. These findings suggest that the nanoplatform-mediated co-delivery of Bru and Sor offers an innovative approach to enhance Sor's effectiveness in HCC treatment.
Collapse
Affiliation(s)
- Fengrui Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
- Key Laboratory of Tongliang District People's Hospital Chongqing 402560 P. R. China
| | - Senlin Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| | - Chengcheng Huang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| | - Zhenfei Bi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| | - Xiao Xiang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| | - Shuqi Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| | - Ruihao Yang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| | - Lu Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University Chongqing 400038 P. R. China
| |
Collapse
|
15
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025; 219:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75123, Sweden.
| |
Collapse
|
16
|
Lei D, Wang W, Zhao J, Zhou Y, Chen Y, Dai J, Qiu Y, Qi H, Li C, Liang B, Liu B, Wang Q, Li R. An injectable gambogic acid loaded nanocomposite hydrogel enhances antitumor effect by reshaping immunosuppressive tumor microenvironment. Mater Today Bio 2025; 31:101611. [PMID: 40104652 PMCID: PMC11919334 DOI: 10.1016/j.mtbio.2025.101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Gambogic acid(GA)is a natural compound that exhibits strong antitumor activity against a variety of tumors. However, its poor water solubility, low specificity, and high toxicity lead to inevitable systemic adverse effects. To minimize side effects, combining gambogic acid (GA) with delivery systems such as nanohydrogels to develop an in situ vaccine system (ISV) shows great promise. In this study, we loaded GA into a novel in situ nanocomposite hydrogel vaccine system (Gel-NPs@GA) along with a near-infrared (NIR) fluorescent dye, IR-1061. The Gel-NPs@GA system allowed for temperature-triggered gelation, simplifying injection and the in vivo formation of a drug-releasing gel, with near-infrared monitoring for drug metabolism. Slow, continuous release of gelatinase-targeted GA nanoparticles from the hydrogel occurs, followed by cleavage of mPEG-peptide-PCL conjugates by gelatinase, causing particle aggregation for endocytosis by tumor cells. This approach tackled solubility issues and curbs excessive GA release, boosting therapeutic drug levels. The sustained GA release induces tumor cell apoptosis, releasing tumor antigens and reprogramming the immune-suppressive tumor microenvironment. In the CT26 colorectal cancer mice model, this in situ vaccine system significantly inhibited tumor growth. By integrating information about immune cell clusters within the tumor microenvironment with RNA sequencing results, we hypothesized that Gel-NPs@GA could synergistically stimulate the immune response through various pathways, promote the maturation of dendritic cells (DCs), increase the infiltration of T cells, and thereby remodel the tumor's immune microenvironment.
Collapse
Affiliation(s)
- Dan Lei
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wanru Wang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianhang Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Yingling Zhou
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Chen
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Juanjuan Dai
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuling Qiu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haoyue Qi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chunhua Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Baorui Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| | - Qin Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| | - Rutian Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| |
Collapse
|
17
|
Chen S, Huang H, Li Q, Cai J, Miao Z, Xie P, Tang S, He D. Carrier-free nanoparticles based on self-assembly of 5-FU and copper-genistein complexes for the combined treatment of hepatocellular carcinoma. Drug Deliv Transl Res 2025; 15:1299-1316. [PMID: 39126575 DOI: 10.1007/s13346-024-01676-w] [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] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
5-Fluorouracil (5-FU) is commonly used as a chemotherapeutic drug for advanced HCC. However, the effectiveness of 5-FU is limited by the emergence of resistance and poor targeting efficiency. Combining 5-FU with natural compounds has shown promise in HCC treatment. In this study, we prepared carrier-free nanoparticles (GEN-Cu-GEN@FUA) containing 5-FU and genistein (GEN) in a synergistic ratio via a green synthesis procedure. The resulting GEN-Cu-GEN@FUA nanoparticles had a spherical or near spherical shape, a dynamic size of 129.3 ± 40.1 nm, and a high drug loading content of approximately 21.40% (5-FU) and 61.48% (GEN). These nanoparticles exhibited approximately 3.6-fold lower IC50 value than 5-FU alone in Bel-7402 cells and resulted in a 3.7-fold greater reduction in tumor weight compared to 5-FU alone in Bel-7402 tumor-bearing BALB/c mice. Importantly, the nanoparticles showed negligible systemic toxicity due to their synergistic effect on cancer cell dysfunction and significant amplification of intracellular glutathione consumption. Our findings suggest that the developed carrier-free nanomedicines offer a highly promising platform for the co-delivery of genistein (GEN) copper(II) complexes and 5-FU, with easy fabrication and great potential for clinical translation in HCC synergistic therapy.
Collapse
Affiliation(s)
- Siwei Chen
- Miluo Maternity and Child Care Hospital, Yueyang, Hunan Province, China
| | - Hongwu Huang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Qi Li
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Jia Cai
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Zhuolin Miao
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery Systemy, Huaihua, Hunan, China
| | - Peikang Xie
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery Systemy, Huaihua, Hunan, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery Systemy, Huaihua, Hunan, China.
| | - Dongxiu He
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
18
|
Mohamed N, Ismail H, Nasr GM, Abdel-Ghany S, Arneth B, Sabit H. Anti-Tumor Potential of Frankincense Essential Oil and Its Nano-Formulation in Breast Cancer: An In Vivo and In Vitro Study. Pharmaceutics 2025; 17:426. [PMID: 40284420 PMCID: PMC12030047 DOI: 10.3390/pharmaceutics17040426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objective: Breast cancer remains the most common malignancy among women worldwide, contributing to high morbidity and mortality rates. Many anti-cancer drugs have been derived from medicinal plants, and frankincense from Boswellia carterii is notable for its anti-inflammatory, anti-neoplastic, and anti-carcinogenic properties. Using gas chromatography/mass spectrometry (GC/MS), 48 components were identified in B. carterii essential oil, and the major constituent was α-pinene (35.81%). Method: In this study, we investigated the anti-tumor effects of frankincense essential oil (FEO) and its nano-formulation with chitosan (FEO-CSNPs) using in vitro breast cancer models (MCF-7, MDA-MB-231, and 4T1 cells) and in vivo mouse mammary carcinoma (4T1) models (Balb/c). Results: The results showed significant reductions in cell viability. At 10 μg/mL, the FEO showed the highest reduction in the C-166 cells, while at 100 μg/mL, the FEO exhibited a stronger cytotoxicity in the MDA-MB-231 and 4T1 cells compared to the FEO-CSNPs and CSNPs. The FEO-CSNPs exhibited cell growth arrest in the S, G2/M, and G1/S phases in the MCF-7, MDA-MB-231, and 4T1 cell lines (36.91%, 23.12%, and 33.58%), in addition to increased apoptosis rates in the MCF-7, MDA-MB-231, and 4T1 cell lines (33.04%, 36.39%, and 42.19%). The wound healing assays revealed a decreased migratory ability in the treated cells. The in vivo experiments in the balb/c mice demonstrated a reduction in the tumor volume, with a histopathological analysis confirming extensive tumor necrosis. Moreover, the FEO and FEO-CSNPs showed notable antioxidant and arginase activity. The gene expression analysis via qPCR indicated the upregulation of tumor suppressor genes and the downregulation of oncogenes. Conclusions: These findings suggest that FEO and its nano-formulation, particularly in the form of FEO-CSNPs as an oral formulation, display enhanced efficacy, warranting further preclinical and clinical research to develop innovative treatment strategies.
Collapse
Affiliation(s)
- Nouran Mohamed
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Hisham Ismail
- Department of Molecular Diagnostics, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, El Sadat City 32897, Menofia, Egypt
| | - Ghada M. Nasr
- Department of Molecular Diagnostics, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, El Sadat City 32897, Menofia, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldinger Str., 35043 Marburg, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| |
Collapse
|
19
|
Liu Y, Xie Y, Chen Y, Duan J, Bao C, Wang J, Feng H, Wang M, Ren Y, Li P, Luo Q, Xu J, Jiang M, Men Y, Wu Y, Li J, Wang G, Lu W. A protease-cleavable liposome for co-delivery of anti-PD-L1 and doxorubicin for colon cancer therapy in mice. Nat Commun 2025; 16:2854. [PMID: 40128211 PMCID: PMC11933685 DOI: 10.1038/s41467-025-57965-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Immune checkpoint blockade therapy using programmed cell death 1 (PD1) or programmed death ligand 1 (PD-L1) has made significant progress in the treatment of advanced cancers, with some patients achieving long-term remission without clinical recurrence. However, only a minority of colon cancer patients respond to the therapy. Here, we report a protease-cleavable anti-PD-L1 antibody liposome, eLipo anti-PD-L1, for enhancing colon cancer therapy. In vivo, eLipo anti-PD-L1 is cleaved by legumain at colon cancer site into pegylated anti-PD-L1 and cancer-homing doxorubicin liposome. Functional assessments show cancer-targeting, legumain-responding, tumor-penetrating, and immune-activating effects, as well as efficacy in treating colon cancer-bearing mice in vivo. Further mechanistic analysis implicates genes related to T cell differentiation and T cell receptor signaling as potential molecular mediators. Lastly, human colorectal cancer tissue evaluations verify expressions of PD-L1 and legumain, hinting a potential translatability. Our study thus suggests that eLipo anti-PD-L1 may be a feasible vector for co-delivery of immunochemotherapy for colon cancer.
Collapse
Affiliation(s)
- Yixuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jialun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Chunjie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jinling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Hexuan Feng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Mengjie Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuxin Ren
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Peishan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jiarui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Min Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yanchen Men
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yang Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Guiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Wanliang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
| |
Collapse
|
20
|
Yao B, Xu F, Tian Z, Dai M, Song J, Li L, Liu X, Lu H, Zhang W. Strong Persistent Luminescence NaYF 4-based Nanoparticles Combined with Manipulated Hyperfractionated Irradiation for X-ray-Excited Photodynamic Therapy Enhancement. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16561-16575. [PMID: 40042361 DOI: 10.1021/acsami.4c20049] [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: 03/21/2025]
Abstract
X-ray-excited photodynamic therapy (X-PDT), a novel synergistic therapy combining radiotherapy (RT) with photodynamic therapy (PDT), demonstrates not only more effective therapeutic outcomes but also overcomes the limitation of PDT's shallow penetration depth. Persistent luminescence nanoparticles (PLNPs) have been employed in X-PDT due to their unique afterglow emission, which yields more light to achieve more effective PDT outcomes using the same irradiation dose. However, at present, persistent luminescent materials used in X-PDT are mainly bulk crystals characterized by a nonuniform size and morphology, which are not suitable for biomedical applications, and the presence of excessive surface defects reduces the luminescence efficiency and the persistent luminescence duration. Herein, the NaYF4:Tb@NaYF4 core-shell nanoparticles with enhanced luminescence and afterglow performance and uniform morphology were prepared via the optimized solvothermal method. Their X-ray excitation optical luminescence (XEOL) and persistent luminescence (XEPL) intensities were enhanced more than 5.2 times and 3.5 times, respectively. The PLNPs were modified with a water-soluble AEP ligand and piggybacked with the photosensitizer Rose Bengal (RB) to construct an efficient X-PDT nanocoupling system. To fully utilize the afterglow of PLNPs, a unique hyperfractionated irradiation plan was designed, and the ROS yield was increased by nearly 50% at the same irradiation dose. In vivo therapeutic efficacy validation using the B16-F10-bearing C57 mouse model demonstrated that hyperfractionated irradiation combined with PLNPs showed significant therapeutic advantages. At a total dose of 2 Gy, the tumor inhibition rate was enhanced from 67.5% to 85% compared to the conventional irradiation strategy. Pathological analysis showed no significant histological damage in major organs, attesting to its negligible side effects. This study offers a novel modality, with both nanoparticles and irradiation strategy improvement, to further improve the X-PDT therapeutic efficacy and reduce side effects.
Collapse
Affiliation(s)
- Bang Yao
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Air Force Medical University, Xi'an, Shaanxi 710032, China
- School of Materials Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Fanyuan Xu
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Zuhong Tian
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Mengyan Dai
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Jiadan Song
- Department of Engineering Physics, Beijing & Key Laboratory of Particle and Radiation Imaging, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Liang Li
- Department of Engineering Physics, Beijing & Key Laboratory of Particle and Radiation Imaging, Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Xiaoxu Liu
- School of Materials Science and Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi 710021, China
| | - Hongbing Lu
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Wenli Zhang
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Air Force Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
21
|
Chattopadhyay S, Goswami A, Sil M. Nanobiotechnology: traditional re-interpreting personalized medicine through targeted therapies and regenerative solutions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04038-6. [PMID: 40100374 DOI: 10.1007/s00210-025-04038-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
Nanobiotechnology is transforming personalized medicine by leveraging the unique properties of nanomaterials to address key challenges in targeted drug delivery, regenerative medicine, and diagnostics. The development of nanocarriers, such as liposomes, polymeric nanoparticles, dendrimers, and metallic nanoparticles, has enabled precise drug delivery with enhanced bioavailability and reduced systemic toxicity. Concurrently, nanostructured scaffolds have advanced regenerative medicine by supporting stem cell differentiation, modulating cellular microenvironments, and enhancing tissue repair. These nanoscale innovations have also led to highly sensitive biosensors and imaging agents, significantly improving early disease detection and biomarker monitoring. Despite these advancements, challenges persist, including nanoparticle-induced cytotoxicity, immunogenicity, scalability issues, and regulatory hurdles requiring extensive evaluation of long-term biocompatibility and pharmacokinetics. Addressing these limitations, recent breakthroughs in AI-assisted nanotechnology and CRISPR-Cas9-mediated gene editing are driving next-generation precision medicine, integrating nanoscale therapeutics with computational approaches to enhance efficacy. Future directions focus on nanorobotics, bioengineered nanovaccines, and theranostic platforms capable of simultaneous diagnosis and treatment, paving the way for real-time, patient-specific interventions. The successful translation of nanomedicine into clinical practice will require interdisciplinary collaboration across nanoscience, bioengineering, and translational medicine to refine nanoparticle functionalization, optimize safety profiles, and ensure equitable access to nanotherapeutics. This review provides a comprehensive overview of these advancements, challenges, and emerging opportunities in nanobiotechnology-driven precision medicine.
Collapse
Affiliation(s)
- Sayantani Chattopadhyay
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603203, Tamil Nadu, India
| | - Arunava Goswami
- Biological Sciences Division, Indian Statistical Institute, 203 B. T. Road, Kolkata, 700108, West Bengal, India.
| | - Moumita Sil
- Biological Sciences Division, Indian Statistical Institute, 203 B. T. Road, Kolkata, 700108, West Bengal, India.
| |
Collapse
|
22
|
Kubbara EA, Bolad A, Malibary H. Advances in Liposomal Interleukin and Liposomal Interleukin Gene Therapy for Cancer: A Comprehensive Review of Preclinical Studies. Pharmaceutics 2025; 17:383. [PMID: 40143046 PMCID: PMC11945541 DOI: 10.3390/pharmaceutics17030383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Preclinical studies on liposomal interleukin (IL) therapy demonstrate considerable promise in cancer treatment. This review explores the achievements, challenges, and future potential of liposomal IL encapsulation, focusing on preclinical studies. METHODS A structured search was conducted using the PubMed and Web of Science databases with the following search terms and Boolean operators: ("liposomal interleukin" OR "liposome-encapsulated interleukin") AND ("gene therapy" OR "gene delivery") AND ("cancer" OR "tumor" OR "oncology") AND ("pre-clinical studies" OR "animal models" OR "in vitro studies". RESULTS Liposomal IL-2 formulations are notable for enhancing delivery and retention at tumor sites. Recombinant human interleukin (rhIL-2) adsorbed onto small liposomes (35-50 nm) substantially reduces metastases in murine models. Hepatic metastasis models demonstrate superior efficacy of liposomal IL-2 over free IL-2 by enhancing immune responses, particularly in the liver. Localized delivery strategies, including nebulized liposomal IL-2 in canine pulmonary metastases and intrathoracic administration in murine sarcoma models, reduce systemic toxicity while promoting immune activation and tumor regression. Liposomal IL gene therapy, delivering cytokine genes directly to tumor sites, represents a notable advancement. Combining IL-2 gene therapy with other cytokines, including IL-6 or double-stranded RNA adjuvants, synergistically enhances macrophage and T-cell activation. Liposomal IL-4, IL-6, and IL-21 therapies show potential across various tumor types. Pairing liposomal IL-2 with chemotherapy or immune agents improves remission and survival. Innovative strategies, including PEGylation and ligand-targeted systems, optimize delivery, release, and therapeutic outcomes. CONCLUSIONS Utilizing immune-stimulatory ILs through advanced liposomal delivery and gene therapy establishes a strong foundation for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Eman A. Kubbara
- Clinical Biochemistry Department, Faculty of Medicine, Rabigh Branch, King Abdulaziz University, Rabigh 21911, Saudi Arabia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Ahmed Bolad
- Department of Microbiology and Unit of Immunology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Husam Malibary
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| |
Collapse
|
23
|
Guo S, Chen M, Yang Y, Zhang Y, Zhuang Y, Dong Y, Tulupov A, Wang X, Cheng J, Bao J, Fan D. Highly efficient tumor oxygen supplementation MnO 2 nano-MOF encapsulated Sorafenib for multiple synergistic CDT/PDT/RT. Int J Pharm 2025; 672:125328. [PMID: 39956406 DOI: 10.1016/j.ijpharm.2025.125328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Tumor growth often creates hypoxic conditions within the tumor microenvironment, which can limit the effectiveness of therapies. To address this issue, a novel "all-in-one" nanoplatform called PCN-224(Hf)@Sorafenib@(PSM) has been developed. This nanoplatform utilizes PCN-224(Hf)-modified MnO2 and combines various therapeutic modalities-chemotherapy, chemodynamic therapy (CDT), photodynamic therapy (PDT), and radiotherapy (RT)-to enhance treatment efficacy. In the PSM nanoplatform, MnO2 decomposes H2O2 to produce oxygen (O2) and reacts with glutathione (GSH) to form Mn2+. This process catalyzes a Fenton-like reaction that generates hydroxyl radicals (·OH), facilitating CDT. When exposed to 635 nm light irradiation, the porphyrin ligand in PCN-224(Hf) produces singlet oxygen (1O2), while the Hf6 clusters contribute to the PDT effects. Furthermore, the nanoplatform enhances radiotherapy by harnessing high-energy radiation. Studies have demonstrated that PSM effectively kills solid tumors even in hypoxic conditions and significantly inhibits tumor growth. This innovative nanoplatform showcases high efficacy in multimodal synergistic tumor treatment, successfully integrating multiple therapeutic approaches to overcome the challenges posed by hypoxia.
Collapse
Affiliation(s)
- Shuangshuang Guo
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Miaomiao Chen
- School of Pharmacy, Xinyang Agriculture and Forestry University, China
| | - Yuhao Yang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yuanyuan Zhang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yuchuan Zhuang
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, United States
| | - Yanbo Dong
- Faculty of Teacher Education, Pingdingshan University, Pingdingshan, Henan 467000, China; Institute of Psychology, The Herzen State Pedagogical University of Russia, Saint Petersburg, Russia
| | - Andrey Tulupov
- Laboratory of MRT Technologies, The Institute International Tomography Center of the Russian Academy of Sciences, Institutskaya Str. 3A, 630090 Novosibirsk, Russia
| | - Xiao Wang
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Jingliang Cheng
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Jianfeng Bao
- Functional Magnetic Resonance and Molecular Imaging Key Laboratory of Henan Province, Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China.
| | - Dandan Fan
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
24
|
Zhang K, Wang T, Huang X, Wu P, Shen L, Yang Y, Wan W, Sun S, Zhang Z. Ultrasound-mediated nanomaterials for the treatment of inflammatory diseases. ULTRASONICS SONOCHEMISTRY 2025; 114:107270. [PMID: 39961217 PMCID: PMC11875835 DOI: 10.1016/j.ultsonch.2025.107270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Sterile and infection-associated inflammatory diseases are becoming increasingly prevalent worldwide. Conventional drug therapies often entail significant drawbacks, such as the risk of drug overdose, the development of drug resistance in pathogens, and systemic adverse reactions, all of which can undermine the effectiveness of treatments for these conditions. Nanomaterials (NMs) have emerged as a promising tool in the treatment of inflammatory diseases due to their precise targeting capabilities, tunable characteristics, and responsiveness to external stimuli. Ultrasound (US), a non-invasive and effective treatment method, has been explored in combination with NMs to achieve enhanced therapeutic outcomes. This review provides a comprehensive overview of the recent advances in the use of US-mediated NMs for treating inflammatory diseases. A comprehensive introduction to the application and classification of US was first presented, emphasizing the advantages of US-mediated NMs and the mechanisms through which US and NMs interact to enhance anti-inflammatory therapy. Subsequently, specific applications of US-mediated NMs in sterile and infection-associated inflammation were summarized. Finally, the challenges and prospects of US-mediated NMs in clinical translation were discussed, along with an outline of future research directions. This review aims to provide insights to guide the development and improvement of US-mediated NMs for more effective therapeutic interventions in inflammatory diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, PR China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, PR China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, PR China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China.
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, PR China; Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
25
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2025; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
26
|
Wang L, Gu M, Zhang X, Kong T, Liao J, Zhang D, Li J. Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413367. [PMID: 39854126 PMCID: PMC11905078 DOI: 10.1002/advs.202413367] [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: 10/21/2024] [Revised: 12/14/2024] [Indexed: 01/26/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
Collapse
Affiliation(s)
- Liyin Wang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Min Gu
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Xiaoli Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | | | - Jun Liao
- Institute of Systems BiomedicineBeijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Dan Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Jingwu Li
- The First Hospital of China Medical UniversityLiaoning110001China
| |
Collapse
|
27
|
Kim K. Hybrid Systems of Gels and Nanoparticles for Cancer Therapy: Advances in Multifunctional Therapeutic Platforms. Gels 2025; 11:170. [PMID: 40136875 PMCID: PMC11941994 DOI: 10.3390/gels11030170] [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: 02/06/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Cancer is a global health concern. Various therapeutic approaches, including chemotherapy, photodynamic therapy, and immunotherapy, have been developed for cancer treatment. Silica nanoparticles, quantum dots, and metal-organic framework (MOF)-based nanomedicines have gained interest in cancer therapy because of their selective accumulation in tumors via the enhanced permeability and retention (EPR) effect. However, bare nanoparticles face challenges including poor biocompatibility, low stability, limited drug-loading capacity, and rapid clearance by the reticuloendothelial system (RES). Gels with unique three-dimensional network structures formed through various interactions such as covalent and hydrogen bonds are emerging as promising materials for addressing these challenges. Gel hybridization enhances biocompatibility, facilitates controlled drug release, and confers cancer-targeting abilities to nanoparticles. This review discusses gel-nanoparticle hybrid systems for cancer treatment developed in the past five years and analyzes the roles of gels in these systems.
Collapse
Affiliation(s)
- Kibeom Kim
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
28
|
Parra-Nieto J, Hidalgo L, Márquez-Cantudo M, García-Castro J, Megias D, Ramirez M, Baeza A. Liposomal-Based Nanoarchitectonics as Bispecific T Cell Engagers in Neuroblastoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11937-11945. [PMID: 39957209 DOI: 10.1021/acsami.5c00633] [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: 02/18/2025]
Abstract
Neuroblastoma (NB) is an aggressive pediatric solid tumor that lacks efficient treatment. In the past few years, the use of engineered lymphocytes endowed with chimeric antigen receptors (CAR T), which improve their natural search and destroy skills against tumoral cells, has provided a highly valuable strategy to eradicate tumors in a specific and safe manner. Unfortunately, despite the excellent results achieved by these cell-based therapies in liquid tumors, their efficacy in the treatment of solid malignancies is usually modest due to the existence of several biological barriers which compromise their efficacy. Herein, a strategy to guide CAR T toward NB cells based on the use of nanometric bispecific T engagers (NBTEs) is presented. These novel bispecific nanoplatforms are based on liposomes and protocells doubly functionalized with synthetic targeting moieties (para-aminobenzylguanidine and fluorescein) able to selectively bind to membrane cell receptors of NB and anti-FITC CAR T, respectively. The binding process of NBTEs to NB cells was monitored by confocal fluorescence microscopy showing the excellent capacity of these nanodevices to place fluorescence labels on the surface of the malignant cells. Then, NB cells previously incubated in the presence of NBTEs were rapidly detected and destroyed by anti-FITC CAR T, which confirmed the excellent capacity of these nanoplatforms to improve the natural capacity of CAR T to eradicate malignant cells. Finally, the high versatility of the NBTE design and its easy-to-tune nature would allow their rapid application to different types of solid tumors.
Collapse
Affiliation(s)
- Jorge Parra-Nieto
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Laura Hidalgo
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Marta Márquez-Cantudo
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Javier García-Castro
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III, Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Diego Megias
- Advanced Optical Microscopy Unit, Instituto de salud Carlos III (ISCIII), Crta, Majadahonda-Pozuelo 2, 28220 Madrid, Spain
| | - Manuel Ramirez
- Servicio de Hematología y Oncología Pediátrica, Hospital Infantil Universitario Niño Jesús, Av. de Menéndez Pelayo, 65, Retiro, 28009 Madrid, Spain
| | - Alejandro Baeza
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
29
|
Liu Y, Song J, Guo Y, Li S, Yuan M, Tang J, Wang Y, Li M, Guo Y, Guo L. Synergistic therapy with celastrol-curcumin multifunctional nanomedicine: Anti-hepatocellular carcinoma and reduced hepatotoxicity. Int J Pharm 2025; 671:125289. [PMID: 39880142 DOI: 10.1016/j.ijpharm.2025.125289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Hepatocellular carcinoma is one of the leading causes of cancer deaths globally and a key hindrance to extending life expectancy. Celastrol (CEL) demonstrates excellent antitumor activity, but faces challenges like low solubility and a narrow therapeutic window, limiting its clinical application. To address these limitations, drug combinations and nano-delivery systems have emerged as effective solutions. Curcumin (CUR), known for its antitumor and hepatoprotective effects, also exhibits good biocompatibility and the ability to mitigate drug-induced liver injury. Considering the complementary properties of CEL and CUR, including CEL's potent antitumor activity and CUR's hepatoprotective effects, we developed a novel self-assembling nanodrug delivery system (CCPN) for the co-loading of both compounds. CCPN nanoparticles were constructed through non-covalent interactions, including hydrogen bonding, π-π stacking, and electrostatic forces, which confer good stability and significantly enhance the solubility and bioavailability of CEL and CUR. Extensive in vitro and in vivo experiments demonstrated that CCPN effectively reduced CEL-induced hepatotoxicity in zebrafish and mouse models, exhibiting good biosafety. Additionally, CUR's fluorescence provides a unique advantage for real-time monitoring of drug distribution and release, facilitating the tracking of therapeutic progress. Furthermore, CCPN nanoparticles enhanced delivery efficiency in HepG2 cells, exhibiting superior anti-liver tumor outcomes, which are associated with the promotion of apoptosis in tumor cells. This study presents CCPN as a promising therapeutic strategy for hepatocellular carcinoma, integrating reduced hepatotoxicity, self-monitoring capabilities, and superior therapeutic efficacy.
Collapse
Affiliation(s)
- Yushi Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China; Department of Pharmacy, Chengdu Seventh People's Hospital, Chengdu 610213 China
| | - Jiawen Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yurou Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Meifeng Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| |
Collapse
|
30
|
Yu S, Rejinold NS, Choi G, Choy JH. Revolutionizing healthcare: inorganic medicinal nanoarchitectonics for advanced theranostics. NANOSCALE HORIZONS 2025; 10:460-483. [PMID: 39648727 DOI: 10.1039/d4nh00497c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Over the last two decades, advancements in nanomaterials and nanoscience have paved the path for the emergence of nano-medical convergence science, significantly impacting healthcare. In our review, we highlight how these advancements are applied in various biomedical technologies such as drug delivery systems, bio-imaging for diagnostic and therapeutic purposes. Recently, novel inorganic nanohybrid drugs have been developed, combining multifunctional inorganic nanomaterials with therapeutic agents (known as inorganic medicinal nanoarchitectonics). These innovative drugs are actively utilized in cutting-edge medical treatments, including targeted anti-cancer therapy, photo and radiation therapy, and immunotherapy. This review provides a detailed overview of the current development status of inorganic medicinal nanoarchitectonics and explores potential future directions in their advancements.
Collapse
Affiliation(s)
- Seungjin Yu
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - N Sanoj Rejinold
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
| | - Goeun Choi
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Ho Choy
- Intelligent Nanohybrid Materials Laboratory (INML), Department of Chemistry, College of Science and Technology, Dankook University, Cheonan 31116, Republic of Korea.
- Division of Natural Sciences, The National Academy of Sciences, Seoul 06579, Republic of Korea
- Tokyo Tech Tokyo Tech World Research Hub Initiative (WRHI), Institute of Innovative Research, Institute of Science Tokyo, Yokohama 226853, Japan
| |
Collapse
|
31
|
Chen Q, Yu T, Gong J, Shan H. Advanced Nanomedicine Delivery Systems for Cardiovascular Diseases: Viral and Non-Viral Strategies in Targeted Therapy. Molecules 2025; 30:962. [PMID: 40005272 PMCID: PMC11858567 DOI: 10.3390/molecules30040962] [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: 01/09/2025] [Revised: 02/08/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular diseases (CVDs) represent a leading global health crisis, significantly impairing patients' quality of life and posing substantial risks to their survival. Conventional therapies for CVDs often grapple with challenges such as inadequate targeting precision, suboptimal therapeutic efficacy, and potential adverse side effects. To address these shortcomings, researchers are intensively developing advanced drug delivery systems characterized by high specificity and selectivity, excellent biodegradability, superior biocompatibility, and minimal toxicity. These innovative systems enable the precise delivery of pharmaceuticals with high drug-loading capacities, minimal leakage, and expansive specific surface areas, thereby enhancing therapeutic outcomes. In this review, we summarize and classify various drug delivery materials targeting CVDs and application values. We also evaluate the feasibility and efficacy of viral and non-viral vectors for the treatment of CVDs, the existing limitations and application prospects are also discussed. We hope that this review will provide new perspectives for the future development of drug delivery systems for the treatment of CVDs, ultimately contributing to improved patient care and outcomes.
Collapse
Affiliation(s)
| | - Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (Q.C.); (J.G.)
| | | | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (Q.C.); (J.G.)
| |
Collapse
|
32
|
Roszkowski S, Durczyńska Z, Szablewska S. Targeted nanodelivery systems for personalized cancer therapy. Rep Pract Oncol Radiother 2025; 29:776-788. [PMID: 40104662 PMCID: PMC11912883 DOI: 10.5603/rpor.103524] [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: 11/27/2023] [Accepted: 11/12/2024] [Indexed: 03/20/2025] Open
Abstract
Conventional cancer therapies such as chemotherapy face challenges such as poor tumor targeting, systemic toxicity, and drug resistance. Nanotechnology offers solutions through advanced drug delivery systems that preferentially accumulate in tumors while avoiding healthy tissues. Recent innovations have enabled the optimization of engineered nanocarriers for extended circulation and tumor localization via both passive and active targeting mechanisms. Passive accumulation exploits the leaky vasculature of tumors, whereas active strategies use ligands to selectively bind cancer cell receptors. Multifunctional nanoparticles also allow the combination of imaging, multiple therapeutic modalities and on-demand drug release within a single platform. Overall, precisely tailored nanotherapeutics that leverage unique pathophysiological traits of malignancies provide opportunities to overcome the limitations of traditional treatment regimens. This emerging field promises more effective and personalized nanomedicine approaches to detect and treat cancer. The key aspects highlighted in this review include the biological barriers associated with nanoparticles, rational design principles to optimize nanocarrier pharmacokinetics and tumor uptake, passive and active targeting strategies, multifunctionality, and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Zofia Durczyńska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Sylwia Szablewska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| |
Collapse
|
33
|
Yu Q, Zhou J, Tao Q, Liu Y, Zhou H, Kang B, Xu JJ. Ultrasound-Activated Copper Matrix Nanosonosensitizer for Cuproptosis-Based Synergy Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1503-1510. [PMID: 39883479 DOI: 10.1021/acsabm.4c01710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Cuproptosis exhibits enormous application prospects in treatment. However, cuproptosis-based therapy is impeded by the limited intracellular copper ions, the nonspecific delivery, uncontrollable release, and chelation of endogenous overproduced glutathione (GSH). In this work, an ultrasound-triggered nanosonosensitizer (p-TiO2-Cu(I)) was constructed for Cu(I) delivery, on-demand release, GSH consumption, and deeper tissue response. When the nanomedicine was internalized into the tumor cells, ultrasound (US) induced the nanosonosensitizer to produce reactive oxygen species (ROS) to achieve sonodynamic therapy (SDT). GSH, acting as a hole trapping agent, improved the efficiency of SDT. Meanwhile, the downgrade of GSH was beneficial to cuproptosis and oxidative damage-based SDT in return. What is more, the US could regulate the release behavior of Cu(I). Cu(I) bonded to mitochondrial proteins and then aggregated the lipoylated protein, bringing about the turbulence of the tricarboxylic acid cycle. The combination of SDT and cuproptosis showed high matching to induce efficient cuproptosis and may inspire other cuproptosis-based nanosonosensitizer designs.
Collapse
Affiliation(s)
- Qiao Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jie Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Qianqian Tao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yong Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Hong Zhou
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Bin Kang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
34
|
Hu P, Zheng J, Wang H, Li Y, Ye T, Li Q, Lan X, Liu C, Liu C. Supramolecular Nanozymes Based on Self-Assembly of Biomolecule for Cancer Therapy. Int J Nanomedicine 2025; 20:2043-2057. [PMID: 39990286 PMCID: PMC11842878 DOI: 10.2147/ijn.s496831] [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: 09/27/2024] [Accepted: 01/10/2025] [Indexed: 02/25/2025] Open
Abstract
Natural enzyme systems possess extraordinary functions and characteristics, making them highly appealing for use in eco-friendly technologies and innovative cancer treatments. However, their inherent instability and structural complexity often limit their practical applications, leading to the exploration of biomolecular nanozyme alternatives. Supramolecular nanozymes, constructed using self-assembly techniques and various non-covalent interactions, have emerged as a promising solution. Amino acids, peptides, and protein motifs offer flexible building blocks for constructing these nanozymes. Importantly, the well-defined structural regulation mechanisms of biomolecular nanozymes, along with their unique properties as fundamental biological modules in living systems-such as selectivity, permeability, retention, and biocompatibility-present new opportunities for cancer therapy. This review highlights recent advances in supramolecular self-assembled nanozymes, including peroxidases, oxidases, catalases, superoxide dismutases, and other nanozyme systems, as building blocks for tumor therapy. Additionally, it discusses precise functional modulation through supramolecular non-covalent interactions and their therapeutic applications in targeting the tumor microenvironment. These studies provide valuable insights that may inspire the design of novel supramolecular nanozymes with enhanced catalytic selectivity, biocompatibility, and tumor-killing efficacy.
Collapse
Affiliation(s)
- Pengcheng Hu
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Jilu Zheng
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Hongjuan Wang
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Yongxin Li
- College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Tao Ye
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
- School of Clinical Medicine, Shandong second Medical University, Weifang, Shandong, 261053, People’s Republic of China
| | - Quanjun Li
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Xiaopeng Lan
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| | - Chunzhao Liu
- College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, People’s Republic of China
| | - Chunlei Liu
- Department of Urology, Department of Primary Healthcare, Department of Cardiology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266071, People’s Republic of China
| |
Collapse
|
35
|
Zhang Y, Lian Y, Zhou C, Cheng J, Zhao S, Liu H, Wang J, Lu X, Shi J, Du G. Self-assembled natural triterpenoids for the delivery of cyclin-dependent kinase 4/6 inhibitors to enhance cancer chemoimmunotherapy. J Control Release 2025; 378:791-802. [PMID: 39732370 DOI: 10.1016/j.jconrel.2024.12.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/22/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Immunogenic cell death (ICD) has recently emerged as a promising strategy in reinforcing anti-PD-L1 blockade immunotherapy of triple-negative breast cancer (TNBC). The CDK4/6 inhibitor palbociclib (PAL), as a clinical star medicine targeting the cell cycle machinery, is an ideal candidate for fabricating a highly efficient ICD inducer for TNBC chemoimmunotherapy. However, the frequently observed chemoresistance and clinical adverse effects, as well as significant antagonistic effects when co-administered with certain chemotherapeutics, have seriously restricted the efficiency of PAL and the feasibility of combination strategies. Herein, we screened and identified six self-assembled natural pentacyclic triterpenoid (PT) molecules that can serve as competent co-administration nanoplatforms for the synergistic or combined delivery of PAL. Analysis of two representative PT-PAL nano-assemblies validated that PT-mediated co-assembly enhances the cytotoxicity and synergy of PAL by inhibiting the PI3K/AKT/mTOR signaling pathway, rather than directly targeting CDK4/6 proteins. Importantly, the PAL nanoassemblies exhibited multiple favorable therapeutic features and stronger accumulative ICD induction, ensuring highly efficient synergistic anti-PD-L1 chemoimmunotherapy by simultaneously facilitating T-cell immune response and reversing the immunosuppressive tumor microenvironment. This study offers possibilities for improving the anticancer efficacy of CDK4/6 inhibitors and potential avenues for clinical applications of chemoimmunotherapy in treating TNBC.
Collapse
Affiliation(s)
- Yongbo Zhang
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China
| | - Yajie Lian
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China
| | - Conglei Zhou
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China
| | - Jianjun Cheng
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China.
| | - Shuang Zhao
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China
| | - Hongjun Liu
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China
| | - Jiacheng Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China
| | - Xin Lu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China
| | - Jiahua Shi
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China.
| | - Guanhua Du
- Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475000, PR China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475000, PR China; Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
36
|
Jia J, Lin R, Liu M, Hou M, Yu H, Lu Q, Ma Y, Zhao T, Zhang F, Mady MF, Elzatahry AA, Wang J, Ji Y, Zhao D, Li X. Dual-Ligand Assisted Anisotropic Assembly for the Construction of NIR-II Light-Propelled Mesoporous Nanomotors. J Am Chem Soc 2025; 147:4198-4209. [PMID: 39871601 DOI: 10.1021/jacs.4c14011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The advent of autonomous nanomotors presents exciting opportunities for nanodrug delivery. However, significant potential remains for enhancing the asymmetry of nanomotors and advancing the development of second near-infrared (NIR-II) light-propelled nanomotors capable of operating within deep tissues. Herein, we developed a dual-ligand assisted anisotropic assembly strategy that enables precise regulation of the interfacial energy between selenium (Se) nanoparticle and periodic mesoporous organosilica (PMO). This strategy facilitates the controllable anisotropic growth of PMO on the Se nanoparticle, leading to the formation of Se&PMO Janus nanohybrids. The exposure ratio of the Se subunit within the Janus nanohybrids can be finely tuned from 0% to 75%. Leveraging the transformability of the Se subunit, a variety of functional MxSe&PMO Janus nanocomposites (MxSe denotes metal selenide) were further derived. As a proof of concept, CuSe&PMO Janus nanohybrids, with NIR-II photothermal properties, were employed as NIR-II light-driven nanomotors. By precisely controlling the exposure ratio of the CuSe subunit within the Janus nanostructure, these CuSe&PMO nanomotors achieved optimal self-propulsion, thus enhancing cellular uptake and promoting deep tumor penetration. Furthermore, the high loading capacity and hydrophobic framework of the PMO subunit enabled the incorporation of hydrophobic disulfiram, thereby significantly boosting the efficacy of synergistic active-motion photothermal therapy.
Collapse
Affiliation(s)
- Jia Jia
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Runfeng Lin
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Minchao Liu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Mengmeng Hou
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Hongyue Yu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Qianqian Lu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Yuzhu Ma
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Tiancong Zhao
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Fan Zhang
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Mohamed F Mady
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha 2713, Qatar
| | - Ahmed A Elzatahry
- William A. Brookshire Department of Chemical and Biomolecular Engineering, Cullen College of Engineering, University of Houston, Houston, Texas 77204, United States
| | - Jiawen Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Yujin Ji
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Dongyuan Zhao
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Xiaomin Li
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
37
|
Nie H, Huang R, Jiang G, Li W, Yang L, Zhang M, Qian M, Guo W, Ye T, Huang R. Modulating active targeting nanoparticle design according to tumor progressions. Acta Pharm Sin B 2025; 15:1143-1158. [PMID: 40177554 PMCID: PMC11959910 DOI: 10.1016/j.apsb.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/25/2024] [Accepted: 11/07/2024] [Indexed: 04/05/2025] Open
Abstract
Targeting drug delivery systems mediated by nanoparticles has shown great potential in the diagnosis and treatment of cancer. However, influences of different tumor progressions on the accumulation of nanoparticles, especially the ligand-modified active targeting nanoparticles are seldom exploited. In this work, the accumulation and penetration of RGD-modified gold nanoparticles (active AuNPs) with different sizes were investigated in orthotopic breast cancer with different tumor progressions. The results showed that the smallest active AuNPs had better accumulation and permeation effects in early tumor tissues with the relatively looser extracellular matrix, larger gaps, lower interstitial fluid pressure, and less receptor expression, which was due to size effects. However, the larger active AuNPs had better accumulation and penetration effects in late tumor tissues with highly expressed target receptors integrin α v β 3 because of the multivalent interactions between larger active nanoparticles and integrin α v β 3. In the midterm, tumor accumulation of active AuNPs was equally influenced by size effects and multivalent interactions. Therefore, RGD-modified nanoparticles with sizes of 7 and 90 nm accumulated more in tumors. This study will guide a rational design of active targeting nanoparticles for enhancing the diagnosis and treatment of tumors based on their progressions.
Collapse
Affiliation(s)
- Huifang Nie
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Rong Huang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Guangwei Jiang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Wenshuai Li
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Lan Yang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Meng Zhang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Min Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Wei Guo
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Tao Ye
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| | - Rongqin Huang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, Fudan University, Shanghai 201203, China
| |
Collapse
|
38
|
Chin B, Meng Lim W, Almurisi SH, Madheswaran T. A quality-by-design approach to develop abemaciclib solid lipid nanoparticles for targeting breast cancer cell lines. Ther Deliv 2025; 16:123-137. [PMID: 39878544 PMCID: PMC11849948 DOI: 10.1080/20415990.2025.2457314] [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: 04/29/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
AIM Abemaciclib (ABE) is an anticancer drug that suffers from low bioavailability and multidrug resistance. This study aims to develop ABE-loaded solid lipid nanoparticles (ABE-SLNs), which will enhance drug solubility and lead to increased cellular uptake and enhanced cytotoxicity when delivering tumor cells. METHODS Melt emulsification followed by ultrasonication was used as a method of preparation and Quality-by-Design (QbD) was utilized to optimize ABE-SLNs. RESULTS The optimized ABE-SLNs consist of Precirol-ATO5 as a lipid and Brij-58 as a surfactant. The particle size, PDI value, and zeta potential of the optimized formulation were 170.4 ± 0.49 nm, 0.25 ± 0.014, and -26.4 ± 0.1 mV, respectively. It also showed sustained release behavior and a high entrapment efficiency of 79.96%. ABE-SLNs exhibited enhanced anticancer activity in the MDA-MB-231 and T47D breast cancer cell lines compared to pure ABE. In Caco-2 human colonic cell lines, ABE-SLNs also showed increased cellular uptake. CONCLUSION The use of QbD to achieve high entrapment efficiency and sustained release in ABE-SLNs, coupled with enhanced cellular uptake and cytotoxicity, represents a novel approach that could set a new standard for nanoparticle-based drug delivery systems.
Collapse
Affiliation(s)
- Bonnie Chin
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Wei Meng Lim
- School of Pharmacy, Monash University, Selangor, Malaysia
| | - Samah Hamed Almurisi
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
- Centre for Bioactive Molecules & Drug Delivery, Institute for Research, Development & Innovation (IRDI), IMU University, Kuala Lumpur, Malaysia
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
- Centre for Bioactive Molecules & Drug Delivery, Institute for Research, Development & Innovation (IRDI), IMU University, Kuala Lumpur, Malaysia
| |
Collapse
|
39
|
Singh P, Pandit S, Balusamy SR, Madhusudanan M, Singh H, Amsath Haseef HM, Mijakovic I. Advanced Nanomaterials for Cancer Therapy: Gold, Silver, and Iron Oxide Nanoparticles in Oncological Applications. Adv Healthc Mater 2025; 14:e2403059. [PMID: 39501968 PMCID: PMC11804848 DOI: 10.1002/adhm.202403059] [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: 08/26/2024] [Revised: 10/07/2024] [Indexed: 01/05/2025]
Abstract
Cancer remains one of the most challenging health issues globally, demanding innovative therapeutic approaches for effective treatment. Nanoparticles, particularly those composed of gold, silver, and iron oxide, have emerged as promising candidates for changing cancer therapy. This comprehensive review demonstrates the landscape of nanoparticle-based oncological interventions, focusing on the remarkable advancements and therapeutic potentials of gold, silver, and iron oxide nanoparticles. Gold nanoparticles have garnered significant attention for their exceptional biocompatibility, tunable surface chemistry, and distinctive optical properties, rendering them ideal candidates for various cancer diagnostic and therapeutic strategies. Silver nanoparticles, renowned for their antimicrobial properties, exhibit remarkable potential in cancer therapy through multiple mechanisms, including apoptosis induction, angiogenesis inhibition, and drug delivery enhancement. With their magnetic properties and biocompatibility, iron oxide nanoparticles offer unique cancer diagnosis and targeted therapy opportunities. This review critically examines the recent advancements in the synthesis, functionalization, and biomedical applications of these nanoparticles in cancer therapy. Moreover, the challenges are discussed, including toxicity concerns, immunogenicity, and translational barriers, and ongoing efforts to overcome these hurdles are highlighted. Finally, insights into the future directions of nanoparticle-based cancer therapy and regulatory considerations, are provided aiming to accelerate the translation of these promising technologies from bench to bedside.
Collapse
Affiliation(s)
- Priyanka Singh
- The Novo Nordisk FoundationCenter for BiosustainabilityTechnical University of DenmarkKogens LyngbyDK‐2800Denmark
| | - Santosh Pandit
- Systems and Synthetic Biology DivisionDepartment of Life SciencesChalmers University of TechnologyGothenburgSE‐412 96Sweden
| | - Sri Renukadevi Balusamy
- Department of Food Science and BiotechnologySejong UniversityGwangjin‐GuSeoul05006Republic of Korea
| | - Mukil Madhusudanan
- The Novo Nordisk FoundationCenter for BiosustainabilityTechnical University of DenmarkKogens LyngbyDK‐2800Denmark
| | - Hina Singh
- Division of Biomedical SciencesSchool of MedicineUniversity of CaliforniaRiversideCA92521USA
| | | | - Ivan Mijakovic
- The Novo Nordisk FoundationCenter for BiosustainabilityTechnical University of DenmarkKogens LyngbyDK‐2800Denmark
- Systems and Synthetic Biology DivisionDepartment of Life SciencesChalmers University of TechnologyGothenburgSE‐412 96Sweden
| |
Collapse
|
40
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [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: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
41
|
Coelho D, Estêvão D, Oliveira MJ, Sarmento B. Radioresistance in rectal cancer: can nanoparticles turn the tide? Mol Cancer 2025; 24:35. [PMID: 39885557 PMCID: PMC11784129 DOI: 10.1186/s12943-025-02232-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
Rectal cancer accounts for over 35% of the worldwide colorectal cancer burden representing a distinctive subset of cancers from those arising in the colon. Colorectal cancers exhibit a continuum of traits that differ with their location in the large intestine. Due to anatomical and molecular differences, rectal cancer is treated differently from colon cancer, with neoadjuvant chemoradiotherapy playing a pivotal role in the control of the locally advanced disease. However, radioresistance remains a major obstacle often correlated with poor prognosis. Multifunctional nanomedicines offer a promising approach to improve radiotherapy response rates, as well as to increase the intratumoral concentration of chemotherapeutic agents, such as 5-Fluorouracil. Here, we revise the main molecular differences between rectal and colon tumors, exploring the complex orchestration beyond rectal cancer radioresistance and the most promising nanomedicines reported in the literature to improve neoadjuvant therapy response rates.
Collapse
Affiliation(s)
- Diogo Coelho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| | - Diogo Estêvão
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Cancer Research Institute, Ghent University, Ghent, Belgium
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, Porto, 4200-319, Portugal
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, Porto, 4200-319, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen 208, Porto, 4200‑135, Portugal.
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal.
| |
Collapse
|
42
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
43
|
Shi Z, Artemenko M, Yu W, Zhang M, Yi C, Chen P, Lin S, Bian Z, Lian B, Meng F, Chen J, Roussel T, Li Y, Chan KKL, Ip PPC, Lai HC, To SKY, Liu X, Peng L, Wong AST. Bola-Amphiphilic Dendrimer Enhances Imatinib to Target Metastatic Ovarian Cancer via β-Catenin-HRP2 Signaling Axis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2884-2898. [PMID: 39752231 PMCID: PMC11744500 DOI: 10.1021/acsami.4c12857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Ovarian cancer is the leading cause of death among all gynecological malignancies, and drug resistance renders the current chemotherapy agents ineffective for patients with advanced metastatic tumors. We report an effective treatment strategy for targeting metastatic ovarian cancer involving a nanoformulation (Bola/IM)─bola-amphiphilic dendrimer (Bola)-encapsulated imatinib (IM)─to target the critical mediator of ovarian cancer stem cells (CSCs) CD117 (c-Kit). Bola/IM offered significantly more effective targeting of CSCs compared to IM alone, through a novel and tumor-specific β-catenin/HRP2 axis, allowing potent inhibition of cancer cell survival, stemness, and metastasis in metastatic and drug-resistant ovarian cancer cells. Promising results were also obtained in clinically relevant patient-derived ascites and organoids alongside high tumor-oriented accumulation and favorable pharmacokinetic properties in mouse models. Furthermore, Bola/IM displayed synergistic anticancer activity when combined with the first-line chemotherapeutic drug cisplatin in patient-derived xenograft mouse models without any adverse effects. Our findings support the use of Bola/IM as a nanoformulation to empower IM, providing targeted and potent treatment of metastatic ovarian cancer. Our study thus represents a significant advancement toward addressing the unmet medical need for improved therapies targeting this challenging disease.
Collapse
Affiliation(s)
- Zeyu Shi
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
- Laboratory
for Synthetic Chemistry and Chemical Biology Limited, Pokfulam, Hong Kong 999077, China
| | - Margarita Artemenko
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
| | - Weiyu Yu
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
| | - Ming Zhang
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
| | - Canhui Yi
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
| | - Peng Chen
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Shuting Lin
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Zhancun Bian
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Aix-Marseille
Université, CNRS, Centre Interdisciplinaire de Nanoscience
de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Baoping Lian
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Fanzhen Meng
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaxuan Chen
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
- Aix-Marseille
Université, CNRS, Centre Interdisciplinaire de Nanoscience
de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Tom Roussel
- Aix-Marseille
Université, CNRS, Centre Interdisciplinaire de Nanoscience
de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Ying Li
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Karen K. L. Chan
- Department
of Obstetrics and Gynecology, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Philip P. C. Ip
- Department
of Pathology, Queen Mary Hospital, University
of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Hung-Cheng Lai
- Department
of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department
of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, Taipei 23561, Taiwan
| | - Sally K. Y. To
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
- Laboratory
for Synthetic Chemistry and Chemical Biology Limited, Pokfulam, Hong Kong 999077, China
| | - Xiaoxuan Liu
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug
Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals
and Biomaterials, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Peng
- Aix-Marseille
Université, CNRS, Centre Interdisciplinaire de Nanoscience
de Marseille, Equipe Labellisée Ligue Contre le Cancer, 13288 Marseille, France
| | - Alice S. T. Wong
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 999077, China
| |
Collapse
|
44
|
Mao M, Wu Y, He Q. Breaking Through Physiological Barriers: Nanorobotic Strategies for Active Drug Delivery. Bioconjug Chem 2025; 36:1-14. [PMID: 39729406 DOI: 10.1021/acs.bioconjchem.4c00480] [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: 12/29/2024]
Abstract
Self-propelled micro/nanomotors (MNMs) represent a groundbreaking advancement in precision drug delivery, offering potential solutions to persistent challenges such as systemic toxicity, limited bioavailability, and nonspecific distribution. By transforming various energy sources into mechanical motion, MNMs are able to autonomously navigate through complex physiological environments, facilitating targeted delivery of therapeutic agents to previously inaccessible regions. However, to achieve efficient in vivo drug delivery, biomedical MNMs must demonstrate their ability to overcome crucial physiological barriers encompassing mucosal surfaces, blood flow dynamics, vascular endothelium, and cellular membrane. This review provides a comprehensive overview of the latest strategies developed to address these obstacles while also analyzing the broader challenges and opportunities associated with clinical translation. Our objective is to establish a solid foundation for future research in medical MNMs by focusing on enhancing drug delivery efficiency and advancing precision medicine, ultimately paving the way for practical theragnostic applications and wider clinical adoption.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
45
|
Chen X, Li F, Cui B, Yan Q, Qiu C, Zhu Z, Wen L, Chen W. Liposomes-mediated enhanced antitumor effect of docetaxel with BRD4-PROTAC as synergist for breast cancer chemotherapy/immunotherapy. Int J Pharm 2025; 668:124973. [PMID: 39566696 DOI: 10.1016/j.ijpharm.2024.124973] [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: 07/31/2024] [Revised: 11/07/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
It has been reported that proteolysis-targeting chimeras (PROTACs) can effectively degrade intracellular oncogenic proteins, providing an ideal strategy for cancer treatment. ARV825, a bromodomain-containing protein 4 (BRD4)-PROTAC, has demonstrated the capacity to enhance the antitumor effect of the classic chemotherapeutic agent docetaxel (DTX). However, there are three major challenges to the broader in vivo application of ARV825: poor solubility, poor permeability, and off-target effects. Additionally, the efficient co-delivery of ARV825 and DTX to tumor tissues for a synergistic therapeutic effect remains unresolved. In this study, liposomes were utilized as co-delivery vehicles for ARV825 and DTX to effectively address these issues. The well-established liposomes significantly improved the solubility of both ARV825 and DTX while maintaining a sustained release profile in blood-mimetic conditions. The co-loaded liposomes accumulated in tumor tissues via the enhanced permeability and retention (EPR) effect. After internalization, ARV825 effectively degraded intracellular BRD4 proteins and downregulated the expression of both Bcl-2 and PD-L1 proteins, thereby increasing tumor cell apoptosis and enhancing the tumor immune response. This, in turn, augmented the antitumor effect of DTX in vivo without undesired side effects. In conclusion, BRD4-PROTAC may serve as a promising synergistic agent alongside the conventional chemotherapeutic agent DTX, with liposomes functioning as effective co-delivery vehicles.
Collapse
Affiliation(s)
- Xixi Chen
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Fang Li
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Binghui Cui
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Qingyi Yan
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Caisheng Qiu
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Zengyan Zhu
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Lijuan Wen
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| | - Weiliang Chen
- School of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| |
Collapse
|
46
|
Van Linthout S, Stellos K, Giacca M, Bertero E, Cannata A, Carrier L, Garcia‐Pavia P, Ghigo A, González A, Haugaa KH, Imazio M, Lopes LR, Most P, Pollesello P, Schunkert H, Streckfuss‐Bömeke K, Thum T, Tocchetti CG, Tschöpe C, van der Meer P, van Rooij E, Metra M, Rosano GM, Heymans S. State of the art and perspectives of gene therapy in heart failure. A scientific statement of the Heart Failure Association of the ESC, the ESC Council on Cardiovascular Genomics and the ESC Working Group on Myocardial & Pericardial Diseases. Eur J Heart Fail 2025; 27:5-25. [PMID: 39576264 PMCID: PMC11798634 DOI: 10.1002/ejhf.3516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 02/07/2025] Open
Abstract
Gene therapy has recently become a reality in the treatment of cardiovascular diseases. Strategies to modulate gene expression using antisense oligonucleotides or small interfering RNA are proving to be safe and effective in the clinic. Adeno-associated viral vector-based gene delivery and CRISPR-Cas9-based genome editing have emerged as efficient strategies for gene delivery and repair in humans. Overall, gene therapy holds the promise not only of expanding current treatment options, but also of intervening in previously untackled causal disease mechanisms with little side effects. This scientific statement provides a comprehensive overview of the various modalities of gene therapy used to treat heart failure and some of its risk factors, and their application in the clinical setting. It discusses specifically the possibilities of gene therapy for hereditary heart diseases and (non)-genetic heart failure. Furthermore, it addresses safety and clinical trial design issues and challenges for future regulatory strategies.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Berlin Institute of Health (BIH) at Charité – Universitätmedizin BerlinBIH Center for Regenerative Therapies (BCRT)BerlinGermany
- German Center for Cardiovascular Research (DZHK)partner site BerlinBerlinGermany
| | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive CareUniversity Medical Centre Mannheim, Heidelberg UniversityMannheimGermany
- German Centre for Cardiovascular Research (DZHK)partner site Heidelberg/MannheimMannheimGermany
- Helmholtz Institute for Translational AngioCardioScience (HI‐TAC)MannheimGermany
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical SciencesNewcastle UniversityNewcastleUK
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK; Department of Medical SciencesUniversity of TriesteTriesteItaly
| | - Edoardo Bertero
- Cardiovascular Unit, Department of Internal MedicineUniversity of GenovaGenovaItaly
| | - Antonio Cannata
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research ExcellenceKing's College LondonLondonUK
| | - Lucie Carrier
- Department of Experimental Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Centre for Cardiovascular Research (DZHK)partner site Hamburg/Kiel/LübeckHamburgGermany
| | - Pablo Garcia‐Pavia
- Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, CIBERCVMadridSpain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- Universidad Francisco de Vitoria (UFV)MadridSpain
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health SciencesMolecular Biotechnology Center "Guido Tarone," University of TorinoTorinoItaly
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA and Department of Pathology, Anatomy and PhysiologyUniversidad de NavarraPamplonaSpain
- IdiSNANavarra Institute for Health ResearchPamplonaSpain
- CIBERCV (Network for Biomedical Research in Cardiovascular Disease)Instituto de Salud Carlos IIMadridSpain
| | - Kristina H. Haugaa
- ProCardio Center for Innovation, Department of CardiologyOslo University Hospital, RikshospitaletOsloNorway
- Faculty of Medicine, Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Massimo Imazio
- Department of Medicine (DMED), University of Udine, and Cardiothoracic Department ASUFCUniversity Hospital Santa Maria della MisericordiaUdineItaly
| | - Luis R. Lopes
- Institute of Cardiovascular ScienceUniversity College LondonLondonUK
- Barts Heart Centre, St Bartholomew's HospitalLondonUK
| | - Patrick Most
- Department of Cardiology, Angiology, PulmonologyUniversity Hospital HeidelbergHeidelbergGermany
| | | | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum MünchenTechnische Universität MünchenMunichGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Katrin Streckfuss‐Bömeke
- Clinic for Cardiology and PneumologyUniversity Medical CenterGöttingenGermany
- German Center for Cardiovascular Research (DZHK), Partner site GöttingenGöttingenGermany
- Institute of Pharmacology and ToxicologyUniversity of WürzburgWürzburgGermany
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC)University Clinic WürzburgWürzburgGermany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS)Hannover Medical SchoolHannoverGermany
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences; Center for Basic and Clinical Immunology Research (CISI); Interdepartmental Center for Clinical and Translational Research (CIRCET); Interdepartmental Hypertension Research Center (CIRIAPA)Federico II UniversityNaplesItaly
| | - Carsten Tschöpe
- Berlin Institute of Health (BIH) at Charité – Universitätmedizin BerlinBIH Center for Regenerative Therapies (BCRT)BerlinGermany
- German Center for Cardiovascular Research (DZHK)partner site BerlinBerlinGermany
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiology, Angiology and Intensive MedicineCampus Virchow KlinikumBerlinGermany
| | - Peter van der Meer
- Department of CardiologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Eva van Rooij
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center UtrechtUtrechtThe Netherlands
- Department of CardiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Marco Metra
- Cardiology, ASST Spedali Civili di Brescia, Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Giuseppe M.C. Rosano
- Cardiovascular Clinical Academic Group, St. George's University Hospitals, NHS TrustUniversity of LondonLondonUK
- Cardiology, San Raffaele Cassino HospitalCassinoItaly
- Department of Human Sciences and Promotion of Quality of LifeSan Raffaele University of RomeRomeItaly
| | - Stephane Heymans
- Centre for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyMaastricht University, CARIM School for Cardiovascular DiseasesMaastrichtThe Netherlands
- European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart (ERN GUARD‐Heart)AmsterdamThe Netherlands
| |
Collapse
|
47
|
Song S, Wang Q, Xie J, Guo Y, He W, Yao Y, Wang H, Huang B, Chen Z, Lin X, He Y, Tian W, Chen Z. A DNA machine-based magnetic resonance imaging nanoprobe for in vivo microRNA detection. Talanta 2025; 281:126867. [PMID: 39277939 DOI: 10.1016/j.talanta.2024.126867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
In situ monitoring microRNA (miRNA) expression in vivo holds immense potential for directly visualizing the occurrence and progression of tumors. However, the significant barrier to developing a probe that can overcome the low abundance of miRNAs while providing an output signal with unlimited tissue penetration depth remains formidable. In this study, we developed a DNA machine-based magnetic resonance imaging nanoprobe (MRINP) for amplified detection of miR-21 in vivo. The MRINP was constructed with superparamagnetic Fe3O4 nanoparticles (NPs), paramagnetic Gd-DOTA complexes, and miR-21-activated DNA machines; the DNA machine was composed of hairpin DNAzyme (HD) strands serving as the DNAzyme walker and hairpin substrate (HS) strands serving as the track. Once uptake into tumor cells, the intracellular miR-21 specifically recognized and hybridized with the HD strand, restoring the activity of DNAzyme. Subsequently, the DNAzyme walker autonomously traveled on the surface of MRINP, and each step movement of the DNAzyme walker resulted in the cleavage of its substrate strands and the ensued release of the Gd-DOTA complex-labeled oligonucleotides, turning on the T1 signal of Gd-DOTA complexes for in situ imaging of miR-21 in tumor-bearing mice. This strategy would offer a promising approach for mapping tumor-specific biomarkers in vivo with unlimited penetration depth.
Collapse
Affiliation(s)
- Sijie Song
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qi Wang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jiangao Xie
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yuheng Guo
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Wen He
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Yuhang Yao
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Hongli Wang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Bingbing Huang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zhitong Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xucong Lin
- Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Yu He
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| | - Wei Tian
- Department of Dermatology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China.
| | - Zhaowei Chen
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
48
|
Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Golshekan M, Abedinzade M, Ahmadi E, Neha S, Najafi M. Revolutionizing Cancer Treatment: Harnessing the Power of Mesenchymal Stem Cells for Precise Targeted Therapy in the Tumor Microenvironment. Curr Top Med Chem 2025; 25:243-262. [PMID: 38797895 DOI: 10.2174/0115680266299112240514103048] [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: 12/17/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
In recent years, mesenchymal stem cells (MSCs) have emerged as promising anti-- cancer mediators with the potential to treat several cancers. MSCs have been modified to produce anti-proliferative, pro-apoptotic, and anti-angiogenic molecules that could be effective against a variety of malignancies. Additionally, customizing MSCs with cytokines that stimulate pro-tumorigenic immunity or using them as vehicles for traditional chemical molecules with anti-cancer characteristics. Even though the specific function of MSCs in tumors is still challenged, promising outcomes from preclinical investigations of MSC-based gene therapy for a variety of cancers inspire the beginning of clinical trials. In addition, the tumor microenvironment (TME) could have a substantial influence on normal tissue stem cells, which can affect the treatment outcomes. To overcome the complications of TME in cancer development, MSCs could provide some signs of hope for converting TME into unequivocal therapeutic tools. Hence, this review focuses on engineered MSCs (En-MSCs) as a promising approach to overcoming the complications of TME.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Rostamzadeh
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Seyed Mohammad Amini
- Radiation Biology Research center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mostafa Golshekan
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahmoud Abedinzade
- Department of Medical Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elham Ahmadi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Singh Neha
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
49
|
Wang X, Jing Z, Huang X, Liu X, Zhang Y, Wang Z, Ma P. PD-L1 antibody conjugated dihydrotanshinone I-loaded polymeric nanoparticle for targeted cancer immunotherapy combining PD-L1 blockade with immunogenic cell death. Int J Pharm 2024; 667:125004. [PMID: 39608587 DOI: 10.1016/j.ijpharm.2024.125004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE Combination immune checkpoint inhibitors (ICI) with chemotherapeutic agents has proven to be highly promising in cancer therapy. However, low response rate, immune-related adverse events, and lack of effectively targeted co-delivery strategy are still major hurdles to overcome for this combination therapeutic regimen. Herein, programmed death-L1 (PD-L1) antibody modified and dihydrotanshinone I (DHT) loaded nanoparticle was prepared for tumor targeting drug delivery, thus achieving immune checkpoint blockade (ICB) and immunogenic cell death (ICD) synergistic anti-tumor effects. METHODS The DHT-loaded nanoparticle (DHT NP) was prepared by the emulsion solvent diffusion method. Atezolizumab (ATEZO) was thiolated with 2-iminothiolane and conjugated to the surface of DHT NP to prepare the ATEZO DHT NP. The drug encapsulation efficiency, drug loading, particle size and drug release were determined. The in vitro cellular uptake, cell proliferation inhibition and apoptosis were evaluated on the HGC-27 tumor cell. The in vivo tumor targeting, anti-tumor efficiency and immune regulation were assessed on tumor bearing mice. RESULTS The optimized ATEZO DHT NP was a spherical nanoparticle of about 250 nm with a continuous drug release profile. It was selectively taken up by the tumor cells through PD-L1 receptor-mediated endocytosis, which resulted in enhanced cytotoxicity and cell apoptosis. In vivo imaging further demonstrated its superior tumor tissue targeting ability. When tumor bearing mice were treated with the ATEZO DHT NP, its synergistic anti-tumor effect was much stronger than that of a single drug. Moreover, the tumor targeting delivery of DHT caused tumor necrosis and initiated ICD with release of tumor-associated antigens, which efficiently up-regulated the population of CD4+ and CD8+ T cells. Notably, there were no obvious system toxicity or tissue damage occur during the whole treatment period. CONCLUSION The ATEZO DHT NP could specifically target to tumor and enhance treatment efficiency through combination of PD-L1 blockade with ICD effect.
Collapse
Affiliation(s)
- Xue Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqi Jing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaobin Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoya Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yujie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhijun Wang
- Department of Geriatric Medicine &National Clinical Research Center of Geriatric Disease, The 2nd Medical Center of Chinese PLA General Hospital, Beijing, China; Department of Interventional Radiology, The 1st & 5th Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Pengkai Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
50
|
Liu D, Lu N, Zang F, Lu M, Zhang J, Zhao Y, Wan H, Wang M, Li QQ, Wang F, Luo S, Ma M, Shi F, Wu H, Tu J, Zhang Y. Magnetic Resonance Imaging-Based Radiogenomic Analysis Reveals Genomic Determinants for Nanoparticle Delivery into Tumors. ACS NANO 2024; 18:34615-34629. [PMID: 39663893 DOI: 10.1021/acsnano.4c09387] [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: 12/13/2024]
Abstract
Even though the enhanced permeability and retention (EPR) effect is applicable for the passive targeting of solid tumors, many nanodrugs have failed to achieve meaningful clinical outcomes due to the heterogeneity of EPR effect. Therefore, understanding the mechanism of the EPR effect is crucial to overcome the obstacles nanomedicines face in clinical translation. The aim of this study was to establish a reliable method to increase awareness of the critical influencing factors of nanoparticle (NP) transport into tumors based on the EPR effect using a combined radiogenomics and clinical magnetic resonance imaging (MRI) technique and gene set pathway enrichment analysis. Employing poly(lactic-co-glycolic acid) (PLGA)-coated Fe3O4 NPs as the contrast agent, the monolayer and multilayer distribution of the NPs were observed and quantitatively analyzed by MRI, improving the accuracy of evaluating vascular permeability by MRI. By performing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of genes and pathways, we identified a variety of genes affecting vascular permeability, such as Cldn1, Dlg2, Bves, Prkag3, Cldn10, and Cldn8, which are related to tight junctions and control the permeability of blood vessels in tumors. The method presented here provides an MRI-supported approach to increase the breadth of data collected from genetic screens, reveals genetic evidence of the presence of NPs in tumors and lays a foundation for clinical patient stratification and personalized treatment.
Collapse
Affiliation(s)
- Di Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Na Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fengchao Zang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Mingze Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jingyue Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Hao Wan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Mengjun Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Qian-Qian Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Shouhua Luo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Ming Ma
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210096, P. R. China
| | - Haoan Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China
| |
Collapse
|