1
|
Liu J, Sun J, Hu J, Xue H, Lei L, Pan X. Biomaterial-based drug delivery strategies for oral mucosa. Colloids Surf B Biointerfaces 2025; 251:114604. [PMID: 40081256 DOI: 10.1016/j.colsurfb.2025.114604] [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: 12/14/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/15/2025]
Abstract
Drug therapy is a key field in modern medicine, and the optimization of its delivery method is crucial. Traditional methods are inherently limited by first-pass effects, high-risk adverse reactions, and patient compliance challenges, which significantly restrict the effectiveness and application potential of drugs. Oral mucosal drug delivery has become a minimally invasive and effective drug delivery strategy. The unique anatomical structure of the oral mucosa facilitates the rapid absorption of drugs into the systemic circulation, thus producing rapid therapeutic effects. However, a complex oral microenvironment and mucosal barrier impede drug absorption. Biomaterials have become an important driving force for the innovative development of oral medicine, owing to their unique and excellent properties. They are widely used for preventing, diagnosing, treating, and rehabilitating oral diseases. This review explores recent advancements in biomaterial-enabled oral mucosal drug delivery systems, analyzing key physiological factors and absorption barriers that impact therapeutic outcomes. Focusing on innovative material engineering strategies highlights significant progress in extending drug residence time and improving delivery precision within the oral cavity. Furthermore, the study identifies critical challenges in translating these advancements from research to clinical practice, emphasizing the need for solutions to bridge this gap.
Collapse
Affiliation(s)
- Junhui Liu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jiao Sun
- Changsha Stomatological Hospital, Changsha 410000, China
| | - Jun Hu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Huaqian Xue
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China.
| | - Xiaoyi Pan
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| |
Collapse
|
2
|
Sun G, Wu Y, Li J, Yang M, Xu H, Li Y, Tong P, Shao R, Liu Y, Kong X. Quercetin liposomes conjugated with hyaluronidase: An efficient drug delivery system to block pancreatic cancer. J Control Release 2025; 382:113642. [PMID: 40127723 DOI: 10.1016/j.jconrel.2025.113642] [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: 11/26/2024] [Revised: 02/03/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025]
Abstract
Pancreatic cancer characterized with intense hydraulic tissue in tumor extracellular matrix (ECM) resists most of chemotherapeutic drugs. Increased levels of hyaluronic acid (HA) represent the primary component of the hydraulic tissue, rendering tumors protective from drug targeting. Quercetin (Que), a natural flavonoid, has the ability to inhibit tumor cell growth in a number of cancers; however, its poor water solubility and low bioavailability largely limit its application in cancer therapy. Hence, we developed an efficient drug delivery system by encapsulation of Que. into liposomes and conjugation with hyaluronidase (HAase) at liposome surface, termed as HQL. In the presence of HAase, HQL were predominantly accumulated at tumor with enhanced permeability and retention effect. Treatment of xenografted tumor mice with HQL gave rise to suppressed tumor growth, while no toxic effects were observed in mice. HQL demonstrated the strong ability to inhibit cell proliferation, promote cell apoptosis, and induce arrest at G2/M cell cycle in pancreatic cancer lines, three-dimensional cultured cell spheroids and pancreatic ductal adenocarcinoma (PDAC)-derived organoids. Mechanistically, HQL downregulated expression of cell cycle-associated protein (CCNB1, CDK1 and PLK1) and cell apoptosis-associated factors PI3K/AKT and Bcl-2. In summary, HQL degraded HA in the tumor microenvironment to enhance nano-particle penetration and inhibited tumor cell growth, eliciting efficacy of anti-tumor therapy. Thereof, HQL may provide a novel efficient drug delivery approach for the adjuvant treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ge Sun
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Ying Wu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Jiekai Li
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Mingjie Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hang Xu
- Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China
| | - Yiping Li
- Department of Oncology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430065, China
| | - Peilin Tong
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Shao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China; Shanghai Key Laboratory of Biliary Tract Diseases, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of General Surgery, Jiading Branch, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201800, China; Shanghai Key Laboratory of Systems Regulation and Clinical Translation for Cancer, Shanghai 200127, China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai 200127, China.
| | - Xianming Kong
- Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Shanghai University of Medicine & Health Sciences, Shanghai 201318, China.
| |
Collapse
|
3
|
Ai Z, Liu B, Chen J, Zeng X, Wang K, Tao C, Chen J, Yang L, Ding Q, Zhou M. Advances in nano drug delivery systems for enhanced efficacy of emodin in cancer therapy. Int J Pharm X 2025; 9:100314. [PMID: 39834843 PMCID: PMC11743866 DOI: 10.1016/j.ijpx.2024.100314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/05/2025] Open
Abstract
Cancer remains one of the leading causes of death worldwide, highlighting the urgent need for novel antitumor drugs. Natural products have long been a crucial source of anticancer agents. Among these, emodin (EMO), a multifunctional anthraquinone compound, exhibits significant anticancer effects but is hindered in clinical applications by challenges such as low solubility, rapid metabolism, poor bioavailability, and off-target toxicity. Nano drug delivery systems offer effective strategies to overcome these limitations by enhancing the solubility, stability, bioavailability, and targeting ability of EMO. While substantial progress has been made in developing EMO-loaded nanoformulations, a comprehensive review on this topic is still lacking. This paper aims to fill this gap by providing an overview of recent advancements in nanocarriers for EMO delivery and their anticancer applications. These carriers include liposomes, nanoparticles, polymeric micelles, nanogels, and others, with nanoparticle-based formulations being the most extensively explored. Nanoformulations encapsulating EMO have demonstrated promising therapeutic results against various cancers, particularly breast cancer, followed by liver and lung cancers. We systematically summarize the preparation methods, materials, and physicochemical properties of EMO-loaded nanopreparations, underscoring key findings on how nanotechnology improves the anticancer efficacy of EMO. This review provides valuable insights for researchers engaged in developing nano delivery systems for anticancer drugs.
Collapse
Affiliation(s)
- Zhenghao Ai
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Bingyao Liu
- Department of Radiology, West China Hospital Sichuan University Jintang Hospital, Chengdu, China
| | - Junyan Chen
- Department of Cardiothoracic Surgery, Luzhou People's Hospital, Luzhou, China
| | - Xinhao Zeng
- Department of Pediatric Surgery, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Ke Wang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chao Tao
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jing Chen
- Department of Clinical Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Liuxuan Yang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qian Ding
- Department of Clinical Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
Luo Q, Xing X, Song Y, Gu B, Hu Q, Liu W, Xiao Y, Wang Z. MiR-29a-3p ameliorate behavioral deficiency in hypoxia-ischemia brain damage in neonatal mice by inhibiting BTG2. Behav Brain Res 2025; 486:115552. [PMID: 40147793 DOI: 10.1016/j.bbr.2025.115552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
It has been reported that miR-29a-3p played a part in series neurological disorders. However, it remains unclear whether miR-29a-3p participate in the pathological mechanism in hypoxia-ischemia (HI) brain injury. In this study, we detected the change of miR-29a-3p level in the ipsilateral cortex following HI brain injury and found that miR-29a-3p was significantly increased at 3 days in the ipsilateral cortex following HI insult in neonatal mice. Therefore, we further explored the role of miR-29a-3p in HI brain injury and its molecular mechanism. The results showed that miR-29a-3p mimics attenuated and miR-29a-3p antagomir aggravated brain infarction volume at 3 days following HI insult. We further found that overexpression of miR-29a-3p also suppressed apoptosis and neuroinflammation, reduced synaptic loss and prevent HI-induced microglial morphological changes 3 days following HI insult. Neurobehavioral tests revealed that overexpression of miR-29a-3p could improve both short-term and long-term behavioral defects after HI injury. Furthermore, we proved that miR-29a-3p targets B-cell translocation gene 2 (BTG2) and further inhibits the expression of Bax by luciferase reporter assay and qRT-PCR. Moreover, overexpression of miR-29a-3p, by applying liposomes through intranasal route, could also achieve the same therapeutic effect in HI injury. Our data showed that by inhibiting BTG2/Bax, increasing level of miR-29a-3p might serve as a strategy to prevent brain damage and behavioral deficiency in HI.
Collapse
Affiliation(s)
- Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaohui Xing
- Department of Neurosurgery, Liaocheng Neuroscience Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China; Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China
| | - Quan Hu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong 271000, PR China
| | - Weiyang Liu
- Jinan Xicheng Experimental High School, Dezhou Road, Jinan, Shandong 1999, PR China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng Neuroscience Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China.
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, PR China.
| |
Collapse
|
5
|
Xu W, Li X, He F, Zhao H, Wu J, Li M, Dai X, Li Y, Hu X, Li X, Cen J, Guo P, Duan S. Folate receptor-targeted pH-sensitive liposomes loaded with TGX-221 against prostate cancer by inhibiting PI3K/110β signaling. NANOSCALE ADVANCES 2025; 7:3267-3280. [PMID: 40212450 PMCID: PMC11979785 DOI: 10.1039/d5na00009b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/05/2025] [Indexed: 05/29/2025]
Abstract
Prostate cancer (PCa) is the most common cancer in men and the leading cause of cancer death worldwide. Overactivation of PI3K signaling has been reported to be associated with PCa. TGX221 is an effective specific inhibitor of PI3K, but its clinical application is greatly limited due to its poor solubility. Herein, by using folic acid-PEG-cholesterol semi-succinate (FA-PEG-CHEMS) as the targeting component, we developed a folate receptor-targeted pH-sensitive liposomal delivery system loaded with TGX221 (FA-Lip-TGX221) that could realize effective delivery and controlled release of drugs in the tumor. The prepared liposomes exhibited a uniform particle size and high stability. In addition, FA-Lip-TGX221 could be effectively internalized by PC-3 cells due to its ability to target folate receptors, thereby accumulating in tumor tissues. Meanwhile, in vitro and in vivo experiments suggested that FA-Lip-TGX221 could activate the PERK-ATF4-CHOP signaling pathway by inhibiting PI3K/110β signaling in PCa, thus significantly promoting endoplasmic reticulum (ER) stress-mediated cancer cell death. In conclusion, FA-Lip-TGX221 is a promising nano-delivery vehicle for the treatment of PCa, and also provide valuable references for all tumors overexpressing folate receptors.
Collapse
Affiliation(s)
- Weibo Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Medical School, Henan University Kaifeng 475004 China
| | - Xiaohan Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Fujin He
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Han Zhao
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Jing Wu
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Mengyu Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaoying Dai
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Yanmin Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaojiao Hu
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaodong Li
- Medical School, Henan University Kaifeng 475004 China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
- The First Affiliated Hospital of Henan University Kaifeng Henan 475004 PR China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
| | - Shaofeng Duan
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
- The First Affiliated Hospital of Henan University Kaifeng Henan 475004 PR China
| |
Collapse
|
6
|
Stępień M, Zajda J, Keppler BK, Timerbaev AR, Matczuk M. Cisplatin meets liposomes for a smarter delivery: A review. Talanta 2025; 295:128331. [PMID: 40382863 DOI: 10.1016/j.talanta.2025.128331] [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: 03/11/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Effective treatment of tumors remains a significant clinical challenge even for approved anticancer drugs such as cisplatin, whose chemotherapy is hindered by inherent toxicity, rapidly acquired resistance, and nonselective mode of action. In the past years, nanodelivery systems have emerged as a key strategy to overcome these limitations due to their potential to improve drug safety, bioavailability, and efficacy. Among various nanostructures applied as carriers for the delivery of cisplatin, liposomes have undergone intensive testing, with the outcome of being advanced to clinical trials. This fact not only triggers further research endeavors toward developing improved liposomal formulations but also makes it timely to highlight recent trends and strategies, showcasing the evolution and application of cisplatin-liposome systems. The present review is aimed at a critical analysis of fabrication, encapsulation, stability testing, release, and cell/animal experimental procedures, focusing on the analytical methodology used to feature these essential practices and providing insights that may help enhance the efficacy of cisplatin chemotherapy.
Collapse
Affiliation(s)
- Marta Stępień
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Stanisława Noakowskiego 3, 00-664, Warsaw, Poland
| | - Joanna Zajda
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Stanisława Noakowskiego 3, 00-664, Warsaw, Poland.
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Währinger Str. 42, 1090, Vienna, Austria
| | - Andrei R Timerbaev
- Institute of Inorganic Chemistry, University of Vienna, Währinger Str. 42, 1090, Vienna, Austria
| | - Magdalena Matczuk
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Stanisława Noakowskiego 3, 00-664, Warsaw, Poland.
| |
Collapse
|
7
|
Togami K, Kanehira Y, Nakamura Y, Ishii H, Abe R, Yamamoto A, Takehara K, Yasuda M, Tada H, Chono S. Pirfenidone encapsulated in succinylated gelatin-coated liposomes exhibits sustained antifibrotic effects in vitro models of renal, pulmonary, and hepatic fibrosis. J Pharm Sci 2025; 114:103819. [PMID: 40345528 DOI: 10.1016/j.xphs.2025.103819] [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: 03/09/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
Fibrosis is characterized by excessive extracellular matrix accumulation, leading to organ dysfunction and irreversible damage in advanced stages. Challenges in sustaining drug levels within fibrotic lesions with the currently used antifibrotic therapies, including pirfenidone, often necessitate high drug doses that can cause systemic side effects. Here, we introduce a succinylated gelatin (SG)-coated liposome (SG-lip) system, which enhances pirfenidone retention and enables enzyme-responsive release at sites of fibrosis in an in vitro model. The SG coating, which ensures high collagen-binding affinity, is degraded by matrix metalloproteinases, which are overexpressed in fibrotic tissues, allowing targeted drug release. In vitro experiments using NRK-49F (kidney fibroblasts), WI-38 (lung fibroblasts), and RI-T (hepatic stellate cells) cultured on collagen I gel, SG-lip prolongs drug retention and sustains localized release at sites of fibrosis. In experiments simulating transient drug exposure by washing away the residual pirfenidone after treatment, pirfenidone-loaded SG-lip significantly inhibit fibroblast proliferation, invasion, and myofibroblast differentiation. Our enzyme-triggered drug delivery system enhances the antifibrotic efficacy of pirfenidone, with the potential to reduce systemic exposure and associated side effects. These findings highlight SG-lip as a promising platform for targeted antifibrotic therapy, offering a novel strategy to improve treatment of fibrosis.
Collapse
Affiliation(s)
- Kohei Togami
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan; Creation Research Institute of Life Science in KITA-no-DAICHI, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan; Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Hokkaido Pharmaceutical University, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8590, Japan.
| | - Yukimune Kanehira
- Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Hokkaido Pharmaceutical University, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8590, Japan
| | - Yuki Nakamura
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Hirotsugu Ishii
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Ryota Abe
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Akiyoshi Yamamoto
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Kanako Takehara
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Mio Yasuda
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan
| | - Hitoshi Tada
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan; Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Hokkaido Pharmaceutical University, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8590, Japan
| | - Sumio Chono
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan; Creation Research Institute of Life Science in KITA-no-DAICHI, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8585, Japan; Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Hokkaido Pharmaceutical University, 7-Jo 15-4-1 Maeda, Teine, Sapporo, Hokkaido 006-8590, Japan
| |
Collapse
|
8
|
Gu Y, Yu W, Li X, Fan Y, Liu Y, Tummatorn J, Jiang S, Yang J. Molecular Engineering of Cordycepin Derivatives for Enhanced Biological Activity and Stability. ChemMedChem 2025; 20:e202400979. [PMID: 39906978 DOI: 10.1002/cmdc.202400979] [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: 12/04/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025]
Abstract
To address the metabolic instability of cordycepin induced by adenosine deaminase (ADA) and to enhance its bioactivity, this study developed eleven novel cordycepin derivatives using molecular engineering techniques. By incorporating sterically hindered protective groups and modifying the glycosyl moiety, the research aimed to improve both stability and efficacy. Antibacterial tests revealed that five derivatives showed significantly greater activity against pathogenic strains compared to cordycepin, with better compatibility with probiotics. Compound 2 c demonstrated moderate antitumor activity against K562 and MGC-803 cells, with IC50 values of 42.21 μM and 27.79 μM, respectively. Additionally, compound 4 b demonstrated notable DPPH free radical scavenging ability. These compounds also showed improved stability in ADA solutions, providing valuable insights into the structure-activity relationships of cordycepin derivatives.
Collapse
Affiliation(s)
- Yiming Gu
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Wei Yu
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Xiang Li
- Qinhuangdao Customs Technology Center, Qinhuangdao, Hebei, 066004, China
| | - Yingjie Fan
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Yanan Liu
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Jumreang Tummatorn
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, Bangkok, 10400, Thailand
| | - Siyu Jiang
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| | - Jingyue Yang
- State Key Laboratory of Metastable Materials Science and Technology, Nano-biotechnology Key Lab of Hebei Province, Applying Chemistry Key Lab of Hebei Province, Yanshan University, Qinhuangdao, 066004, China
| |
Collapse
|
9
|
Zhou Y, Ge Q, Wang X, Wang Y, Sun Q, Wang J, Yang T, Wang C. Advances in Lipid Nanoparticle-Based Disease Treatment. ChemMedChem 2025; 20:e202400938. [PMID: 39962990 DOI: 10.1002/cmdc.202400938] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/11/2025] [Indexed: 05/09/2025]
Abstract
Lipid nanoparticles (LNPs) have emerged as a transformative platform for the targeted delivery of therapeutic agents, revolutionizing treatment paradigms across a spectrum of diseases. Since the inception of liposomes in the 1960s, lipid-based nanotechnology has evolved to address limitations such as poor bioavailability, off-target effects, and instability, thereby enhancing the efficacy and safety of drug administration. This review highlights the latest advancements in LNPs technology, focusing on their application in cancer therapy, gene therapy, infectious disease management, glaucoma, and other clinical areas. Recent studies underscore the potential of LNPs to deliver messenger RNA (mRNA) and small interfering RNA (siRNA) for precise genetic intervention, exemplified by breakthroughs in RNA interference and CRISPR-Cas9 genome editing. Additionally, LNPs have been successfully employed to ameliorate conditions, demonstrating their versatility in addressing both acute and chronic disorders. However, challenges persist concerning large-scale manufacturing, long-term stability, and comprehensive safety evaluations. Future research must focus on optimizing formulations, exploring synergistic combinations with existing therapies, and expanding the scope of treatable diseases. The integration of LNPs into personalized medicine and the exploration of applications in other diseases represent promising avenues for further investigation. LNPs are poised to play an increasingly central role in the development of next-generation therapeutics.
Collapse
Affiliation(s)
- Yujie Zhou
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| | - Qiqi Ge
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| | - Xin Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| | - Yuhui Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| | - Qianqian Sun
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| | - Tie Yang
- Chia Tai Tianqing Pharmaceutical Group Co., LTD, Nanjing, 211100, Jiangsu, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu, China (Jianhao Wang), (Cheng Wang
| |
Collapse
|
10
|
Kart U, Raimbekova A, Yegorov S, Hortelano G. Immune Modulation with Oral DNA/RNA Nanoparticles. Pharmaceutics 2025; 17:609. [PMID: 40430900 PMCID: PMC12115334 DOI: 10.3390/pharmaceutics17050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 05/29/2025] Open
Abstract
The oral delivery of DNA/RNA nanoparticles represents a transformative approach in immunotherapy and vaccine development. These nanoparticles enable targeted immune modulation by delivering genetic material to specific cells in the gut-associated immune system, triggering both mucosal and systemic immune responses. Unlike parenteral administration, the oral route offers a unique immunological environment that supports both tolerance and activation, depending on the formulation design. This review explores the underlying mechanisms of immune modulation by DNA/RNA nanoparticles, their design and delivery strategies, and recent advances in their application. Emphasis is placed on strategies to overcome physiological barriers such as acidic pH, enzymatic degradation, mucus entrapment, and epithelial tight junctions. Special attention is given to the role of gut-associated lymphoid tissue in mediating immune responses and the therapeutic potential of these systems in oral vaccine platforms, food allergies, autoimmune diseases, and chronic inflammation. Despite challenges, recent advances in nanoparticle formulation support the translation of these technologies into clinical applications for both therapeutic immunomodulation and vaccination.
Collapse
Affiliation(s)
| | | | | | - Gonzalo Hortelano
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Ave., Astana 010000, Kazakhstan; (U.K.); (A.R.); (S.Y.)
| |
Collapse
|
11
|
Xiao Y, Tong Z, Xu H, Jia Z, Wang C, Cao Y, Song L, Hao S, Yang J, Zhou Y, Xie Y, Wu P, He T, Wu Y, Petersen RB, Peng A, Zhang C, Chen H, Zheng L, Huang K. A rationally designed injury kidney targeting peptide library and its application in rescuing acute kidney injury. SCIENCE ADVANCES 2025; 11:eadt3943. [PMID: 40315322 PMCID: PMC12047437 DOI: 10.1126/sciadv.adt3943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Acute kidney injury (AKI) has high incidence and mortality rates. Present treatments are mostly symptomatic and cause side effects due to systemic distribution; thus, targeted kidney drug delivery is desired. Transmembrane kidney injury molecule-1 (KIM1) is expressed at low levels in normal kidneys but markedly up-regulated following injury, making it an ideal marker/target for injured kidneys. Here, assisted by AlphaFold, we constructed a library of 1885 peptides that target the extracellular Ig V domain of KIM1 based on interacting fragments from 47 potential KIM1 binding proteins followed by systemic optimization according to their binding energies with KIM1. Experimental validation of top candidates (TKP 1-5) demonstrated that TKP 4 efficiently targeted injured renal cells/kidneys, with its specificity demonstrated in KIM1 knockout cells/mice. TKP 4-decorating liposomes were loaded with nystatin, a renal-protective compound with systemic side effects, and efficiently targeted injured mouse kidneys and alleviated AKI. This work establishes a virtual platform to screen/identify drug delivery candidates with broad research/therapeutic potentials.
Collapse
Affiliation(s)
- Yushuo Xiao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhijian Tong
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huidie Xu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhouyan Jia
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Wang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Cao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liangliang Song
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Siyu Hao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Yang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yihao Zhou
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Yunhao Xie
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Peng Wu
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Tong He
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yancai Wu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Robert B. Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI 48859, USA
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Tongren Hospital of Wuhan University, Wuhan 430070, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hong Chen
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Zheng
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan 430030, China
- Tongji-RongCheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
12
|
Cui Y, Song M, Liu R, Xi Z, Zhao L, Cen L. Microfluidic development of liposome nanoparticles encapsulated with yam polysaccharide. J Pharm Sci 2025; 114:103718. [PMID: 40054528 DOI: 10.1016/j.xphs.2025.103718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 04/29/2025]
Abstract
A novel liposome nanoparticle loaded with yam polysaccharide was developed in this study via a tailor-made microfluidic device and further optimized through the Box-Behnken Design method of surface analysis to exploit combinational immunomodulatory effects of liposomes and yam polysaccharide. A chip of 100 μm in height and 200 μm in width with a mixing channel with embedded baffles to enhance fluid mixing was first designed and fabricated. Liposome nanoparticles were obtained by manipulating the operating parameters of the microfluidic chip. The formulation of yam polysaccharide loaded liposomes (YPL1) was optimized using response-surface analysis, and their physicochemical properties were characterized. In vitro cellular assays were performed to assess the effect of YPL on the activity of mouse dendritic cells (DC) and the secretion of immune-related cytokines to investigate the immune-enhancing effect of YPL in vitro. The resulting YPL had a mean size of 154.2±3.8 nm with a narrow size distribution (PDI=0.083) and had a high entrapment efficiency (EE) of 79.02%. The YPL exhibited good stability at 4°C over 14 days of storage, and an in vitro sustained release duration of approximately 30 h. The YPL demonstrated excellent biocompatibility and stimulated the expression of immune-related cytokines, especially TNF-α. Therefore, the YPL with well controlled particle size and high loading capacity, capable of exhibiting potent immunostimulatory activity was successfully developed in this study via a microfluidic device and optimized using response-surface analysis. The current YPL engineering methodology could further serve as an experimental platform for liposome nanoparticle development.
Collapse
Affiliation(s)
- Yuehan Cui
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Meng Song
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200000, PR China
| | - Ruilai Liu
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200000, PR China
| | - Zhenhao Xi
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200000, PR China.
| | - Ling Zhao
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, PR China; Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200000, PR China.
| | - Lian Cen
- Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200000, PR China.
| |
Collapse
|
13
|
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
|
14
|
Kelley M, Sasaninia K, Badaoui A, Glassman I, Abnousian A, Rai N, Tiwari RK, Venketaraman V. The effects of cyclic peptide [R4W4] in combination with first-line therapy on the survival of Mycobacterium avium. Front Cell Infect Microbiol 2025; 15:1547376. [PMID: 40308965 PMCID: PMC12041069 DOI: 10.3389/fcimb.2025.1547376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
Background Mycobacterium avium (M. avium) is a nontuberculous mycobacterium (NTM) that can cause pulmonary and extrapulmonary infections mostly in immunocompromised individuals, such as those with HIV and diabetes. Traditionally, rifampicin (RIF) and azithromycin (AZ) have been used for a 12-month duration as first-line antibiotics against M. avium. Due to the increased multidrug resistance, novel ways, such as enhancement of macrophages response, are needed to provide adequate immune response required to clear M. avium infection. Methods and findings In this study, we aim to study the effects of using THP-1 cells, which are monocyte-like cells, to induce a macrophage response and control M. avium infection when used in combination with traditional treatments such as RIF and AZ in free and liposomal forms. Traditional treatments' effects are studied when used alone and in combination therapy with cyclic peptide [R4W4] (liposomal encapsulated and liposomal combination). Colony-forming units (CFU) counts were assessed for all samples 3 hours, 4 days, and 8 days post-treatment. A significant reduction in the intracellular viability of M. avium was observed when THP-1 cells were treated with liposomal combination [R4W4]+RIF and liposomal combination [R4W4]+AZ compared to when treated with liposomal RIF or liposomal AZ alone, respectively. Conclusion Our findings show that liposomal combination [R4W4] is a promising adjuvant therapy to increase M. avium susceptibility to known antibiotics.
Collapse
Affiliation(s)
- Melissa Kelley
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Kayvan Sasaninia
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Ali Badaoui
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Ira Glassman
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Arbi Abnousian
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Nadia Rai
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Rakesh K. Tiwari
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific-Northwest, Western University of Health Sciences, Lebanon, OR, United States
| | - Vishwanath Venketaraman
- Department of Biomedical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
15
|
Alrumaihi F, Babiker AY, Khan A. Lipid-Based Nanoformulations of [6]-Gingerol for the Chemoprevention of Benzo[a] Pyrene-Induced Lung Carcinogenesis: Preclinical Evidence. Pharmaceuticals (Basel) 2025; 18:574. [PMID: 40284009 PMCID: PMC12030401 DOI: 10.3390/ph18040574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: [6]-Gingerol ([6]-G), a bioactive compound derived from Zingiber officinale (ginger), exhibits strong anticancer potential but is hindered by poor aqueous solubility and low bioavailability. This study aimed to develop and evaluate PEGylated liposomal [6]-G (6-G-Lip) to enhance its stability, bioavailability, and chemopreventive efficacy in benzo[a]pyrene (BaP)-induced lung carcinogenesis. Methods: 6-G-Lip was synthesized using a modified thin-film hydration technique and characterized for size, polydispersity index (PDI), zeta potential, encapsulation efficiency (EE%), and release kinetics. The chemopreventive effects were assessed in BaP-induced lung cancer in Swiss albino mice, with prophylactic 6-G-Lip administration from two weeks before BaP exposure through 21 weeks. Cancer biomarkers, antioxidant enzyme activity, reactive oxygen species (ROS) generation, induction of apoptosis, and histopathological alterations were analyzed. Results: 6-G-Lip exhibited a particle size of 129.7 nm, a polydispersity index (PDI) of 0.16, a zeta potential of -18.2 mV, and an encapsulation efficiency (EE%) of 91%, ensuring stability and effective drug loading. The formulation exhibited a controlled release profile, with 26.5% and 47.5% of [6]-G released in PBS and serum, respectively, at 72 h. 6-G-Lip significantly lowered cancer biomarkers, restored antioxidant defenses (SOD: 5.60 U/min/mg protein; CAT: 166.66 μm H2O2/min/mg protein), reduced lipid peroxidation (MDA: 3.3 nm/min/mg protein), and induced apoptosis (42.2%), highlighting its chemopreventive efficacy. Conclusions: This study is the first to prepare, characterize, and evaluate PEGylated [6]-G-Lip for the chemoprevention of lung cancer. It modulates oxidative stress, restores biochemical homeostasis, and selectively induces apoptosis. These findings support 6-G-Lip as a promising nanotherapeutic strategy for cancer prevention.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (F.A.); (A.Y.B.)
| | - Ali Yousif Babiker
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (F.A.); (A.Y.B.)
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
16
|
Wong CN, Lee SK, Lim YM, Yang SB, Chew YL, Chua AL, Liew KB. Recent Advances in Vitamin E TPGS-Based Organic Nanocarriers for Enhancing the Oral Bioavailability of Active Compounds: A Systematic Review. Pharmaceutics 2025; 17:485. [PMID: 40284480 PMCID: PMC12030195 DOI: 10.3390/pharmaceutics17040485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Background: D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), an amphiphilic derivative of natural vitamin E, functions as both a drug efflux inhibitor and a protector against enzymatic degradation and has been widely incorporated into nano-formulations for drug design and delivery. Objective: This systematic review evaluates TPGS-based organic nanocarriers, emphasizing their potential to enhance bioavailability of active compounds which include drugs and phytochemicals, improve pharmacokinetic profiles, and optimize therapeutic outcomes, eventually overcoming the limitations of conventional oral active compounds delivery. Search strategy: Data collection was carried out by entering key terms (TPGS) AND (Micelle OR Liposome OR Nanoparticle OR Nanotube OR Dendrimer OR Niosome OR Nanosuspension OR Nanomicelle OR Nanocrystal OR Nanosphere OR Nanocapsule) AND (Oral Bioavailability) into the Scopus database. Inclusion criteria: Full-text articles published in English and relevant to TPGS, which featured organic materials, utilized an oral administration route, and included pharmacokinetic study, were included to the final review. Data extraction and analysis: Data selection was conducted by two review authors and subsequently approved by all other authors through a consensus process. The outcomes of the included studies were reviewed and categorized based on the types of nanocarriers. Results: An initial search of the database yielded 173 records. After screening by title and abstract, 52 full-text articles were analyzed. A total of 21 papers were excluded while 31 papers were used in this review. Conclusions: This review concludes that TPGS-based organic nanocarriers are able to enhance the bioavailability of various active compounds, including several phytochemicals, leveraging TPGS's amphiphilic nature, inhibition of efflux transporters, protection against degradation, and stabilization properties. Despite using the same excipient, variability in particle size, zeta potential, and encapsulation efficiency among nanocarriers indicates the need for tailored formulations. A comprehensive approach involving the development and standardized comparison of diverse TPGS-incorporated active compound formulations is essential to identify the optimal TPGS-based nanocarrier for improving a particular active compound's bioavailability.
Collapse
Affiliation(s)
- Chee Ning Wong
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia; (C.N.W.); (Y.M.L.)
| | - Siew-Keah Lee
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia; (C.N.W.); (Y.M.L.)
| | - Yang Mooi Lim
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Malaysia; (C.N.W.); (Y.M.L.)
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan;
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia;
| | - Ang-Lim Chua
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh 47000, Malaysia;
| | - Kai Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Cyberjaya 63000, Malaysia
| |
Collapse
|
17
|
Li G, Chen Y, Zhang X, Tang A, Yang H. Advances in Microfluidics-Enabled Dimensional Design of Micro-/Nanomaterials for Biomedical Applications: A Review. ACS APPLIED MATERIALS & INTERFACES 2025; 17:19203-19229. [PMID: 40105107 DOI: 10.1021/acsami.4c22581] [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: 03/20/2025]
Abstract
Biomedical materials are of great significance for preventing and treating major diseases and protecting human health. At present, more stringent requirements have been put forward for the preparation methods and dimension control of biomedical materials based on the urgent demand for high-performance biomedical materials, especially the existence of various physiological size thresholds in vitro/in vivo. Microfluidic platforms break the limitations of traditional micro-/nanomaterial synthesis, which provide a miniaturized and highly controlled environment for size-dependent biomaterials. In this review, the basic conceptions and technical characteristics of microfluidics are first described. Then the syntheses of biomedical materials with different dimensions (0D, 1D, 2D, 3D) driven by microfluidics have been systematically summarized. Meanwhile, the applications of microfluidics-driven biomedical materials, including diagnosis, anti-inflammatory, drug delivery, antibacterial, and disease therapy, are discussed. Furthermore, the challenges and developments in the research field are further proposed. This work is expected to facilitate the convergence between the bioscience and engineering communities and continue to contribute to this emerging field.
Collapse
Affiliation(s)
- Guangyao Li
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Laboratory of Advanced Mineral Materials, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Ying Chen
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Laboratory of Advanced Mineral Materials, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Xuming Zhang
- Department of Applied Physics, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Aidong Tang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Laboratory of Advanced Mineral Materials, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Huaming Yang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Laboratory of Advanced Mineral Materials, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Hunan Key Laboratory of Mineral Materials and Application, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China
| |
Collapse
|
18
|
Li M, Liu F, Guo L, Fan W, Wang J, Lu B, Hong G, Zhang W, Tian S, Mao J, Xie J. Distribution and Pharmacokinetic Characteristics of Cordycepin in Rat: Investigated by UHPLC-HRMS/MS and Blood-Brain Synchronous Microdialysis. Biomed Chromatogr 2025; 39:e70038. [PMID: 40071940 DOI: 10.1002/bmc.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/11/2025] [Accepted: 02/12/2025] [Indexed: 05/14/2025]
Abstract
Cordycepin, a natural adenosine derivative, exhibits multiple pharmacological effects on organisms. However, its distribution and metabolic characteristics have not been fully elucidated in vivo. In this study, ultra-high liquid chromatography tandem high-resolution mass spectrometry (UHPLC-HRMS/MS) was used to investigate the pharmacokinetic characteristics and effects of cordycepin on endogenous adenosine and inosine. Microdialysis was used for real-time monitoring of unbound drug in brain and blood, whereas conventional tissue homogenate methods assessed distribution in various tissues. Results showed that the distribution pattern of cordycepin was as follows: kidney > liver > heart > lung > spleen > brain. Cordycepin administration significantly altered the levels of adenosine and inosine in heart and liver. Synchronous microdialysis sampling for the pharmacokinetic profile indicated that cordycepin was rapidly consumed and 3'-deoxyinosine was generated as the main metabolite. The Cmax values of cordycepin in the rat blood and brain after exposure (10 mg/kg, i.p.) were 7.8 and 5.4 ng/mL, respectively. Mean residence time in blood and brain was 102.2 and 137.0 min, respectively. Inhibition of adenosine deaminase by racemic 9-(2-hydroxy-3-nonyl) adenine hydrochloride (EHNA) enhanced cordycepin levels in the blood. This work provides a solid basis for understanding the metabolism of cordycepin and its pharmacological effects.
Collapse
Affiliation(s)
- Mengjiao Li
- Flavour Science Research Center, College of Chemistry, Zhengzhou University, Zhengzhou, China
- Beijing Life Science Academy (BLSA), Beijing, China
| | - Fuqiang Liu
- Flavour Science Research Center, College of Chemistry, Zhengzhou University, Zhengzhou, China
- Beijing Life Science Academy (BLSA), Beijing, China
| | - Lulu Guo
- Beijing Life Science Academy (BLSA), Beijing, China
| | - Wu Fan
- Zhengzhou Tobacco Research Institute of China National Tobacco Company, Zhengzhou, China
| | | | - Binbin Lu
- Zhengzhou Tobacco Research Institute of China National Tobacco Company, Zhengzhou, China
| | - Guangfeng Hong
- Zhengzhou Tobacco Research Institute of China National Tobacco Company, Zhengzhou, China
| | - Wenjuan Zhang
- Zhengzhou Tobacco Research Institute of China National Tobacco Company, Zhengzhou, China
| | - Shu Tian
- Inner Mongolia Kunming Cigarette Limited Liability Company, Huhhot, China
| | - Jian Mao
- Flavour Science Research Center, College of Chemistry, Zhengzhou University, Zhengzhou, China
- Beijing Life Science Academy (BLSA), Beijing, China
- Zhengzhou Tobacco Research Institute of China National Tobacco Company, Zhengzhou, China
| | - Jianping Xie
- Flavour Science Research Center, College of Chemistry, Zhengzhou University, Zhengzhou, China
- Beijing Life Science Academy (BLSA), Beijing, China
| |
Collapse
|
19
|
Wang P, Chen Z, Li P, Al Mamun A, Ning S, Zhang J, Tang C, Sun T, Xiao J, Wei X, Wu F. Multi-targeted nanogel drug delivery system alleviates neuroinflammation and promotes spinal cord injury repair. Mater Today Bio 2025; 31:101518. [PMID: 39935893 PMCID: PMC11810842 DOI: 10.1016/j.mtbio.2025.101518] [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: 06/23/2024] [Revised: 12/26/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Spinal cord injury (SCI) is significantly hampered by an inflammatory microenvironment, prompting continued efforts in drug development to address inflammation. Research shows that quercetin (Que) exhibits excellent performance in reducing inflammation and neuroprotection. However, its application is limited by poor solubility, notable side effects, and the unique pathophysiology of the spinal cord. In this study, we introduce a novel multifunctional liposome hydrogel drug delivery system (QLipTC@HDM), obtained by incorporating liposomes with blood-spinal cord barrier penetration and injury site targeting properties (LipTC) into a dual-network viscous hydrogel (HDM). Our results demonstrate that encapsulating Que in LipTC (QLipTC) enhances solubility, minimizes toxic side effects, facilitates lesion targeting, and aids in crossing the blood-spinal cord barrier. Moreover, encapsulation in HDM significantly prolongs the retention of QLipTC at the injury site after local administration. Crucially, our findings reveal that QLipTC@HDM induces M2 phenotype transformation in glial cells and in mice with SCI, thereby mitigating inflammation. This intervention additionally preserves the integrity of the blood-spinal cord barrier, optimizes the spinal cord microenvironment, reduces glial scarring, promotes axonal regeneration, and enhances motor function recovery in SCI mice. In summary, our investigations highlight the potential of this disease-specific drug delivery system as a promising therapeutic approach for the treatment and management of SCI.
Collapse
Affiliation(s)
- Penghui Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Zaifeng Chen
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Ping Li
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Abdullah Al Mamun
- Central Laboratory of the Lishui Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Shaoxia Ning
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Jinjing Zhang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Chonghui Tang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Tianmiao Sun
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Jian Xiao
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
- Central Laboratory of the Lishui Hospital of Wenzhou Medical University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Xiaojie Wei
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| | - Fenzan Wu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
- Cixi Biomedical Research Institute of Wenzhou Medical University, Ningbo, Zhejiang, 315300, China
| |
Collapse
|
20
|
Kojima T, Asakura K, Gobbo P, Banno T. Programmed Fabrication of Vesicle-Based Prototissue Fibers with Modular Functionalities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409066. [PMID: 39927512 PMCID: PMC12021080 DOI: 10.1002/advs.202409066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/22/2024] [Indexed: 02/11/2025]
Abstract
Multicellular organisms have hierarchical structures where multiple cells collectively form tissues with complex 3D architectures and exhibit higher-order functions. Inspired by this, to date, multiple protocell models have been assembled to form tissue-like structures termed prototissues. Despite recent advances in this research area, the programmed assembly of protocells into prototissue fibers with emergent functions still represents a significant challenge. The possibility of assembling prototissue fibers will open up a way to a novel type of prototissue subunit capable of hierarchical assembly into unprecedented soft functional materials with tunable architectures, modular and distributed functionalities. Herein, the first method to fabricate freestanding vesicle-based prototissue fibers with controlled lengths and diameters is devised. Importantly, it is also shown that the fibers can be composed of different specialized modules that, for example, can endow the fiber with magnetotaxis capabilities, or that can work synergistically to take an input diffusible chemical signals and transduce it into a readable fluorescent output through a hosted enzyme cascade reaction. Overall, this research addresses an important challenge of prototissue engineering and will find important applications in 3D bio-printing, tissue engineering, and soft robotics as next-generation bioinspired materials.
Collapse
Affiliation(s)
- Tomoya Kojima
- Department of Applied ChemistryKeio University3‐14‐1 Hiyoshi, Kohoku‐kuYokohamaKanagawa223‐8522Japan
| | - Kouichi Asakura
- Department of Applied ChemistryKeio University3‐14‐1 Hiyoshi, Kohoku‐kuYokohamaKanagawa223‐8522Japan
| | - Pierangelo Gobbo
- Department of Chemical and Pharmaceutical SciencesUniversity of TriesteVia L. Giorgieri 1Trieste34127Italy
- National Interuniversity Consortium of Materials Science and Technology Unit of TriesteVia G. Giusti 9Firenze50121Italy
| | - Taisuke Banno
- Department of Applied ChemistryKeio University3‐14‐1 Hiyoshi, Kohoku‐kuYokohamaKanagawa223‐8522Japan
| |
Collapse
|
21
|
Shaikh S, Chary PS, Mehra NK. Tyrosine Kinase Inhibitor Lenvatinib Based Nano Formulations and Cutting-Edge Scale-Up Technologies in revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1749-1784. [PMID: 40091597 DOI: 10.1021/acsabm.4c01527] [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: 03/19/2025]
Abstract
Lenvatinib (LEN), a tyrosine kinase inhibitor, has emerged as a promising therapeutic agent for various solid tumors. Nevertheless, a number of constraints, including diminished bioavailability, incapacity to elicit localized inflammation, and inability to selectively accumulate at the tumor site, may impede the comprehensive exploitation of its versatile tyrosine kinase inhibitory capabilities. In order to achieve targeted delivery of LEN while also reducing its high dose used in conventional therapeutics, nanoformulation approaches can be adopted. The integration of LEN into various nanoformulations, such as nanoparticles, nanocrystals, high density lipoproteins (HDLs), liposomes, and micelles, is discussed, highlighting the advantages of these innovative approaches in a comparative manner; however, given that the current methods of nanoformulation synthesis employ toxic organic solvents and chemicals, there is an imperative need for exploring alternative, environmentally friendly approaches. The multifaceted effects of nanocarriers have rendered them profoundly applicable within the biomedical domain, serving as instrumental entities in various capacities such as vehicles for drug delivery and genetic material, diagnostic agents, facilitators of photothermal therapy, and radiotherapy. However, the scalability of these nanotechnological methodologies must be rigorously investigated and addressed to refine drug delivery mechanisms. This endeavor offers promising prospects for revolutionizing strategies in cancer therapeutics, thereby laying the foundation for future research in scale-up techniques in the pursuit of more effective and less toxic therapies for cancer.
Collapse
Affiliation(s)
- Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| |
Collapse
|
22
|
Xiao S, Rao L, Yan C, Nie L, Wang L, Zhao Y, Zhang S, Zhan W, Qin D, Zhuang M. Aptamer Functionalized Liposomes Co-Loaded with Exenatide-4 and Coenzyme Q10 Ameliorate Type 2 Diabetes Mellitus by Improving Pancreatic β Cell Function. Int J Nanomedicine 2025; 20:3363-3378. [PMID: 40125440 PMCID: PMC11928442 DOI: 10.2147/ijn.s510240] [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: 12/04/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Oxidative stress has been shown to disrupt β-cell function and promote the development of type 2 diabetes mellitus (T2DM). Exenatide-4 (Ext-4) is a widely used anti-glycemic drug but cannot restore pancreatic β-cells' structure and function. Coenzyme Q10 (CoQ10) has great antioxidant activities but shows suboptimal therapeutic effects because of its poor solubility and poor bioavailability. To further enhance the therapeutic efficacy of the drugs, a pancreas-targeting liposomal co-delivery system encapsulating Ext-4 and CoQ10 ((E+Q)-Lip-Apt) was designed, using the aptamers as the targeting ligands. Methods (E+Q)-Lip-Apt was prepared by thin film dispersion method and its optimal formulation was obtained through single-factor experiments and orthogonal experiments. The pancreatic β-cell protecting effect of (E+Q)-Lip-Apt was investigated both in vitro and in vivo. Results (E+Q)-Lip-Apt exhibited uniform size, good dispersion, and high encapsulation efficiency (EE) for both Ext-4 and CoQ10. The in vitro results showed that (E+Q)-Lip-Apt manifested superior capacity in scavenging ROS, enhancing mitochondrial membrane potential, and reducing malondialdehyde (MDA) content compared to Ext-4 in MIN6 cells. In vivo investigations demonstrated that (E+Q)-Lip-Apt significantly improved glucose tolerance, insulin sensitivity, hepatic lipid metabolism, oxidative stress, and enhanced antioxidant enzyme activity in diabetic mice. Moreover, Hematoxylin-eosin staining (H&E) and Immunohistochemistry (IHC) results indicated that (E+Q)-Lip-Apt could improve liver and pancreatic lesions, restoring the structure and function of β-cells in diabetic mice. Conclusion (E+Q)-Lip-Apt could improve oxidative stress, thereby restoring pancreatic β-cell function, and alleviating diabetes.
Collapse
Affiliation(s)
- Shangying Xiao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Lei Rao
- Medical College, Shaoguan University, Shaoguan, People’s Republic of China
| | - Canying Yan
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Ling Nie
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Leiqi Wang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Yingyin Zhao
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Shihao Zhang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - WeiMao Zhan
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Dongyun Qin
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| | - Manjiao Zhuang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Dongguan, People’s Republic of China
| |
Collapse
|
23
|
Nkune NW, Abrahamse H. The Combination of Active-Targeted Photodynamic Therapy and Photoactivated Chemotherapy for Enhanced Cancer Treatment. JOURNAL OF BIOPHOTONICS 2025:e70005. [PMID: 40083278 DOI: 10.1002/jbio.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
Scientists have been actively investigating novel therapies that can effectively eradicate cancer cells with negligible side effects in normal tissues when used alone or in a combinatorial approach. Photodynamic therapy has emerged as a promising non-invasive therapy that integrates photosensitizer, oxygen, and a specific wavelength of light for the treatment of cancer. Despite encouraging outcomes yielded by PDT, conventional PSs are faced with longstanding challenges such as poor water solubility, a short half-life, and off-target toxicity. Development of nanotherapeutics has shown great potential in overcoming this issue. The tumor microenvironment is inherently hypoxic, and this promotes tumor resistance to PDT, as it is oxygen-dependent. Photoactivated chemotherapy, an oxygen-independent light-based therapy, utilizes chemotherapeutic regimens that remain inert until exposed to light, allowing target-specific activation while minimizing off-target toxicity. Integration of these techniques can improve selectivity and yield synergistic cytotoxic effects that could improve cancer treatment.
Collapse
Affiliation(s)
- Nkune Williams Nkune
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, South Africa
| |
Collapse
|
24
|
Joseph-Mullol B, Royo M, Preat V, Moliné T, Ferrer B, Aparicio G, Cortés-Hernández J, Solé C. Topical miRNA Delivery via Elastic Liposomal Formulation: A Promising Genetic Therapy for Cutaneous Lupus Erythematosus (CLE). Int J Mol Sci 2025; 26:2641. [PMID: 40141283 PMCID: PMC11942213 DOI: 10.3390/ijms26062641] [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: 12/30/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Cutaneous lupus erythematosus (CLE) is a chronic autoimmune skin disorder with limited therapeutic options, particularly for refractory discoid lupus (DLE), which often results in scarring and atrophy. Recent studies have identified miR-31, miR-485-3p, and miR-885-5p as key regulators of inflammation, apoptosis, and fibrosis in CLE skin lesions. This research investigates a novel topical miRNA therapy using DDC642 elastic liposomes to target these pathways in CLE. DDC642 liposomes were complexed with miRNAs (anti-miR-31, anti-miR-485-3p, pre-miR-885-5p) and characterized through dynamic light scattering and Cryo-TEM. Cytotoxicity, cellular penetration, and therapeutic efficacy were evaluated in primary keratinocytes, PBMCs, and immune 3D-skin organoids. miRNA lipoplexes were successfully synthesized with optimized particle size, surface charge, and encapsulation efficiency. These lipoplexes exhibited effective cellular penetration and low cytotoxicity. Anti-miR-31 lipoplexes reduced miR-31 and NF-κB levels while increasing STK40 and PPP6C expression. Pre-miR-885-5p lipoplexes elevated miR-885-5p levels and downregulated PSMB5 and NF-κB in keratinocytes. While anti-miR-485-3p lipoplexes reduced T-cell activation markers. Anti-miR-31 and pre-miR-885-5p lipoplexes successfully modulated inflammatory pathways in 3D-skin CLE models. miRNA lipoplexes represent promising candidates for pioneering topical genetic therapies for CLE. Further studies, including animal models, are necessary to validate and optimize these findings.
Collapse
Affiliation(s)
- Blanca Joseph-Mullol
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (B.J.-M.); (M.R.)
| | - Maria Royo
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (B.J.-M.); (M.R.)
| | - Veronique Preat
- Louvain Drug Research Institute—Advanced Drug Delivery and Biomaterial, Universite Catholique de Louvain, 1200 Brussels, Belgium;
| | - Teresa Moliné
- Department of Pathology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (T.M.); (B.F.)
| | - Berta Ferrer
- Department of Pathology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (T.M.); (B.F.)
| | - Gloria Aparicio
- Department of Dermatology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Josefina Cortés-Hernández
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (B.J.-M.); (M.R.)
| | - Cristina Solé
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (B.J.-M.); (M.R.)
| |
Collapse
|
25
|
Ghanem R, Youf R, Haute T, Buin X, Riool M, Pourchez J, Montier T. The (re)emergence of aerosol delivery: Treatment of pulmonary diseases and its clinical challenges. J Control Release 2025; 379:421-439. [PMID: 39800241 DOI: 10.1016/j.jconrel.2025.01.017] [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: 10/04/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Aerosol delivery represents a rapid and non-invasive way to directly reach the lungs while escaping the hepatic first-pass effect. The development of pulmonary drugs for respiratory diseases such as cystic fibrosis, lung infections, pulmonary fibrosis or lung cancer requires an enhanced understanding of the relationships between the natural physiology of the respiratory system and the pathophysiology of these conditions. This knowledge is crucial to better predict and thereby control drug deposition. Moreover, aerosol administration faces several challenges, including the pulmonary tract, immune system, mucociliary clearance, the presence of fluid on the airway surfaces, and, in some cases, bacterial colonisation. Each of them directly influences on the bioavailability of the active molecule. In addition to these challenges, particle size and the device used to administer the treatment are critical factors that can significantly impact the biodistribution of the drugs. Nanoparticles are very promising in the development of new formulations for aerosol drug delivery, as they can be fine-tuned to reach the entire pulmonary tract and overcome the difficulties encountered along the way. However, to properly assess drug delivery, preclinical studies need to be more thorough to efficiently enhance drug delivery.
Collapse
Affiliation(s)
- Rosy Ghanem
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, F-29200 Brest, France
| | - Raphaëlle Youf
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Tanguy Haute
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Xavier Buin
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Martijn Riool
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Jérémie Pourchez
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F - 42023 Saint-Etienne, France
| | - Tristan Montier
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, F-29200 Brest, France.
| |
Collapse
|
26
|
Brenner AJ, Patel T, Bao A, Phillips WT, Michalek JE, Youssef M, Weinberg JS, Kamiya Matsuoka C, Hedrick MH, LaFrance N, Moore M, Floyd JR. Convection enhanced delivery of Rhenium ( 186Re) Obisbemeda ( 186RNL) in recurrent glioma: a multicenter, single arm, phase 1 clinical trial. Nat Commun 2025; 16:2079. [PMID: 40055350 PMCID: PMC11889265 DOI: 10.1038/s41467-025-57263-1] [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: 04/25/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Rhenium (186Re) Obisbemeda (186RNL), chelated-186Re encapsulated in nanoliposomes and delivered to brain tumors via convection enhanced delivery (CED), was evaluated in a Phase 1 dose escalation trial (NCT01906385). The primary objective was to determine the maximum tolerated dose (MTD). Secondary objectives included safety and tolerability, dose distribution, the overall response rate (ORR), disease-specific progression-free survival (PFS), and overall survival (OS). 21 patients received up to 22.3 mCi 186RNL over 6 dosing cohorts. Most adverse events (AEs) were unrelated to 186RNL and the MTD was not reached. Although not predefined outcomes, the mOS and mPFS were 11 and 4 months, respectively, and found to correlate with radiation absorbed dose to the tumor and percent tumor treated. When dichotomized by absorbed dose of 100 Gy, the mOS and mPFS were 17 months and 6 months, respectively, for >100 Gy, compared to 6 (mOS) and 2 (mPFS) months, respectively, for <100 Gy. For ORR, 57.1% exhibited stable disease (SD), 4.8% partial response, and 38.1% progressive disease. Overall, patients received radiation absorbed doses without significant toxicity higher than possible with external beam radiation therapy (EBRT) and demonstrated mOS beyond standard of care for recurrent glioblastoma (~8 months).
Collapse
Affiliation(s)
- Andrew J Brenner
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA.
| | - Toral Patel
- UT Southwestern Medical Center of Dallas, Dallas, TX, USA
| | - Ande Bao
- Case Western Reserve University, Cleveland, OH, USA
| | | | - Joel E Michalek
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | | - John R Floyd
- Mays Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
27
|
Wang J, Gu X, Gao X, Chen J, Lv Z, Zhang S, Ni S, Shi F, Chen X, Cao L, Wang Z, Xiao W. Formulation and optimization of glycyrrhetinic acid-modified pH-sensitive curcumin liposomes for anti-hepatocellular carcinoma. Pharm Dev Technol 2025; 30:233-245. [PMID: 39935270 DOI: 10.1080/10837450.2025.2465549] [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: 07/26/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
In order to enhance the therapeutic value of curcumin in liver cancer treatment, glycyrrhetinic acid-modified pH-sensitive curcumin liposomes (GA-pH-Lip@Cur) was developed.GA-pH-Lip@Cur was prepared using a thin film dispersion ultrasonication method, and the optimal formulation process was selected through single-factor experiments and a Box-Behnken design-response surface methodology. The liposomes were evaluated for their morphological appearance, particle size, in vitro release at different pH levels, and biocompatibility. The anti-tumor effect of GA-pH-Lip@Cur was assessed using cell viability assays (CCK-8). The in vivo hepatic targeting and anti-liver tumor efficacy of GA-pH-Lip@Cur were evaluated through pharmacokinetic and pharmacological experiments. The results indicated that optimized GA-pH-Lip@Cur exhibited uniform particle size distribution, good stability, pH-sensitive in vitro release with sustained behavior. Compared to conventional liposomes, GA-pH-Lip@Cur showed prolonged average retention time in vivo and significantly increased curcumin distribution in liver tissues, indicating excellent liver targeting. Both in vitro and in vivo evaluations demonstrated the effectiveness of GA-pH-Lip@Cur in inhibiting liver cancer cell proliferation and suppressing liver tumor growth in tumor-bearing mice. In conclusion, GA-pH-Lip@Cur, by leveraging the acidic tumor microenvironment and overexpression of glycyrrhetinic acid receptors in liver cells, encapsulates curcumin to improve its bioavailability, and target its delivery to the liver tumor sites.
Collapse
Affiliation(s)
- Jie Wang
- Nanjing University of Chinese Medicine, Nanjing, China
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Xuemei Gu
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Xia Gao
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Jing Chen
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Zhiyang Lv
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyu Zhang
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Siyu Ni
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Fei Shi
- Hanlin College of Nanjing University of Chinese Medicine, Taizhou, China
| | - Xialin Chen
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Liang Cao
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Zhenzhong Wang
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| | - Wei Xiao
- Nanjing University of Chinese Medicine, Nanjing, China
- National Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co., Ltd, Lianyungang, China
| |
Collapse
|
28
|
Hammond J, Richards CJ, Ko Y, Jonker T, Åberg C, Roos WH, Lira RB. Membrane Fusion-Based Drug Delivery Liposomes Transiently Modify the Material Properties of Synthetic and Biological Membranes. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408039. [PMID: 40007088 PMCID: PMC11947515 DOI: 10.1002/smll.202408039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Many drug targets are located in intracellular compartments of cells but they often remain inaccessible to standard imaging and therapeutic agents. To aid intracellular delivery, drug carrier nanoparticles have been used to overcome the barrier imposed by the plasma membrane. The carrier must entrap large amounts of cargo, efficiently and quickly deliver the cargo in the cytosol or other intracellular compartments, and must be inert; they should not induce cellular responses or alter the cell state in the course of delivery. This study demonstrates that cationic liposomes with high charge density efficiently fuse with synthetic membranes and the plasma membrane of living cells. Direct fusion efficiently delivers large amounts of cargo to cells and cell-like vesicles within seconds, bypassing slow and often inefficient internalization-based pathways. These effects depend on liposome charge density, concentration, and the helper lipid. However, fusion-mediated cargo delivery results in the incorporation of large amounts of foreign lipids, causing changes to the material properties of these membranes, namely modifications in membrane packing and fluidity, induction of membrane curvature, decrease in surface tension, and the formation of (short-lived) pores. Importantly, these effects are transient and liposome removal allows cells to recover their state prior to liposome interaction.
Collapse
Affiliation(s)
- Jayna Hammond
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Ceri J. Richards
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyRijksuniversiteit GroningenGroningenThe Netherlands
| | - YouBeen Ko
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Thijs Jonker
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Christoffer Åberg
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyRijksuniversiteit GroningenGroningenThe Netherlands
| | - Wouter H. Roos
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
| | - Rafael B. Lira
- Moleculaire BiofysicaZernike InstituutRijksuniversiteit GroningenGroningenThe Netherlands
- Present address:
Department of BionanoscienceKavli Institute of NanoscienceDelft University of TechnologyDelftThe Netherlands
| |
Collapse
|
29
|
Li S, Li N, Yin Q, Zhang Z, Hu H, Hao L. cRGD-based MRI imaging-enhanced nanoplatform helps DOX target pancreatic cancer. Sci Rep 2025; 15:7217. [PMID: 40021813 PMCID: PMC11871140 DOI: 10.1038/s41598-025-91549-0] [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: 11/11/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025] Open
Abstract
This project aims to construct cRGD functionalized mesoporous silica nanoparticles and cRGD modified mesoporous silica nanoparticles for the diagnosis and treatment of tumors, providing new ideas for targeted therapy of tumors. The mesoporous silica nanoparticles were doped with gadolinium in situ to provide excellent imaging; cRGD was coupled on the particle surface to confer particle targeting; and hyaluronic acid was loaded onto the particles by electrostatic adsorption, thereby improving the biocompatibility of the particles and prolonging their in vivo circulation time.Taking pancreatic cancer as a model, we studied its targeting ability to pancreatic cancer and its phagocytosis to cancer cells; Using methods such as cell growth experiments and flow cytometry, the anti-cancer effect and pro apoptotic effect of the system were studied. In vivo distribution, tumor targeting and therapeutic efficacy of nanoparticles evaluated in a mouse model of pancreatic cancer with loaded tumors.Evaluate the bioavailability and enrichment of nanoparticles in tumor tissue using MRI technology. Evaluate the therapeutic effect and safety through changes in tumor volume, histopathological examination, and prognosis. Characterization of the synthesis results proved that cRGD-HA-DOX-Gd2O3@MSN (cHDG@MSN) was successfully synthesized with a particle size of 230.83 ± 12.36 nm.In vitro drug release experiments of DOX were carried out at different pH values (5.5 and 7.4), where the release was only up to 22.65% at pH 7.4, whereas DOX release was increased up to 78.75% at pH = 5.5.The results confirm the pH responsiveness of this nanocarrier platform.The results of cytotoxicity studies showed that cHDG@MSN itself is not cytotoxic. Biosafety evaluation and hemolysis test results confirmed that the probe is highly biocompatible.Notably, Gd3+ significantly enhanced the T1 contrast of the system according to MR imaging results.The apoptosis rates of SW1990 cells treated with PBS, DOX and cHDG@MSN in flow cytometry were 13.97%, 18.38% and 29.02%, respectively, demonstrating the effectiveness of the nanoprobes at the cellular level. Animal experiments demonstrated the effectiveness of nanoprobes at the pathological level and imaging level.Cells and animals demonstrated that cHDG@MSN effectively inhibited the proliferation of pancreatic cancer cells. This research further verified the pH sensitivity of the constructed compound drug delivery system to achieve accurate diagnosis and treatment of pancreatic cancer tumor cells.
Collapse
Affiliation(s)
- Silong Li
- Medical Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang Province, 161006, People's Republic of China
| | - Na Li
- Department of Imaging Medicine and Nuclear Medicine, School of Clinical Medicine, Jiamusi University, Jiamusi, 154002, People's Republic of China
| | - Qiangqiang Yin
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People's Republic of China
| | - Zhichen Zhang
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People's Republic of China
| | - Haifeng Hu
- Medical Imaging Center, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, 161000, People's Republic of China
| | - Liguo Hao
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People's Republic of China.
| |
Collapse
|
30
|
Filipiuc SI, Simionescu N, Stanciu GD, Coroaba A, Marangoci NL, Filipiuc LE, Pinteala M, Uritu CM, Tamba BI. Fluorescent Rhein-Loaded Liposomes for In Vivo Biodistribution Study. Pharmaceutics 2025; 17:307. [PMID: 40142971 PMCID: PMC11944368 DOI: 10.3390/pharmaceutics17030307] [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: 02/13/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Objectives: This work aimed to develop and investigate liposomes incorporating Rhein (Lip-Rh) into the liposomal membrane to enhance the compound's water solubility and oral bioavailability. Methods: Liposomes were produced by the thin lipid film technique, with a phosphatidylcholine-to-cholesterol molar ratio of 5:1, dissolved in chloroform and methanol, and thereafter hydrated with ultrapure water and subjected to sonication. The resultant liposomes were studied from a physicochemical perspective using DLS, zeta potential, STEM, UV-Vis, and fluorescence spectroscopies, while oral bioavailability was assessed by fluorescence imaging. Additionally, cell viability assays were performed on tumour cells (MCF-7) in comparison to normal cells (HGFs). Results: The resultant nanoparticles exhibited relatively uniform sizes and narrow size distribution. In vivo fluorescence imaging studies performed on Wistar rats demonstrated significantly enhanced oral bioavailability for Lip-Rh, with rapid absorption into the bloodstream observed one hour after administration, in contrast to the free compound dissolved in vegetable oil. Cell viability assays demonstrated higher cytotoxicity of Lip-Rh towards MCF-7 cells compared to HGF cells, highlighting the selective therapeutic potential of the product. Moreover, we determined that the optimal dose of Rhein per kilogram of body weight, when encapsulated in liposomes, is approximately 2.5 times less than when Rhein is delivered in its unencapsulated form. Conclusions: Lip-Rh is a promising candidate for oncological treatments, presenting three key advantages: increased cytotoxicity towards tumour cells, protection of normal tissues, and the practicality of oral delivery. Additional investigation is required to explore its application in anticancer therapy, whether as monotherapy or as a complementary treatment.
Collapse
Affiliation(s)
- Silviu Iulian Filipiuc
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania; (S.I.F.); (N.S.); (A.C.); (N.L.M.); (M.P.)
- Advanced Center for Research and Development in Experimental Medicine “Prof. Ostin C. Mungiu”, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (L.E.F.); (B.I.T.)
| | - Natalia Simionescu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania; (S.I.F.); (N.S.); (A.C.); (N.L.M.); (M.P.)
| | - Gabriela Dumitrița Stanciu
- Advanced Center for Research and Development in Experimental Medicine “Prof. Ostin C. Mungiu”, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (L.E.F.); (B.I.T.)
| | - Adina Coroaba
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania; (S.I.F.); (N.S.); (A.C.); (N.L.M.); (M.P.)
| | - Narcisa Laura Marangoci
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania; (S.I.F.); (N.S.); (A.C.); (N.L.M.); (M.P.)
| | - Leontina Elena Filipiuc
- Advanced Center for Research and Development in Experimental Medicine “Prof. Ostin C. Mungiu”, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (L.E.F.); (B.I.T.)
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania; (S.I.F.); (N.S.); (A.C.); (N.L.M.); (M.P.)
| | - Cristina Mariana Uritu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487 Iasi, Romania; (S.I.F.); (N.S.); (A.C.); (N.L.M.); (M.P.)
- Advanced Center for Research and Development in Experimental Medicine “Prof. Ostin C. Mungiu”, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (L.E.F.); (B.I.T.)
| | - Bogdan Ionel Tamba
- Advanced Center for Research and Development in Experimental Medicine “Prof. Ostin C. Mungiu”, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (G.D.S.); (L.E.F.); (B.I.T.)
| |
Collapse
|
31
|
Liu J, Wang T, Dong J, Lu Y. The blood-brain barriers: novel nanocarriers for central nervous system diseases. J Nanobiotechnology 2025; 23:146. [PMID: 40011926 PMCID: PMC11866817 DOI: 10.1186/s12951-025-03247-8] [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: 11/16/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
The central nervous system (CNS) diseases are major contributors to death and disability worldwide. However, the blood-brain barrier (BBB) often prevents drugs intended for CNS diseases from effectively crossing into the brain parenchyma to deliver their therapeutic effects. The blood-brain barrier is a semi-permeable barrier with high selectivity. The BBB primarily manages the transport of substances between the blood and the CNS. To enhance drug delivery for CNS disease treatment, various brain-based drug delivery strategies overcoming the BBB have been developed. Among them, nanoparticles (NPs) have been emphasized due to their multiple excellent properties. This review starts with an overview of the BBB's anatomical structure and physiological roles, and then explores the mechanisms, both endogenous and exogenous, that facilitate the NP passage across the BBB. The text also delves into how nanoparticles' shape, charge, size, and surface ligands affect their ability to cross the BBB and offers an overview of different nanoparticle classifications. This review concludes with an examination of the current challenges in utilizing nanomaterials for brain drug delivery and discusses corresponding directions for solutions. This review aims to propose innovative diagnostic and therapeutic approaches for CNS diseases and enhance drug design for more effective delivery across the BBB.
Collapse
Affiliation(s)
- Jiajun Liu
- State Key Laboratory of Green Biomanufacturing, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ting Wang
- State Key Laboratory of Green Biomanufacturing, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Jian Dong
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yuan Lu
- State Key Laboratory of Green Biomanufacturing, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
32
|
Pandey P, Arya DK, Kumar A, Kaushik A, Mishra YK, Rajinikanth PS. Dual ligand functionalized pH-sensitive liposomes for metastatic breast cancer treatment: in vitro and in vivo assessment. J Mater Chem B 2025; 13:2682-2694. [PMID: 39841132 DOI: 10.1039/d4tb02570a] [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: 01/23/2025]
Abstract
This research demonstrates the design and development of a novel dual-targeting, pH-sensitive liposomal (pSL) formulation of 5-Fluorouracil (5-FU), i.e., (5-FU-iRGD-FA-pSL) to manage breast cancer (BC). The motivation to explore this formulation is to overcome the challenges of systemic toxicity and non-specific targeting of 5-FU, a conventional chemotherapeutic agent. The proposed formulation also combines folic acid (FA) and iRGD peptides as targeting ligands to enhance tumor cell specificity and penetration, while the pH-sensitive liposomes ensure the controlled drug release in the acidic tumor microenvironment. The physicochemical characterization revealed that 5-FU-iRGD-FA-pSL possesses optimal size, low polydispersity index, and favorable zeta potential, enhancing its stability and targeting capabilities. In vitro studies demonstrated significantly enhanced cellular uptake, cytotoxicity, and inhibition of cell migration in MCF-7 BC cells compared to free 5-FU and non-targeted liposomal formulations. DAPI staining revealed significant apoptotic features, including chromatin condensation (CC) and nuclear fragmentation (NF), with 5-FU-iRGD-FA-pSL inducing more pronounced apoptosis compared to 5-FU-pSL. Furthermore, in vivo analysis in a BC rat model showed superior anti-tumor efficacy, reduced systemic toxicity, and improved safety profile of the 5-FU-iRGD-FA-pSL formulation. This dual-targeting pSL system presents a promising approach for enhancing the therapeutic index of 5-FU, offering a potential strategy for more effective BC treatment.
Collapse
Affiliation(s)
- Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Dilip Kumar Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| | - Anit Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland 33805, FL, USA
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Alsion 2, 6400 Sønderborg, Denmark
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
33
|
Torres J, Valenzuela Oses JK, Rabasco-Álvarez AM, González-Rodríguez ML, García MC. Innovations in Cancer Therapy: Endogenous Stimuli-Responsive Liposomes as Advanced Nanocarriers. Pharmaceutics 2025; 17:245. [PMID: 40006612 PMCID: PMC11858840 DOI: 10.3390/pharmaceutics17020245] [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: 12/21/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Recent advancements in nanotechnology have revolutionized cancer therapy-one of the most pressing global health challenges and a leading cause of death-through the development of liposomes (L), lipid-based nanovesicles known for their biocompatibility and ability to encapsulate both hydrophilic and lipophilic drugs. More recent innovations have led to the creation of stimuli-responsive L that release their payloads in response to specific endogenous or exogenous triggers. Dual- and multi-responsive L, which react to multiple stimuli, offer even greater precision, improving therapeutic outcomes while reducing systemic toxicity. Additionally, these smart L can adjust their physicochemical properties and morphology to enable site-specific targeting and controlled drug release, enhancing treatment efficacy while minimizing adverse effects. This review explores the latest advancements in endogenous stimuli-responsive liposomal nanocarriers, as well as dual- and multi-responsive L that integrate internal and external triggers, with a focus on their design strategies, mechanisms, and applications in cancer therapy.
Collapse
Affiliation(s)
- Jazmín Torres
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| | - Johanna Karina Valenzuela Oses
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| | - Antonio María Rabasco-Álvarez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain;
| | - María Luisa González-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain;
| | - Mónica Cristina García
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| |
Collapse
|
34
|
Eftaiha AF, Suryabrahmam B, Morris NB, Qaroush AK, Assaf KI, Foudeh DM, Hammad SB, Ashkar R. Modification of Liposomal Properties by an Engineered Gemini Surfactant. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3042-3052. [PMID: 39862190 PMCID: PMC11823631 DOI: 10.1021/acs.langmuir.4c03043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/22/2024] [Accepted: 12/16/2024] [Indexed: 01/27/2025]
Abstract
Lipid membranes form the primary structure of cell membranes and serve as configurable interfaces across numerous applications including biosensing technologies, antifungal treatments, and therapeutic platforms. Therefore, the modification of lipid membranes by additives has important consequences in both biological processes and practical applications. In this study, we investigated a nicotinic-acid-based gemini surfactant (NAGS) as a chemically tunable molecular additive for modulating the structure and phase behavior of liposomal membranes. We specifically focused on NAGS with hydrocarbon chains that mirror those of lipid molecules. By introducing NAGS to phosphatidylcholine membranes with lipids of identical and varied chain lengths or degrees of unsaturation, we demonstrated the effects of headgroup interactions and chain mismatch between NAGS and membrane lipids. Using small-angle X-ray scattering, we showed that regardless of chain compatibility or mismatch, NAGS reduced the thickness and packing density of fluid lipid membranes. Further observations by fluorescence microscopy revealed the emergence of ordered-disordered domains upon cooling to room temperature. The observed phases were quite distinct from those of lipid membranes with analogous chain compositions, emphasizing the importance of NAGS headgroup chemistry in mediating domain formation and stabilization. These findings open new possibilities for exploiting NAGS in tuning the structure and organization of liposomal membranes with potential applications in drug delivery and biomedical imaging.
Collapse
Affiliation(s)
- Ala’a F. Eftaiha
- Department
of Chemistry, Faculty of Science, The Hashemite
University, Zarqa 13133, Jordan
- Department
of Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
- Center
for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Buti Suryabrahmam
- Department
of Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
- Center
for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Nicholas B. Morris
- Department
of Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
- Center
for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Abdussalam K. Qaroush
- Department
of Chemistry, Faculty of Science, The University
of Jordan, Amman 11942, Jordan
| | - Khaleel I. Assaf
- Department
of Chemistry, Faculty of Science, Al-Balqa
Applied University, Al-Salt 19117, Jordan
| | - Dina M. Foudeh
- Department
of Chemistry, Faculty of Science, The University
of Jordan, Amman 11942, Jordan
| | - Suhad B. Hammad
- Department
of Chemistry, Faculty of Science, The University
of Jordan, Amman 11942, Jordan
| | - Rana Ashkar
- Department
of Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
- Center
for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, Virginia 24061, United States
- Macromolecular
Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
35
|
Fan X, Cheng D, Niu B, Wang X, Zhang P. Current research status, applications and challenges of ketorolac-based sustained-release and controlled-release formulations. Int J Pharm 2025; 670:125162. [PMID: 39793634 DOI: 10.1016/j.ijpharm.2024.125162] [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: 10/02/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025]
Abstract
Ketorolac, a nonsteroidal anti-inflammatory drug, exhibits moderate antipyretic and anti-inflammatory properties, as well as potent analgesic effects. It is widely used in clinical practice for pain relief in cases of mild and severe pain such as postoperative pain, fractures, sprains, toothaches and cancer pain. Due to its relatively short half-life, patients experiencing pain often need frequent injections or oral medications, leading to poor patient compliance. Thus, it is crucial to create long-acting sustained-release formulations of ketorolac. This paper provides an overview of the research, applications, and challenges associated with ketorolac sustained-release formulations over the past decade, based on a comprehensive review of the literature. The aim is to provide fresh insights for the research and development of long-acting, sustained-release, and controlled-release formulations of ketorolac.
Collapse
Affiliation(s)
- Xiaoling Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China; Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, Shandong, PR China
| | - Dongfang Cheng
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, Shandong, PR China.
| | - Baohua Niu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, Shandong, PR China
| | - Xiuzhi Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, PR China
| | - Peng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China.
| |
Collapse
|
36
|
Zhao R, Lan D, Xia B, Dong M, Mu J, Zhao Y. PET-Based Dual-Modal Probes for In Vivo Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409713. [PMID: 39873346 DOI: 10.1002/smll.202409713] [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: 10/19/2024] [Revised: 11/07/2024] [Indexed: 01/30/2025]
Abstract
Molecular imaging has significantly advanced the detection and analysis of in vivo metabolic processes, while single-modal techniques remain limited. Dual-modal imaging, particularly positron emission tomography (PET)-based combinations has emerged as a powerful solution, offering enhanced capabilities through integration with magnetic resonance imaging (MRI) or near-infrared fluorescence (NIRF) imaging. This review highlights recent progress in PET-based dual-modal imaging, focusing on the development of various bimodal probes derived from antibodies, nanoparticles, and peptides, and key applications including image-guided surgery and disease assessment. PET-based dual-modal imaging holds substantial potential for advancing research and diagnostics by improving resolution and providing functional insights. By combining complementary modalities, these systems deliver a more comprehensive view of disease processes, leading to more accurate diagnoses and targeted treatments. Future research prioritizes optimizing probe design for enhanced biocompatibility and safety, facilitating clinical translation, and broadens applications beyond cancer. Through interdisciplinary collaboration, PET-based dual-modal probes are poised to play a pivotal role in improving patient outcomes, particularly in diagnosing and managing complex diseases.
Collapse
Affiliation(s)
- Runge Zhao
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Deren Lan
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Beibei Xia
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - MengJie Dong
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Jing Mu
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yongsheng Zhao
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| |
Collapse
|
37
|
Darwish A, Sándor N, Szenti I, Marosvölgyi T, Juhász K, Rónavári A, Kachal E, Kutus B, Kónya Z, Balogi Z. Highly Stable Antitumor Silver-Lipid Nanoparticles Optimized for Targeted Therapy. Int J Nanomedicine 2025; 20:1351-1366. [PMID: 39911260 PMCID: PMC11796454 DOI: 10.2147/ijn.s498208] [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: 09/27/2024] [Accepted: 12/21/2024] [Indexed: 02/07/2025] Open
Abstract
Background Silver nanoparticles (AgNPs) have a broad spectrum of biocidal effects, allowing also their antitumor application. To enhance bioavailability, minimize adverse effects and enable targeted drug delivery AgNPs may be encapsulated in liposomes. In this study we aimed to create highly stable and effective antitumor AgNP lipid formulations (LAgs). Methods Uncapped and citrate-stabilized AgNPs were encapsulated by the lipid film hydration method using several phospholipid mixtures, followed by the essential removal of unencapsulated AgNPs by size exclusion chromatography (SEC). Purified LAgs were characterized by UV-VIS, DLS, XRD, ICP-MS, transmission electron microscopy (TEM) and glycerol-based density gradient centrifugation (DGC). Liposomal stability was assessed by carboxyfluorescein (CF) leakage, while antitumor effects of purified LAgs were tested in MTT, clonogenic and 3D spheroid invasion experiments. Results The presence of AgNPs inside SEC-purified liposomes was confirmed by TEM, XRD and ICP-MS. Encapsulation efficiency was estimated to be between 18.7 and 25.5%. Purified LAgs had higher density as compared to free AgNPs revealed by DGC, indicating that a considerable fraction of liposomes contained AgNPs. LAgs with PC/PG, PC/PG/SM/Chol, and in particular PC/PG/SM displayed the highest stability assessed by CF leakage, whereas high content of neutral or negatively charged phospholipids was destabilizing. As shown by MTT and colony formation assays, viability and survival of A375 and RPMI-7951 melanoma cells were severely impaired by LAgs at a higher or comparable level as caused by free AgNPs. Used as a non-tumor control, HEK293 cells were less vulnerable to LAgs as compared to free AgNPs. Finally, applying the most stable lipid composition, PC/PG/SM-LAg-c, and in part PC/PG/SM-LAg-u effectively inhibited a tissue-like invasion of melanoma spheroids. Conclusion Altogether, highly stable purified LAg formulations were created, which effectively block survival, clonogenic potential and invasion of melanoma cells, therefore could be promising NP platforms for targeted tumor therapy.
Collapse
Affiliation(s)
- Ammar Darwish
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, H-7624, Hungary
| | - Nikolett Sándor
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, H-7624, Hungary
| | - Imre Szenti
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, H-6720, Hungary
| | - Tamás Marosvölgyi
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, H-7624, Hungary
| | - Kata Juhász
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, H-7624, Hungary
| | - Andrea Rónavári
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, H-6720, Hungary
| | - Edi Kachal
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, H-6720, Hungary
| | - Bence Kutus
- Department of Molecular and Analytical Chemistry, University of Szeged, Szeged, H-6720, Hungary
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, H-6720, Hungary
| | - Zsolt Balogi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, H-7624, Hungary
| |
Collapse
|
38
|
Li H, Xu D, Cai W, Liu J, Bing Z, Zhang Q. PEGylated Nanoliposomal Doxorubicin Conjugated with Specific TREM2 Peptides for Glioma-Targeting Therapy. Adv Healthc Mater 2025; 14:e2403096. [PMID: 39711286 DOI: 10.1002/adhm.202403096] [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: 08/17/2024] [Revised: 12/12/2024] [Indexed: 12/24/2024]
Abstract
PEGylated liposomes can deliver anti-cancer drugs to brain tumors, and achieve enhanced permeability and retention effects. Triggering receptor expressed on myeloid cells 2 (TREM2) is an excellent biomarker for precise therapy of glioma. The present study is aimed at designing PEGylated nanoliposomal doxorubicin (PLD) conjugated with peptides targeting TREM2 for glioma-targeting therapy. The specific peptides are designed with the Rosetta Peptiderive Protocol. Schrodinger's peptide-specific version of Glide is used for molecular docking. PLD modified with peptides (peptide-PLD) are engineered and prepared. Cell cycle, apoptosis, cell invasion and migration, cell viability, and colony-formation assays are performed to analyze glioma cell functions. The anti-tumor effects of peptide-PLD are validated in an intracranial U87-MG cells orthotopic glioma model. The targeting peptides HLRKLRKR and LRKLRLRL showed specific affinity for TREM2 and better cellular uptake in U87-MG cells. PLD with peptide modification demonstrated stable doxorubicin loading, small sizes (<60 nm), and enrichment in the mouse brain. Peptide-PLD treatment inhibited the Akt/GSK3β/β-catenin pathway, thereby inhibiting cell invasion and migration, and colony-forming ability in U87-MG cells. The peptide modification of PLD achieved better suppression of glioma development than PLD. Overall, TREM2-targeting peptides are successfully designed, and peptide-PLD served as a potent drug delivery carrier for glioma-targeting therapy.
Collapse
Affiliation(s)
- Hongyan Li
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Gansu Provincial Isotope Laboratory, Lanzhou, 730300, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Duling Xu
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Weihua Cai
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiadi Liu
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Zhitong Bing
- Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Qiyue Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000, China
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
39
|
Vanić Ž, Jøraholmen MW, Škalko-Basnet N. Challenges and considerations in liposomal hydrogels for the treatment of infection. Expert Opin Drug Deliv 2025; 22:255-276. [PMID: 39797393 DOI: 10.1080/17425247.2025.2451620] [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: 10/30/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
INTRODUCTION Liposomal hydrogels are novel drug delivery systems that comprise preformed liposomes incorporated in hydrogels destined for mostly localized drug therapy, herewith antimicrobial therapy. The formulation benefits from versatility of liposomes as lipid-based nanocarriers that enable delivery of various antimicrobials of different lipophilicities, and secondary vehicle, hydrogel, that assures better retention time of formulation at the infection site. Especially in an era of alarming antimicrobial resistance, efficient localized antimicrobial therapy that avoids systemic exposure of antimicrobial and related side effects is crucial. AREAS COVERED We provide an overview of liposomal hydrogels that were developed for superior delivery of antimicrobials at different infections sites, with focus on skin and vaginal infections. The review summarizes the challenges of infection site and most common infection-causing pathogens and offers commentary on most relevant features the formulation needs to optimize to increase the therapy outcome. We discuss the impact of liposomal composition, size, and choice of polymer-forming hydrogel on antimicrobial outcome based on the literature overview and own experience in the field. EXPERT OPINION Liposomal hydrogels offer improved therapy outcome in localized antimicrobial therapy. By fine-tuning of liposomal as well as hydrogel properties, formulations with superior performance can be optimized targeting specific infection site.
Collapse
Affiliation(s)
- Željka Vanić
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - May Wenche Jøraholmen
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Tromsø, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
40
|
Yılmaz E, Kacaroglu D, Ozden AK, Aydogan N. Gold nanoparticles decorated FOLFIRINOX loaded liposomes for synergistic therapy of pancreatic cancer. Int J Pharm 2025; 669:125067. [PMID: 39672312 DOI: 10.1016/j.ijpharm.2024.125067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Pancreatic cancer is predicted to be the second highest cause of cancer deaths by 2030, with a mortality rate of 98 % and a 5-year survival rate of only 4-8 %. FOLFIRINOX which consists of four main ingredients has shown superior efficacy in treating patients with pancreatic cancer compared to other agents and combinations. However, toxicities have prevented full-dose use of FOLFIRINOX. In this study, we present the design of a liposome nanosystem that enables the sequential release of a drug combination that is called FOLFIRINOX using lipid-based nanosystem synergistic chemo/photothermal therapy approaches. The co-eccentric liposome allowed us to locate the drug molecules in different locations giving us the flexibility to release them in a selected order. Core liposome (L2) has a melting temperature of 53.63 °C, it was decorated by gold nanoparticle (L2@AuNP) to bring photothermal responsiveness. The outer liposome structure had a lower melting temperature, which facilitated the sequential release process. The efficacy of photothermal therapy for nanosystem was calculated. The results indicate that coating L2@AuNP nanostructure with L1 liposomes improves efficacy by stabilizing gold nanoparticles. FOLFIRINOX components are encapsulated in a concentric liposome structure according to the order of administration into the body. The concentric liposome structure enables the sequential release of multiple drugs due to the varying phase transition temperatures of the liposomes. The cytotoxic effect of these formulations was evaluated on Panc-1 pancreatic cancer cells; the lowest cell viability was obtained in 4 Liposome(L) under 5 min NIR irradiation. Combination therapy has a higher therapeutic efficacy (70.45 %) when compared to chemotherapy and photothermal therapy used separately. The study's results show the potential of combination therapies to improve therapeutic outcomes, providing a promising path for future research and clinical application.
Collapse
Affiliation(s)
- Emine Yılmaz
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey
| | - Demet Kacaroglu
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Ayse Kevser Ozden
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Nihal Aydogan
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Beytepe, Ankara, Turkey; Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara, 06800, Turkey.
| |
Collapse
|
41
|
Ding Y, Li B, Yi Y, Liu C, Wen J, Jian X, Li Y. Progress in the role of nanoparticles in the diagnosis and treatment of bone and joint tuberculosis. Front Med (Lausanne) 2025; 12:1536547. [PMID: 39926423 PMCID: PMC11804262 DOI: 10.3389/fmed.2025.1536547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Bone and joint tuberculosis (BJTB), caused by Mycobacterium tuberculosis (MTB), is a prevalent form of extrapulmonary tuberculosis that poses significant challenges to global public health due to difficulties in early diagnosis, prolonged treatment cycles, and drug resistance. Recent advancements in nanotechnology have introduced novel solutions for the early detection and precise treatment of BJTB, leveraging unique physicochemical properties such as high specific surface area, targeted delivery capabilities, sustained drug release, and excellent biocompatibility. In diagnostic applications, nanomaterials markedly enhance the sensitivity and accuracy of detection methods while reducing testing time. These technologies are adaptable to resource-limited settings, enabling earlier patient intervention and mitigating disease progression risk. In therapeutic applications, nanomaterials prolong drug retention in bone tissue through targeted delivery, thereby decreasing medication frequency and minimizing toxic side effects, which significantly improves treatment efficacy. Despite substantial progress, further research is required to address long-term safety concerns, broaden clinical applicability, and evaluate performance under complex pathological conditions. This review summarizes recent advancements in nanomaterials for diagnosing and treating BJTB and identifies key areas for future research, laying the groundwork for advancing precision medicine and personalized treatments.
Collapse
Affiliation(s)
- Yitong Ding
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Baiyun Li
- Department of Nursing, Hunan Normal University, Changsha, Hunan, China
| | - Yangfei Yi
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Can Liu
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Jie Wen
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
- Department of Pediatric Orthopedics, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Xiaohong Jian
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yufei Li
- Department of Anatomy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| |
Collapse
|
42
|
Zhao W, Li X, Guan J, Yan S, Teng L, Sun X, Dong Y, Wang H, Tao W. Potential and development of cellular vesicle vaccines in cancer immunotherapy. Discov Oncol 2025; 16:48. [PMID: 39812959 PMCID: PMC11735706 DOI: 10.1007/s12672-025-01781-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Cancer vaccines are promising as an effective means of stimulating the immune system to clear tumors as well as to establish immune surveillance. In this paper, we discuss the main platforms and current status of cancer vaccines and propose a new cancer vaccine platform, the cytosolic vesicle vaccine. This vaccine has a unique structure that can integrate antigen and adjuvant carriers to improve the delivery efficiency and immune activation ability, which brings new ideas for cancer vaccine design. Tumor exosomes carry antigens and MHC-peptide complexes, which can provide tumor antigens to antigen-processing cells and increase the chances of recognition of tumor antigens by immune cells. DEVs play a role in amplifying the immune response by acting as carriers for the dissemination of antigenic substances in dendritic cells. OMVs, with their natural adjuvant properties, are one of the advantages for the preparation of antitumor vaccines. This paper presents the advantages of these three bacteria/extracellular vesicles as cancer vaccines and discusses the potential applications of functionally modified extracellular vesicles as cancer vaccines after cellular engineering or genetic engineering, as well as current clinical trials of extracellular vesicle vaccines. In summary, extracellular vesicle vaccines are a promising direction for cancer vaccine research.
Collapse
Affiliation(s)
- Wenxi Zhao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Xianjun Li
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Jialu Guan
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Shuai Yan
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Lizhi Teng
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Xitong Sun
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Yuhan Dong
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Hongyue Wang
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Weiyang Tao
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, No. 23, Youzheng Street, Nangang District, Harbin, 150001, China.
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
43
|
Tie S, Xue M, Chen Y, Wu Y, Gu S, Tan M. Enhancing stability, bioavailability and nutritional intervention effect of procyanidins using bio-based delivery systems: A review. Int J Biol Macromol 2025; 287:138517. [PMID: 39647725 DOI: 10.1016/j.ijbiomac.2024.138517] [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: 06/25/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Procyanidins (PCs), a kind of polyphenolic compound, have attracted extensive attention due to their strong antioxidant, anti-inflammatory and other activities. However, PCs are susceptible to complex micro-environments, resulting in low stability, poor target tissue delivery and bioavailability, which limits their biological effects. Therefore, it is urgent to find some suitable pathways to protect PCs, avoid their degradation, and maximize their health benefits in nutritional intervention. This review focused on the design and construction of different types of bio-based delivery systems loaded with PCs, such as nanoparticles, microparticles, emulsions, liposomes, hydrogels and fibers. The advantages and biological effects of PCs-based delivery systems in promoting cellular uptake, realizing targeted release of organs, cells and organelles, and even nutritional intervention for different chronic diseases were summarized. Furthermore, the development prospects and challenges of delivery systems in the field of precision nutrition were discussed. The construction of these delivery systems can effectively improve the stability and bioavailability of PCs, and maximize their precise nutritional intervention for various chronic diseases.
Collapse
Affiliation(s)
- Shanshan Tie
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China
| | - Mengmeng Xue
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
| | - Yannan Chen
- College of Food Science and Engineering, Shandong Agricultural University, Taian 270018, China
| | - Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, China.
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China.
| |
Collapse
|
44
|
Bisht A, Avinash D, Sahu KK, Patel P, Das Gupta G, Kurmi BD. A comprehensive review on doxorubicin: mechanisms, toxicity, clinical trials, combination therapies and nanoformulations in breast cancer. Drug Deliv Transl Res 2025; 15:102-133. [PMID: 38884850 DOI: 10.1007/s13346-024-01648-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Doxorubicin is a key treatment for breast cancer, but its effectiveness often comes with significant side effects. Its actions include DNA intercalation, topoisomerase II inhibition, and reactive oxygen species generation, leading to DNA damage and cell death. However, it can also cause heart problems and low blood cell counts. Current trials aim to improve doxorubicin therapy by adjusting doses, using different administration methods, and combining it with targeted treatments or immunotherapy. Nanoformulations show promise in enhancing doxorubicin's effectiveness by improving drug delivery, reducing side effects, and overcoming drug resistance. This review summarizes recent progress and difficulties in using doxorubicin for breast cancer, highlighting its mechanisms, side effects, ongoing trials, and the potential impact of nanoformulations. Understanding these different aspects is crucial in optimizing doxorubicin's use and improving outcomes for breast cancer patients. This review examines the toxicity of doxorubicin, a drug used in breast cancer treatment, and discusses strategies to mitigate adverse effects, such as cardioprotective agents and liposomal formulations. It also discusses clinical trials evaluating doxorubicin-based regimens, the evolving landscape of combination therapies, and the potential of nanoformulations to optimize delivery and reduce systemic toxicity. The review also discusses the potential of liposomes, nanoparticles, and polymeric micelles to enhance drug accumulation within tumor tissues while sparing healthy organs.
Collapse
Affiliation(s)
- Anjali Bisht
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Dubey Avinash
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Chaumuhan, Mathura, 281406, UP, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
45
|
Yang Z, Shi X, Qiu L. Tunable supramolecular self-assemblies based on cyclodextrin polymer as a loading platform for water-soluble drugs. Carbohydr Polym 2025; 347:122743. [PMID: 39486972 DOI: 10.1016/j.carbpol.2024.122743] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 11/04/2024]
Abstract
Drug loading capacity is a crucial character of nano-scaled drug carriers to achieve high quality pharmaceutical preparations. However, efficient encapsulation of water-soluble small molecular drugs still faces large obstacles in many cases. Herein, we designed a novel supramolecular delivery system constructed by poly(β-cyclodextrin) containing benzoic acid groups (PCD-PA) and adamantyl terminated poly(ethylene glycol) (PEG-AD) to provide multiple intermolecular interactions for competent loading of water-soluble small-molecular drugs. PCD-PA had multiple host molecules, and PEG-AD could be inserted via host-guest interaction in different proportion to adjust the composition of supramolecular carrier. Meanwhile, π-π stacking and electrostatic interaction furnished by benzoic acid groups served as binding force for drug entrapment, which led to considerable loading capacity for several water-soluble drugs. Among the drugs with different chemical structures, mitoxantrone hydrochloride and doxorubicin hydrochloride bearing anthraquinone rings and several protonable amino groups acquired the highest loading content as about 14 % in PCD-PA3/PEG-AD supramolecular self-assemblies. Further computational simulations investigated the mechanism of drug loading based on the interactions between the carrier materials and the payloads. In addition, the weakly acidic environment obviously accelerated the release of certain drugs. All in all, this self-assembled supramolecular nano-system displayed great potentials as a delivery platform for diverse water-soluble drugs.
Collapse
Affiliation(s)
- Zhuting Yang
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Xuezhang Shi
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Liyan Qiu
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
46
|
Pimple P, Shah J, Singh P. Emerging Phytochemical Formulations for Management of Rheumatoid Arthritis: A Review. Curr Drug Deliv 2025; 22:15-40. [PMID: 38299275 DOI: 10.2174/0115672018270434240105110330] [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: 07/11/2023] [Revised: 12/02/2023] [Accepted: 12/17/2023] [Indexed: 02/02/2024]
Abstract
Rheumatoid arthritis (RA) is a T-cell-mediated chronic inflammatory disorder affecting 0.5-1% of the global population. The disease with unknown etiology causes slow destruction of joints, advancing to significant deterioration of an individual's quality of life. The present treatment strategy comprises the use of disease-modifying anti-rheumatic drugs (DMARDs) coupled with or without nonsteroidal anti-inflammatory drugs or glucocorticoids. Additionally, involves co-therapy of injectable biological DMARDs in case of persistent or recurrent arthritis. The availability of biological DMARDs and the implementation of the treat-to-target approach have significantly improved the outcomes for patients suffering from RA. Nevertheless, RA requires continuous attention due to inadequate response of patients, development of tolerance and severe side effects associated with long-term use of available treatment regimens. An estimated 60-90% of patients use alternative methods of treatment, such as herbal therapies, for the management of RA symptoms. Over the past few decades, researchers have exploring natural phytochemicals to alleviate RA and associated symptoms. Enormous plant-origin phytochemicals such as alkaloids, flavonoids, steroids, terpenoids and polyphenols have shown anti-inflammatory and immunomodulatory activity against RA. However, phytochemicals have certain limitations, such as high molecular weight, poor water solubility, poor permeability, poor stability and extensive first-pass metabolism, limiting absorption and bioavailability. The use of nanotechnology has aided to extensively improve the pharmacokinetic profile and stability of encapsulated drugs. The current review provides detailed information on the therapeutic potential of phytochemicals. Furthermore, the review focuses on developed phytochemical formulations for RA, with emphasis on clinical trials, regulatory aspects, present challenges, and future prospects.
Collapse
Affiliation(s)
- Prachi Pimple
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| | - Jenny Shah
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| | - Prabha Singh
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| |
Collapse
|
47
|
Wang X, Cao Z, Su J, Ge X, Zhou Z. Oral barriers to food-derived active peptides and nano-delivery strategies. J Food Sci 2025; 90:e17672. [PMID: 39828408 DOI: 10.1111/1750-3841.17672] [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: 10/20/2024] [Revised: 12/04/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
Food-derived bioactive peptides are a class of peptides from natural protein. It may have biological effects on the human body and play a significant role in protecting human physiological health and regulating physiological metabolism, such as lowering blood pressure, lowering cholesterol, antioxidant, antibacterial, regulating immune activity, and so on. However, most of the natural food-derived functional peptides need to overcome a variety of barriers in the body to enter the blood circulation system and target to specific tissues to generate physiological activity. During this process, the bioavailability of the functional peptides will be reduced. The nano-delivery system can offer the feasibility to overcome these obstacles and improve the stability and bioavailability of food-derived active peptides by nanoencapsulation. This work summarizes the application of food-derived bioactive peptides and the obstacles during the delivery pathway in vivo. Moreover, the different nano-delivery systems used for bioactive peptides and their application were summarized, which could provide ideas for oral delivery of food-derived bioactive peptides.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Zhaoxin Cao
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Jingyi Su
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
| | - Xuemei Ge
- Department of Food Science and Technology, College of Light Industry Science and Engineering, Nanjing Forestry University, Nanjing, P. R. China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Zhiyong Zhou
- College of Medicine and Health Sciences, China Three Gorges University, Yichang, P. R. China
| |
Collapse
|
48
|
Mo L, Yang C, Dai Y, Liu W, Gong Y, Guo Y, Zhu Y, Cao Y, Xiao X, Du S, Lu S, He J. Novel drug delivery systems for hirudin-based product development and clinical applications. Int J Biol Macromol 2025; 287:138533. [PMID: 39657884 DOI: 10.1016/j.ijbiomac.2024.138533] [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: 09/23/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Hirudin, a natural biological polypeptide macromolecule secreted by the salivary glands of medicinal leech, is a specific thrombin inhibitor with multiple favourable bioactivities, including anti-coagulation, anti-fibrotic, and anti-tumour. Despite several anticoagulants have been widely applied in clinic, hirudin shows advantages in reducing the incidence of bleeding side effects by virtue of its high specificity in binding to thrombin. As a result, hirudin has been tested in clinical practice to prevent and treat several complex diseases. However, the application of this polypeptide macromolecule is compromised by its low bioavailability and bioactivity due to poor serum stability and susceptibility to protease degradation in vivo. To overcome these drawbacks, several studies have proposed novel drug delivery systems (NDDSs) to prevent the degradation and increase the targeting efficiency of hirudin. This systematic review summarises the clinical research on hirudin, including its classification and bioactivities, and highlights the opportunities and challenges in the clinical use of hirudin. The NDDSs designed to enhance the bioavailability and bioactivity of hirudin are discussed to explore its application in the treatment of related diseases. This review may considerably contribute to the advancement of delivery science and technology, particularly in the context of polypeptide-based therapeutics.
Collapse
Affiliation(s)
- Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Wei Liu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuhong Gong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430061, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yan Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430061, PR China
| | - Xuecheng Xiao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430061, PR China
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430061, PR China.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430061, PR China.
| |
Collapse
|
49
|
Agrawal SS, Baliga V, Londhe VY. Liposomal Formulations: A Recent Update. Pharmaceutics 2024; 17:36. [PMID: 39861685 PMCID: PMC11769406 DOI: 10.3390/pharmaceutics17010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 01/27/2025] Open
Abstract
Liposome-based drug delivery technologies have showed potential in enhancing medication safety and efficacy. Innovative drug loading and release mechanisms highlighted in this review of next-generation liposomal formulations. Due to poor drug release kinetics and loading capacity, conventional liposomes have limited clinical use. Scientists have developed new liposomal carrier medication release control and encapsulation methods to address these limits. Drug encapsulation can be optimized by creating lipid compositions that match a drug's charge and hydrophobicity. By selecting lipids and adding co-solvents or surfactants, scientists have increased drug loading in liposomal formulations while maintaining stability. Nanotechnology has also created multifunctional liposomes with triggered release and personalized drug delivery. Surface modification methods like PEGylation and ligand conjugation can direct liposomes to disease regions, improving therapeutic efficacy and reducing off-target effects. In addition to drug loading, researchers have focused on spatiotemporal modulation of liposomal carrier medication release. Stimuli-responsive liposomes release drugs in response to bodily signals. Liposomes can be pH- or temperature-sensitive. To improve therapeutic efficacy and reduce systemic toxicity, researchers added stimuli-responsive components to liposomal membranes to precisely control drug release kinetics. Advanced drug delivery technologies like magnetic targeting and ultrasound. Pro Drug, RNA Liposomes approach may improve liposomal medication administration. Magnetic targeting helps liposomes aggregate at illness sites and improves drug delivery, whereas ultrasound-mediated drug release facilitates on-demand release of encapsulated medicines. This review also covers recent preclinical and clinical research showing the therapeutic promise of next-generation liposomal formulations for cancer, infectious diseases, neurological disorders and inflammatory disorders. The transfer of these innovative liposomal formulations from lab to clinical practice involves key difficulties such scalability, manufacturing difficulty, and regulatory limits.
Collapse
Affiliation(s)
- Surendra S. Agrawal
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DU), Sawangi (M), Wardha 442001, Maharashtra, India;
| | - Vrinda Baliga
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| | - Vaishali Y. Londhe
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| |
Collapse
|
50
|
Peng S, Zhang Y, Zhao X, Wang Y, Zhang Z, Zhang X, Li J, Zheng H, Zhang Y, Shi H, Li H, Liu L. Pathologic Tissue Injury and Inflammation in Mice Immunized with Plasmid DNA-Encapsulated DOTAP-Based Lipid Nanoparticles. Bioconjug Chem 2024; 35:2015-2026. [PMID: 39656061 DOI: 10.1021/acs.bioconjchem.4c00536] [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: 12/19/2024]
Abstract
Ionizable cationic lipids have been developed to mitigate the toxicity of quaternary ammonium lipids, such as DOTAP. Despite its toxicity, DOTAP can promote localization of lipid nanoparticles (LNPs) in target tissues, serving as one of the ionizable cationic helper lipids. Notably, DOTAP-based nanoadjuvants prepared via microfluidic methods showed a better T-cell response. Previous studies showed that DOTAP-based LNPs prepared by the lipid-film method resulted in obvious adverse events. Therefore, our research focused on evaluating the tissue localization and adverse toxicity of a DOTAP-based delivery system prepared through microfluidic techniques. We assessed the delivery efficacy, biodistribution, inflammatory response, and pathological injury in various tissues. In our study, the plasmid DNA encoding the receptor-binding domain (RBD) of SARS-CoV-2 was encapsulated using a mixture of lipids that included DOTAP, DOPE, cholesterol, and DMG-PEG2000 via microfluidic mixing. The LNP-RBDs were smaller than those prepared via the traditional lipid membrane system. We found that LNP-DNA complexes can be effectively delivered and expressed in muscle tissue, with specific antibodies in serum induced postimmunization. Initial distribution of the liposomes was observed in the muscle and liver. Interestingly, both LNPs and DNA showed sustained presence in the lungs and spleen in the group immunized with DNA-encapsulated DOTAP-based LNPs, whereas lower amounts of DNA were detected in the group immunized with dissociative DNA. We detected obvious inflammatory responses and pathological injuries in the muscle, heart, and liver, and the side effects decreased when the immunization dose decreased. These findings suggest that DOTAP-based LNPs have obvious advantages for targeting the lungs and spleen. Additionally, inflammatory responses and pathological injuries occur in a dose-dependent manner in the muscles, heart, and liver. In conclusion, these findings contribute to the development of an LNP delivery system with DOTAP, highlighting its potential to enhance tissue localization and promote high levels of expression when coordinated with ionizable lipids.
Collapse
Affiliation(s)
- Shasha Peng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Yifan Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| | - Xin Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Yibin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Zihan Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Xin Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jiali Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Huiwen Zheng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Haijing Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Heng Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Kunming, Yunnan 650118, China
| |
Collapse
|