1
|
Ruan S, Du Y, Zhang X, Zhang X, Kang H. Ameliorative Hypoglycemic Effect of 1-DNJ via Structural Derivatization Followed by Assembly Into Selenized Nanovesicles. Int J Nanomedicine 2025; 20:4399-4413. [PMID: 40230538 PMCID: PMC11995926 DOI: 10.2147/ijn.s516833] [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: 01/10/2025] [Accepted: 04/06/2025] [Indexed: 04/16/2025] Open
Abstract
Purpose 1-Deoxynojirimycin (1-DNJ), a phytomedicine derived from mulberry leaves and certain bacteria, can inhibit α-glycosidase activity and alleviate insulin resistance, thereby lowering blood glucose levels. However, its short half-life and limited in vivo residence compromise its therapeutic efficacy. This study aimed to optimize the structure of 1-DNJ and develop nano-formulation to ameliorate its pharmacokinetic properties and therapeutic effects. Methods We synthesized N-oleoyl-1-DNJ (N-1-DNJ) and formulated it into selenized nanovesicles using a thin-film hydration method combined with in situ reduction. Results The resulting N-1-DNJ-loaded selenized nanovesicles (N-1-DNJ-Se@NVs) exhibited improved physiological stability and sustained release compared to non-selenized versions. In vivo pharmacokinetic studies in GK rats revealed that N-1-DNJ-Se@NVs presented prolonged absorption, higher mean retention time, and enhanced area under the blood drug concentration versus time curve (AUC), indicating superior bioavailability. Furthermore, N-1-DNJ-Se@NVs demonstrated long-lasting hypoglycemic effect and increased cellular uptake efficiency. Conclusion Our findings suggest that structural derivatization improves the oral delivery of 1-DNJ and prolongs its therapeutic effect via selenized nanovesicles, positioning N-1-DNJ-Se@NVs as a promising nanomedicine for diabetes management.
Collapse
Affiliation(s)
- Shuxian Ruan
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
| | - Yanli Du
- The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xianyuan Zhang
- Meizhou Baoyuantang Pharmaceutical Co., Ltd, Meizhou, People’s Republic of China
| | - Xingwang Zhang
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
| | - Hao Kang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, Anhui College of Traditional Chinese Medicine, Wuhu, People’s Republic of China
| |
Collapse
|
2
|
Li W, Tan CH, Baek JS, Jiang L, Ng NKJ, Chong KKL, Wong JJ, Gao L, Kline KA, Loo SCJ. Anti-Intracellular MRSA Activity of Antibiotic-Loaded Lipid-Polymer Hybrid Nanoparticles and Their Effectiveness in Murine Skin Wound Infection Models. ACS Infect Dis 2025; 11:750-761. [PMID: 39949070 DOI: 10.1021/acsinfecdis.4c01016] [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: 03/15/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a significant concern for skin and soft tissue infections. Apart from biofilm formation, these bacteria can reside intracellularly in phagocytic and nonphagocytic mammalian cells, complicating treatment with conventional antibiotics. Lipid-polymer hybrid nanoparticle (LPN) systems, combining the advantages of polymeric nanoparticles and liposomes, represent a new generation of nanocarriers with the potential to address these therapeutic challenges. In this study, gentamicin (Gen) and vancomycin (Van) were encapsulated in LPNs and evaluated for their ability to eliminate intracellular MRSA in phagocytic macrophage RAW-Blue cells and nonphagocytic epithelial HaCaT cells. Compared to free antibiotics at 100 μg/mL, LPN formulations significantly reduced intracellular bacterial loads in both cell lines. Specifically, LPN-Van resulted in approximately 0.7 Log CFU/well reduction in RAW-Blue cells and 0.3 Log CFU/well reduction in HaCaT cells. LPN-Gen showed a more pronounced reduction, with approximately 1.26 Log CFU/well reduction in RAW-Blue cells and 0.45 Log CFU/well reduction in HaCaT cells. In vivo, LPN-Van at 500 μg/mL significantly reduced MRSA biofilm viability compared to untreated controls (p < 0.001), achieving 98% eradication based on median values. In comparison, free vancomycin achieved a nonstatistically significant 79.2% reduction in biofilm viability compared to control. Prophylactically, LPN-Van at 500 μg/mL decreased MRSA levels to the limit of detection, resulting in a ∼3.5 Log reduction in the median CFU/wound compared to free vancomycin. No acute dermal toxicity was observed for LPN-Van based on histological analysis. These data indicate that LPNs show promise as a drug delivery platform technology to address intracellular infections.
Collapse
Affiliation(s)
- Wenrui Li
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- NTU Institute for Health Technologies, Interdisciplinary Graduate Program, Nanyang Technological University, 61 Nanyang Drive, Singapore 637335, Singapore
| | - Chuan Hao Tan
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jong-Suep Baek
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Lai Jiang
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Noele Kai Jing Ng
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Kelvin Kian Long Chong
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jun Jie Wong
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Liheng Gao
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Kimberly A Kline
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Say Chye Joachim Loo
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Singapore Center for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore
| |
Collapse
|
3
|
Mitrović D, Zaklan D, Đanić M, Stanimirov B, Stankov K, Al-Salami H, Pavlović N. The Pharmaceutical and Pharmacological Potential Applications of Bilosomes as Nanocarriers for Drug Delivery. Molecules 2025; 30:1181. [PMID: 40076403 PMCID: PMC11901966 DOI: 10.3390/molecules30051181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Nano-drug delivery systems provide targeted solutions for addressing various drug delivery challenges, leveraging nanotechnology to enhance drug solubility and permeability. Liposomes, explored for several decades, face hurdles, especially in oral delivery. Bile-acid stabilized vesicles (bilosomes) are flexible lipid vesicles, composed of phospholipids or other surfactants, along with amphiphilic bile salts, and they show superior stability and pharmacokinetic behavior in comparison to conventional vesicular systems (liposomes and niosomes). Bilosomes enhance skin penetration, fluidize the stratum corneum, and improve drug stability. In oral applications, bilosomes overcome drawbacks, offering improved bioavailability, controlled release, and reduced side effects. Vaccines using bilosomes demonstrate efficacy, and bilosomes for intranasal, inhalation, ocular, and buccal applications enhance drug delivery, offering targeted, efficient, and controlled activities. Formulations vary based on active substances and optimization techniques, showcasing the versatility and potential of bilosomes across diverse drug delivery routes. Therefore, the aim of this comprehensive review was to critically explore the state-of-the-art of bilosomes in drug delivery and potential therapeutic applications.
Collapse
Affiliation(s)
- Darko Mitrović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (D.M.); (D.Z.)
| | - Dragana Zaklan
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (D.M.); (D.Z.)
| | - Maja Đanić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia;
| | - Bojan Stanimirov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (B.S.); (K.S.)
| | - Karmen Stankov
- Department of Biochemistry, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (B.S.); (K.S.)
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6845, Australia;
- UWA Medical School, University of Western Australia, Perth, WA 6009, Australia
| | - Nebojša Pavlović
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (D.M.); (D.Z.)
| |
Collapse
|
4
|
Dong X, Lin Y, Li K, Liang G, Huang X, Pan J, Wang L, Zhang D, Liu T, Wang T, Yan X, Zhang L, Li X, Qu X, Jia D, Li Y, Zhang H. Consensus statement on extracellular vesicles in liquid biopsy for advancing laboratory medicine. Clin Chem Lab Med 2025; 63:465-482. [PMID: 38896030 DOI: 10.1515/cclm-2024-0188] [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: 02/07/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024]
Abstract
Extracellular vesicles (EVs) represent a diverse class of nanoscale membrane vesicles actively released by cells. These EVs can be further subdivided into categories like exosomes and microvesicles, based on their origins, sizes, and physical attributes. Significantly, disease-derived EVs have been detected in virtually all types of body fluids, providing a comprehensive molecular profile of their cellular origins. As a result, EVs are emerging as a valuable addition to liquid biopsy techniques. In this collective statement, the authors share their current perspectives on EV-related research and product development, with a shared commitment to translating this newfound knowledge into clinical applications for cancer and other diseases, particularly as disease biomarkers. The consensus within this document revolves around the overarching recognition of the merits, unresolved questions, and existing challenges surrounding EVs. This consensus manuscript is a collaborative effort led by the Committee of Exosomes, Society of Tumor Markers, Chinese anti-Cancer Association, aimed at expediting the cultivation of robust scientific and clinically applicable breakthroughs and propelling the field forward with greater swiftness and efficacy.
Collapse
Affiliation(s)
- Xingli Dong
- 558113 Central Laboratory, Department of Hematology and Oncology, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen Clinical Research Center for hematologic disease, Shenzhen University General Hospital , Shenzhen, Guangdong, China
| | - Yusheng Lin
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Thoracic Surgery, 47885 The First Affiliated Hospital of Jinan University , Guangzhou, China
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Kai Li
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Gaofeng Liang
- 74623 School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology , Luoyang, China
| | - Xiaoyi Huang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Heilongjiang Province, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Tingjiao Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Tong Wang
- 47885 MOE Key Laboratory of Tumor Molecular Biology, College of Life Science and Technology, Jinan University , Guangzhou, China
| | - Xiaomei Yan
- Department of Chemical Biology, 534787 MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University , Xiamen, China
| | - Long Zhang
- 12377 MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University , Hangzhou, China
| | - Xiaowu Li
- Department of Hepatobiliary Surgery, 558113 Shenzhen Key Laboratory, Shenzhen University General Hospital , Shenzhen, Guangdong, China
| | - Xiujuan Qu
- Department of Medical Oncology, 159407 The First Hospital of China Medical University , Shenyang, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Hao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
- Institute of Precision Cancer Medicine and Pathology, and Department of Pathology, School of Medicine, Jinan University, Guangzhou, P.R. China
| |
Collapse
|
5
|
Qi C, Xing H, Ding N, Feng W, Wu Y, Zhang X, Yu Y. Nanometerizing Taxifolin Into Selenized Liposomes to Ameliorate Its Hypoglycemic Effect by Optimizing Drug Release and Bioavailability. Int J Nanomedicine 2025; 20:2225-2240. [PMID: 40007903 PMCID: PMC11853828 DOI: 10.2147/ijn.s510378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose Diabetes mellitus (DM) remains a significant health challenge, with traditional treatments often failing to provide lasting solutions. Taxifolin (Tax), a potential phytomedicine with antioxidant and anti-hyperglycemic properties, suffers from low water solubility and poor bioavailability, necessitating advanced delivery systems. This study aims to nanometerize taxifolin (Tax) into selenized liposomes (Tax-Se@LPs) for enhanced oral delivery and hypoglycemic effect. Methods Tax-Se@LPs were fabricated through a thin-film hydration/in situ reduction technique. The resulting nanomedicine was characterized through in vitro release studies, pharmacokinetic and pharmacodynamic evaluations, cellular uptake assays, and formulation stability tests. Results The optimized Tax-Se@LPs demonstrated an average particle size of 185.3 nm and an entrapment efficiency of 95.25% after optimization. In vitro release studies revealed that Tax-Se@LPs exhibited a slower and more sustained release profile compared to conventional liposomes, favoring gastrointestinal drug absorption. Pharmacokinetic evaluations in normal rats indicated that Tax-Se@LPs achieved a relative bioavailability of 216.65%, significantly higher than Tax suspensions and unmodified liposomes. Furthermore, in diabetic GK rats, Tax-Se@LPs resulted in a maximal blood glucose reduction of 46.8% and exhibited a more sustained therapeutic duration compared to other formulations. Cellular uptake tests manifested that selenization altered the internalization mechanisms of liposomes while preserving their absorption aptness by intestinal epithelial cells. The physiological and in vitro stability of Tax-Se@LPs was also reinforced by selenization. Conclusion Overall, Tax-Se@LPs not only improve the oral bioavailability of Tax but also enhance its therapeutic efficacy. These findings underscore the potential of Tax-Se@LPs as a promising therapeutic strategy for DM management.
Collapse
Affiliation(s)
- Chunli Qi
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, People’s Republic of China
- Institute of Laboratory Animals, Jinan University, Guangzhou, 510632, People’s Republic of China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, People’s Republic of China
| | - Huijie Xing
- Institute of Laboratory Animals, Jinan University, Guangzhou, 510632, People’s Republic of China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, People’s Republic of China
| | - Ning Ding
- Department of Animal, Plant and Food, Guangzhou Customs Technology Center, Guangzhou, 510623, People’s Republic of China
| | - Weifeng Feng
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, People’s Republic of China
| | - Yongyi Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People’s Republic of China
| | - Xingwang Zhang
- Department of Pharmaceutics, School of Pharmacy, Jinan University, Guangzhou, 511443, People’s Republic of China
| | - Yigang Yu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510641, People’s Republic of China
| |
Collapse
|
6
|
Yang H, Song Y, Zhang Q, Wang M, Jia T, Pan Q, Sun K, Guan X, Pan M, Chen F, Yan B. Engineering Protein-Based Lipid-Binding Nanovesicles via Catechol-Amine-Derived Coacervation with Their Underlying Interfacial Mechanisms. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3199-3208. [PMID: 39893691 DOI: 10.1021/acs.langmuir.4c03941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The development of nonphospholipid nanovesicles has garnered tremendous attention as a viable alternative to traditional liposomal nanovesicles. Protein/peptide-based nanovesicles have demonstrated their potential to reduce immunogenicity while enhancing bioactivity. However, a fundamental understanding of how proteinaceous vesicles interact with lipids and cell membranes remains elusive. In this study, we engineered a series of protamine-based nonphospholipid nanovesicles by modulating intramolecular catechol-amine interactions. By grafting trihydroxybenzene (GA) and catechol (CA) groups onto the protamine (Prot), a salt-triggered coacervation was observed in an alkaline environment with the size of as-prepared vesicles ranging from 200 to 1200 nm. The bonding affinity to lipid interfaces followed the order of Prot-CA-Fe3+(25 μM) > Prot-CA-Fe3+(10 μM) > Prot-CA > original Prot with the underlying nanomechanics investigated by the lipid bubble force measurement. Direct quantification of interactions between the nanovesicles and living human gingival fibroblasts was performed by using surface charge difference mapping. Introducing trace amounts of Fe3+ (at 10 and 25 μM) enhanced vesicle-lipid interactions via the synergy of catechol-amine interactions and Fe3+-induced complexation. This work provides improved valuable insights into the interactions between nanovesicles and cell membranes, offering an energetic paradigm for modulating cell-target delivery processes via intramolecular short-range interactions.
Collapse
Affiliation(s)
- Haibing Yang
- Department of Stomatology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Yao Song
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Zhang
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Moran Wang
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Tianqi Jia
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Qing Pan
- Department of Stomatology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Kanda Sun
- Department of Stomatology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
| | - Xiang Guan
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Mingfei Pan
- Department of Stomatology, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou 213164, China
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Feng Chen
- Key Laboratory for Bio-Electromagnetic Environment and Advanced Medical Theranostics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, China
| | - Bin Yan
- National Engineering Laboratory for Clean Technology of Leather Manufacture, College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
7
|
Yeo S, Wu H, Song YK, Yoon I, Lee WK. Encapsulation of synthesized purpurin-18-N-aminoimide methyl ester in lipid nanovesicles for use as agents in photodynamic cancer therapy. J Pharm Sci 2025; 114:1504-1511. [PMID: 39832621 DOI: 10.1016/j.xphs.2025.01.001] [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: 11/13/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
This study aimed to synthesize purpurin-18-N-aminoimide methyl ester (P18 N AI ME) and encapsulate it into lipid nanovesicles (LNVs) for potential application as photodynamic therapy (PDT) agents in cancer therapy. PDT, a light-induced treatment, offers several advantages over conventional cancer treatments, such as minimal invasiveness and localized action. P18 N AI ME, a chlorine class photosensitizer model drug, was synthesized in an attempt to treat tumor in deeper tissues by interacting long-wavelength light. LNVs were introduced to improve anticancer effect and photostability of P18 N AI ME. LNVs using glycerol monostearate demonstrated smaller particle sizes and more sustained release profiles than those using lauric acid. In photocytotoxicity against HeLa (human cervical carcinoma) and A549 (human lung carcinoma) cell lines, P18 N AI ME-LNVs demonstrated safety under dark conditions and enhanced anticancer effects under light conditions compared to P18 N AI ME alone. The inhibitory concentration values (IC50) were 0.86 μM (P18 N AI ME) and 0.68 μM (LNVs) in HeLa cell line and 0.85 μM (P18 N AI ME) and 0.64 μM (LNVs) in A549 cell line. These findings suggest that P18 N AI ME-LNVs hold promise as PDT agents in cancer therapy.
Collapse
Affiliation(s)
- Sooho Yeo
- College of Pharmacy & Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea; Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea.
| | - Huiqiang Wu
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea
| | - Young Kyu Song
- Research Center of Dr. i&B Co., DaeJeon, Republic of Korea
| | - Il Yoon
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea.
| | - Woo Kyoung Lee
- Center for Nano Manufacturing and Department of Nanoscience and Engineering, Inje University, Gimhae 50834, Republic of Korea.
| |
Collapse
|
8
|
Ravi Mythili VM, Rajendran RL, Arun R, Thasma Loganathbabu VK, Reyaz D, Nagarajan AK, Ahn BC, Gangadaran P. Emerging Strategies for Revascularization: Use of Cell-Derived Extracellular Vesicles and Artificial Nanovesicles in Critical Limb Ischemia. Bioengineering (Basel) 2025; 12:92. [PMID: 39851366 PMCID: PMC11762151 DOI: 10.3390/bioengineering12010092] [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: 11/27/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Critical limb ischemia (CLI) poses a substantial and intricate challenge in vascular medicine, necessitating the development of innovative therapeutic strategies to address its multifaceted pathophysiology. Conventional revascularization approaches often fail to adequately address the complexity of CLI, necessitating the identification of alternative methodologies. This review explores uncharted territory beyond traditional therapies, focusing on the potential of two distinct yet interrelated entities: cell-derived extracellular vesicles (EVs) and artificial nanovesicles. Cell-derived EVs are small membranous structures naturally released by cells, and artificial nanovesicles are artificially engineered nanosized vesicles. Both these vesicles represent promising avenues for therapeutic intervention. They act as carriers of bioactive cargo, including proteins, nucleic acids, and lipids, that can modulate intricate cellular responses associated with ischemic tissue repair and angiogenesis. This review also assesses the evolving landscape of CLI revascularization through the unique perspective of cell-derived EVs and artificial nanovesicles. The review spans the spectrum from early preclinical investigations to the latest translational advancements, providing a comprehensive overview of the current state of research in this emerging field. These groundbreaking vesicle therapies hold immense potential for revolutionizing CLI treatment paradigms.
Collapse
Affiliation(s)
- Vijay Murali Ravi Mythili
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Raksa Arun
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Vasanth Kanth Thasma Loganathbabu
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Danyal Reyaz
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - ArulJothi Kandasamy Nagarajan
- Integrative Genetics and Molecular Oncology Group, Department of Genetic Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu 603203, Tamil Nadu, India; (V.M.R.M.); (R.A.); (V.K.T.L.); (D.R.); (A.K.N.)
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
9
|
Mahmoud TM, Abdelfatah MM, Omar MM, Hasan OA, Wali SM, El-Mofty MS, Ewees MG, Salem AE, Abd-El-Galil TI, Mahmoud DM. Enhancing the Therapeutic Effect and Bioavailability of Irradiated Silver Nanoparticle-Capped Chitosan-Coated Rosuvastatin Calcium Nanovesicles for the Treatment of Liver Cancer. Pharmaceutics 2025; 17:72. [PMID: 39861720 PMCID: PMC11769262 DOI: 10.3390/pharmaceutics17010072] [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: 11/18/2024] [Revised: 12/14/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Liver cancer is a prevalent form of carcinoma worldwide. A novel chitosan-coated optimized formulation capped with irradiated silver nanoparticles (INops) was fabricated to boost the anti-malignant impact of rosuvastatin calcium (RC). Using a 23-factorial design, eight formulations were produced using the solvent evaporation process. The formulations were characterized in vitro to identify the optimal formulation (Nop). The FTIR spectra showed that the fingerprint region is not superimposed with that of the drug; DSC thermal analysis depicted a negligible peak shift; and XRPD diffractograms revealed the disappearance of the typical drug peaks. Nop had an entrapment efficiency percent (EE%) of 86.2%, a polydispersity index (PDI) of 0.254, a zeta potential (ZP) of -35.3 mV, and a drug release after 12 h (Q12) of 55.6%. The chitosan-coated optimized formulation (CS.Nop) showed significant mucoadhesive strength that was 1.7-fold greater than Nop. Physical stability analysis of CS.Nop revealed negligible alterations in VS, ZP, PDI, and drug retention (DR) at 4 °C. The irradiated chitosan-coated optimized formulation capped with silver nanoparticles (INops) revealed the highest inhibition effect on carcinoma cells (97.12%) compared to the chitosan-coated optimized formulation (CS.Nop; 81.64) and chitosan-coated optimized formulation capped with silver nanoparticles (CS.Nop.AgNPs; 92.41). The bioavailability of CS-Nop was 4.95-fold greater than RC, with a residence time of about twice the free drug. CS.Nop has displayed a strong in vitro-in vivo correlation with R2 0.9887. The authors could propose that novel INop could serve as an advanced platform to improve oral bioavailability and enhance hepatic carcinoma recovery.
Collapse
Affiliation(s)
- Tamer Mohamed Mahmoud
- Pharmaceutics and Industrial Pharmacy Department, Al-Manara College for Medical Sciences, Maysan 62010, Iraq;
| | | | - Mahmoud Mohamed Omar
- Department of Pharmaceutics and Pharmaceutical Technology, Deraya University, Minia 61519, Egypt;
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Omiya Ali Hasan
- Department of Pharmaceutics and Pharmaceutical Technology, Deraya University, Minia 61519, Egypt;
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Saad M. Wali
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Mohamed S. El-Mofty
- Oral Medicine, Periodontology, Oral Diagnosis and Radiology Department, Ain Shams University, Cairo 11566, Egypt;
- Oral Medicine, Periodontology, Oral Diagnosis and Radiology Department, Nahda University, Beni-Suef 62764, Egypt
| | - Mohamed G. Ewees
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Nahda University, Beni-Suef 62764, Egypt;
- Department of Pharmacology and Toxicology, College of Pharmacy, Almaaqal University, Basrah 61014, Iraq
| | - Amel E. Salem
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Cairo 11562, Egypt;
| | | | - Dina Mohamed Mahmoud
- Department of Pharmaceutics, Faculty of Pharmacy, Nahda University, Beni Suef 62764, Egypt;
| |
Collapse
|
10
|
Xie L, Hong Y, Hu Y, Li H, Lou J, Zhou X. Prioritizing oral bioavailability in drug development strategies. Future Med Chem 2025; 17:149-151. [PMID: 39723437 PMCID: PMC11749343 DOI: 10.1080/17568919.2024.2444871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Lijuan Xie
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, PR China
| | - Yingjing Hong
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, PR China
| | - Yun Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, PR China
| | - Hongmei Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, PR China
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, PR China
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming, PR China
| |
Collapse
|
11
|
Aulifa DL, Amarilis B, Ichsani LN, Maharani DS, Shabrina AM, Hanifah H, Wulandari RP, Rusdin A, Subra L, Budiman A. A Comprehensive Review: Mesoporous Silica Nanoparticles Greatly Improve Pharmacological Effectiveness of Phytoconstituent in Plant Extracts. Pharmaceuticals (Basel) 2024; 17:1684. [PMID: 39770526 PMCID: PMC11677945 DOI: 10.3390/ph17121684] [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: 10/24/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Medicinal plants are increasingly being explored due to their possible pharmacological properties and minimal adverse effects. However, low bioavailability and stability often limit efficacy, necessitating high oral doses to achieve therapeutic levels in the bloodstream. Mesoporous silica nanoparticles (MSNs) offer a potential solution to these limitations. Due to their large surface area, substantial pore volume, and ability to precisely control pore size. MSNs are also capable of efficiently incorporating a wide range of therapeutic substances, including herbal plant extracts, leading to potential use for drug containment and delivery systems. Therefore, this review aimed to discuss and summarize the successful developments of herbal plant extracts loaded into MSN, focusing on preparation, characterization, and the impact on efficacy. Data were collected from publications on Scopus, PubMed, and Google Scholar databases using the precise keywords "mesoporous silica nanoparticle" and "herbal extract". The results showed that improved phytoconstituent bioavailability, modified release profiles, increased stability, reduced dose and toxicity are the primary benefits of this method. This review offers insights on the significance of integrating MSNs into therapeutic formulations to improve pharmacological characteristics and effectiveness of medicinal plant extracts. Future prospects show favorable potential for therapeutic applications using MSNs combined with herbal medicines for clinical therapy.
Collapse
Affiliation(s)
- Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (R.P.W.); (A.R.)
| | - Bunga Amarilis
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Luthfia Nur Ichsani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Devita Salsa Maharani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Ayunda Myela Shabrina
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Hanifah Hanifah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| | - Rizky Prasiska Wulandari
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (R.P.W.); (A.R.)
| | - Agus Rusdin
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (R.P.W.); (A.R.)
| | - Laila Subra
- Department of Pharmacy, Faculty of Bioeconomic, Food and Health Sciences, Universiti Geomatika Malaysia, Kuala Lumpur 54200, Malaysia;
| | - Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (B.A.); (L.N.I.); (D.S.M.); (A.M.S.); (H.H.)
| |
Collapse
|
12
|
Said AR, Arafa MF, El-Dakroury WA, Alshehri S, El Maghraby GM. Bilosomes and Niosomes for Enhanced Intestinal Absorption and In Vivo Efficacy of Cytarabine in Treatment of Acute Myeloid Leukemia. Pharmaceuticals (Basel) 2024; 17:1572. [PMID: 39770414 PMCID: PMC11677554 DOI: 10.3390/ph17121572] [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: 10/23/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Cytarabine (CTR) is a hydrophilic anticancer drug used to treat leukemia. It suffers from poor permeability and intestinal metabolism, diminishing its oral bioavailability. BACKGROUND/OBJECTIVES The objective was to develop and evaluate niosomes and bilosomes for enhanced intestinal absorption; hence, oral bioavailability. RESULTS CTR-loaded niosomes and bilosomes with vesicle sizes of 152 and 204.3 nm were successfully prepared with acceptable properties. The presence of bile salts increased the zeta potential of bilosomes. The recorded entrapment efficiency of cytarabine was acceptable for such a hydrophilic drug. CTR-bilosomes showed a pH-dependent drug release pattern with preferred release in pH 6.8. Intestinal absorption behavior indicated a site-dependent CTR absorption pattern with unfavorable absorption in the distal intestine. Niosomal and bilosomal formulations enhanced intestinal absorption parameters with evidence for a predominant paracellular absorption mechanism that bypasses intestinal barriers. The investigation of the anti-leukemic effect of niosomal and bilosomal formulations indicated that both formulations ameliorated the blood parameters, reflecting significant improvement in leukemia treatment compared with the drug solution. Pathological examination of blood films revealed decreased blast cells in peripheral blood in groups treated with tested formulations. METHODS Tested formulations were prepared according to the pro-concentrate method and characterized for particle size, zeta potential, entrapment efficiency, and in vitro release. CTR-loaded niosomes and bilosomes were evaluated for enhanced intestinal absorption utilizing the single-pass in situ intestinal perfusion method in rabbits, and the anti-leukemic effect was assessed using the benzene-induced leukemia model in rats. CONCLUSIONS This study introduced surfactant vesicles for enhanced oral bioavailability of CTR.
Collapse
Affiliation(s)
- Abdelrahman R. Said
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (M.F.A.); (G.M.E.M.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Egypt
| | - Mona F. Arafa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (M.F.A.); (G.M.E.M.)
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Walaa A. El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Egypt
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Gamal M. El Maghraby
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (M.F.A.); (G.M.E.M.)
- Faculty of Pharmacy, Alsalam University, Tanta 31527, Egypt
| |
Collapse
|
13
|
Liu F, Meng F, Yang Z, Wang H, Ren Y, Cai Y, Zhang X. Exosome-biomimetic nanocarriers for oral drug delivery. CHINESE CHEM LETT 2024; 35:109335. [DOI: 10.1016/j.cclet.2023.109335] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Xie M, Meng F, Wang P, Díaz-García AM, Parkhats M, Santos-Oliveira R, Asim MH, Bostan N, Gu H, Yang L, Li Q, Yang Z, Lai H, Cai Y. Surface Engineering of Magnetic Iron Oxide Nanoparticles for Breast Cancer Diagnostics and Drug Delivery. Int J Nanomedicine 2024; 19:8437-8461. [PMID: 39170101 PMCID: PMC11338174 DOI: 10.2147/ijn.s477652] [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: 05/10/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Data published in 2020 by the International Agency for Research on Cancer (IARC) of the World Health Organization show that breast cancer (BC) has become the most common cancer globally, affecting more than 2 million women each year. The complex tumor microenvironment, drug resistance, metastasis, and poor prognosis constitute the primary challenges in the current diagnosis and treatment of BC. Magnetic iron oxide nanoparticles (MIONPs) have emerged as a promising nanoplatform for diagnostic tumor imaging as well as therapeutic drug-targeted delivery due to their unique physicochemical properties. The extensive surface engineering has given rise to multifunctionalized MIONPs. In this review, the latest advancements in surface modification strategies of MIONPs over the past five years are summarized and categorized as constrast agents and drug delivery platforms. Additionally, the remaining challenges and future prospects of MIONPs-based targeted delivery are discussed.
Collapse
Affiliation(s)
- Mengjie Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China / Guangdong Key Laboratory of Traditional Chinese Medicine Informatization / International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
| | - Fansu Meng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, 528400, People’s Republic of China
| | - Panpan Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
| | | | - Marina Parkhats
- B. I. Stepanov Institute of Physics, National Academy of Sciences of Belarus, Minsk, 220072, Belarus
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro, RJ, 21941906, Brazil
| | | | - Nazish Bostan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Honghui Gu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Lina Yang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Qi Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Zhenjiang Yang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, People’s Republic of China
| | - Haibiao Lai
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, 528400, People’s Republic of China
| | - Yu Cai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China / Guangdong Key Laboratory of Traditional Chinese Medicine Informatization / International Science and Technology Cooperation Base of Guangdong Province/School of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, People’s Republic of China
| |
Collapse
|
15
|
Fu S, Bao X, Mao Z, Lv Y, Zhu B, Chen Y, Zhou M, Tian S, Zhou F, Ding Z. Tetrastigma hemsleyanum polysaccharide ameliorates cytokine storm syndrome via the IFN-γ-JAK2/STAT pathway. Int J Biol Macromol 2024; 275:133427. [PMID: 38936586 DOI: 10.1016/j.ijbiomac.2024.133427] [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: 01/29/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an disease characterized by pulmonary edema and widespread inflammation, leading to a notably high mortality rate. The dysregulation of both pro-inflammatory and anti-inflammatory systems, results in cytokine storm (CS), is intricately associated with the development of ALI/ARDS. Tetrastigma hemsleyanum polysaccharide (THP) exerts remarkable anti-inflammatory and immunomodulatory effects against the disease, although its precise role in pathogenesis remains unclear. In the present study, an ALI/ARDS model was established using bacterial lipopolysaccharides. THP administration via aerosol inhalation significantly mitigated lung injury, reduced the number of inflammatory cells, and ameliorated glycerophospholipid metabolism. Furthermore, specific CS-related pathways were investigated by examining the synergy between tumor necrosis factor-α and interferon-γ used to establish CS models. The results indicated that THP effectively decreased inflammatory damage and cell death. The RNA sequencing revealed the involvement of the Janus kinase (JAK) 2-signal transducers and activators of transcription (STAT) signaling pathway in exerting the mentioned effects. Additionally, THP inhibited the activation of the JAK-STAT pathway, thereby alleviating the CS both in vivo and in vitro. Overall, THP exhibited marked therapeutic potential against ALI/ARDS and CS, primarily by targeting the IFN-γ-JAK2/STAT signaling pathway.
Collapse
Affiliation(s)
- Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Zian Mao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yishan Lv
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Bingqi Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yuchi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Mingyuan Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shasha Tian
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
16
|
Li H, Guan M, Zhang NN, Wang Y, Liang T, Wu H, Wang C, Sun T, Liu S. Harnessing nanomedicine for modulating microglial states in the central nervous system disorders: Challenges and opportunities. Biomed Pharmacother 2024; 177:117011. [PMID: 38917758 DOI: 10.1016/j.biopha.2024.117011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Microglia are essential for maintaining homeostasis and responding to pathological events in the central nervous system (CNS). Their dynamic and multidimensional states in different environments are pivotal factors in various CNS disorders. However, therapeutic modulation of microglial states is challenging due to the intricate balance these cells maintain in the CNS environment and the blood-brain barrier's restriction of drug delivery. Nanomedicine presents a promising avenue for addressing these challenges, offering a method for the targeted and efficient modulation of microglial states. This review covers the challenges faced in microglial therapeutic modulation and potential use of nanoparticle-based drug delivery systems. We provide an in-depth examination of nanoparticle applications for modulating microglial states in a range of CNS disorders, encompassing neurodegenerative and autoimmune diseases, infections, traumatic injuries, stroke, tumors, chronic pain, and psychiatric conditions. This review highlights the recent advancements and future prospects in nanomedicine for microglial modulation, paving the way for future research and clinical applications of therapeutic interventions in CNS disorders.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Meng Guan
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ning-Ning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
17
|
Chen J, Zhao Y, Cheng J, Wang H, Pan S, Liu Y. The Antiviral Potential of Perilla frutescens: Advances and Perspectives. Molecules 2024; 29:3328. [PMID: 39064906 PMCID: PMC11279397 DOI: 10.3390/molecules29143328] [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: 06/07/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Viruses pose a significant threat to human health, causing widespread diseases and impacting the global economy. Perilla frutescens, a traditional medicine and food homologous plant, is well known for its antiviral properties. This systematic review examines the antiviral potential of Perilla frutescens, including its antiviral activity, chemical structure and pharmacological parameters. Utilizing bioinformatics analysis, we revealed the correlation between Perilla frutescens and antiviral activity, identified overlaps between Perilla frutescens target genes and virus-related genes, and explored related signaling pathways. Moreover, a classified summary of the active components of Perilla frutescens, focusing on compounds associated with antiviral activity, provides important clues for optimizing the antiviral drug development of Perilla frutescens. Our findings indicate that Perilla frutescens showed a strong antiviral effect, and its active ingredients can effectively inhibit the replication and spread of a variety of viruses in this review. The antiviral mechanisms of Perilla frutescens may involve several pathways, including enhanced immune function, modulation of inflammatory responses, and inhibition of key enzyme activities such as viral replicase. These results underscore the potential antiviral application of Perilla frutescens as a natural plant and provide important implications for the development of new antiviral drugs.
Collapse
Affiliation(s)
- Jing Chen
- Department of Bioinformatics and Intelligent Diagnosis, School of Medicine, Jiangsu University, Zhenjiang 212003, China; (J.C.); (Y.Z.); (J.C.); (H.W.)
| | - Yi Zhao
- Department of Bioinformatics and Intelligent Diagnosis, School of Medicine, Jiangsu University, Zhenjiang 212003, China; (J.C.); (Y.Z.); (J.C.); (H.W.)
| | - Jie Cheng
- Department of Bioinformatics and Intelligent Diagnosis, School of Medicine, Jiangsu University, Zhenjiang 212003, China; (J.C.); (Y.Z.); (J.C.); (H.W.)
| | - Haoran Wang
- Department of Bioinformatics and Intelligent Diagnosis, School of Medicine, Jiangsu University, Zhenjiang 212003, China; (J.C.); (Y.Z.); (J.C.); (H.W.)
| | - Shu Pan
- Computer Science School, Jiangsu University of Science and Technology, Zhenjiang 212003, China;
| | - Yuwei Liu
- Department of Bioinformatics and Intelligent Diagnosis, School of Medicine, Jiangsu University, Zhenjiang 212003, China; (J.C.); (Y.Z.); (J.C.); (H.W.)
| |
Collapse
|
18
|
Zhang M, Yuan Q, Wang P, Zhang F, Wu D, Bai H, Liu J, Liu H, Yuan X. Bone Marrow Mesenchymal Stem Cell-Derived Nanovesicles Containing H8 Improve Hepatic Glucose and Lipid Metabolism and Exert Ameliorative Effects in Type 2 Diabetes. Int J Nanomedicine 2024; 19:6643-6658. [PMID: 38979532 PMCID: PMC11230129 DOI: 10.2147/ijn.s455021] [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: 01/23/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Purpose Nanovesicles (NVs) derived from bone mesenchymal stem cells (BMSCs) as drug delivery systems are considered an effective therapeutic strategy for diabetes. However, its mechanism of action remains unclear. Here, we evaluated the efficacy and molecular mechanism of BMSC-derived NVs carrying the curcumin analog H8 (H8-BMSCs-NVs) on hepatic glucose and lipid metabolism in type 2 diabetes (T2D). Subjects and Methods Mouse BMSCs were isolated by collagenase digestion and H8-BMSCs-NVs were prepared by microvesicle extrusion. The effects of H8-BMSCs-NVs on hepatic glucose and lipid metabolism were observed in a T2D mouse model and a HepG2 cell insulin resistance model. To evaluate changes in potential signaling pathways, the PI3K/AKT/AMPK signaling pathway and expression levels of G6P and PEPCK were assessed by Western blotting. Results H8-BMSCs-NVs effectively improved lipid accumulation in liver tissues and restored liver dysfunction in T2D mice. Meanwhile, H8-BMSCs-NVs effectively inhibited intracellular lipid accumulation in the insulin resistance models of HepG2 cells. Mechanistic studies showed that H8-BMSCs-NVs activated the PI3K/AKT/AMPK signaling pathway and decreased the expression levels of G6P and PEPCK. Conclusion These findings demonstrate that H8-BMSCs-NVs improved hepatic glucose and lipid metabolism in T2D mice by activating the PI3K/AKT/AMPK signaling pathway, which provides novel evidence suggesting the potential of H8-BMSCs-NVs in the clinically treatment of T2D patients.
Collapse
Affiliation(s)
- Meng Zhang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
- The First Hospital of Qiqihar, Qiqihar, People's Republic of China
| | - Qi Yuan
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Peiwen Wang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Fan Zhang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Dan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - He Bai
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Jieting Liu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Haifeng Liu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Xiaohuan Yuan
- College of Life Science, Mudanjiang Medical University, Mudanjiang, People's Republic of China
| |
Collapse
|
19
|
Kumar D, Sachdeva K, Tanwar R, Devi S. Review on novel targeted enzyme drug delivery systems: enzymosomes. SOFT MATTER 2024; 20:4524-4543. [PMID: 38738579 DOI: 10.1039/d4sm00301b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
The goal of this review is to present enzymosomes as an innovative means for site-specific drug delivery. Enzymosomes make use of an enzyme's special characteristics, such as its capacity to accelerate the reaction rate and bind to a particular substrate at a regulated rate. Enzymosomes are created when an enzyme forms a covalent linkage with a liposome or lipid vesicle surface. To construct enzymosomes with specialized activities, enzymes are linked using acylation, direct conjugation, physical adsorption, and encapsulation techniques. By reducing the negative side effects of earlier treatment techniques and exhibiting efficient medication release, these cutting-edge drug delivery systems improve long-term sickness treatments. They could be a good substitute for antiplatelet medication, gout treatment, and other traditional medicines. Recently developed supramolecular vesicular delivery systems called enzymosomes have the potential to improve drug targeting, physicochemical characteristics, and ultimately bioavailability in the pharmaceutical industry. Enzymosomes have advantages over narrow-therapeutic index pharmaceuticals as focusing on their site of action enhances both their pharmacodynamic and pharmacokinetic profiles. Additionally, it reduces changes in normal enzymatic activity, which enhances the half-life of an enzyme and accomplishes enzyme activity on specific locations.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmaceutical Sciences, Om Sterling Global University, Hisar, 125001, Haryana, India.
| | - Komal Sachdeva
- School of Pharmaceutical Sciences, Om Sterling Global University, Hisar, 125001, Haryana, India.
| | - Rajni Tanwar
- Department of Pharmaceutical Sciences, Starex University, Gurugram, India
| | - Sunita Devi
- School of Pharmaceutical Sciences, Om Sterling Global University, Hisar, 125001, Haryana, India.
| |
Collapse
|
20
|
Han R, He H, Lu Y, Lu H, Shen S, Wu W. Oral targeted drug delivery to post-gastrointestinal sites. J Control Release 2024; 370:256-276. [PMID: 38679163 DOI: 10.1016/j.jconrel.2024.04.047] [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: 02/18/2024] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
As an essential branch of targeted drug delivery, oral targeted delivery is attracting growing attention in recent years. In addition to site-specific delivery for the treatment of locoregional diseases in the gastrointestinal tract (GIT), oral targeted delivery to remote sites beyond the GIT emerges as a cutting-edge research topic. This review aims to provide an overview of the fundamental concepts and most recent advances in this field. Owing to the physiological barriers existing in the GIT, carrier systems should be transported across the enteric epithelia to target remote sites. Recently, pioneer investigations have validated the transport of intact micro- or nanocarriers across gastrointestinal barriers and subsequently to various distal organs and tissues. The microfold (M) cell pathway is the leading mechanism underlying the oral absorption of particulates, but the contribution of the transcellular and paracellular pathways should not be neglected either. In addition to well-acknowledged physicochemical and biological factors, the formation of a protein corona may also influence the biological fate of carrier systems. Although in an early stage of conceptualization, oral targeted delivery to remote diseases has demonstrated promising potential for the treatment of inflammation, tumors, and diseases inflicting the lymphatic and mononuclear phagocytosis systems.
Collapse
Affiliation(s)
- Rongze Han
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China; Fudan Zhangjiang Institute, Shanghai 201203, China
| | - Huiping Lu
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Shun Shen
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China; Fudan Zhangjiang Institute, Shanghai 201203, China.
| |
Collapse
|
21
|
Yin W, Ma H, Qu Y, Wang S, Zhao R, Yang Y, Guo ZN. Targeted exosome-based nanoplatform for new-generation therapeutic strategies. Biomed Mater 2024; 19:032002. [PMID: 38471163 DOI: 10.1088/1748-605x/ad3310] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/12/2024] [Indexed: 03/14/2024]
Abstract
Exosomes, typically 30-150 nm in size, are lipid-bilayered small-membrane vesicles originating in endosomes. Exosome biogenesis is regulated by the coordination of various mechanisms whereby different cargoes (e.g. proteins, nucleic acids, and lipids) are sorted into exosomes. These components endow exosomes with bioregulatory functions related to signal transmission and intercellular communication. Exosomes exhibit substantial potential as drug-delivery nanoplatforms owing to their excellent biocompatibility and low immunogenicity. Proteins, miRNA, siRNA, mRNA, and drugs have been successfully loaded into exosomes, and these exosome-based delivery systems show satisfactory therapeutic effects in different disease models. To enable targeted drug delivery, genetic engineering and chemical modification of the lipid bilayer of exosomes are performed. Stimuli-responsive delivery nanoplatforms designed with appropriate modifications based on various stimuli allow precise control of on-demand drug delivery and can be utilized in clinical treatment. In this review, we summarize the general properties, isolation methods, characterization, biological functions, and the potential role of exosomes in therapeutic delivery systems. Moreover, the effective combination of the intrinsic advantages of exosomes and advanced bioengineering, materials science, and clinical translational technologies are required to accelerate the development of exosome-based delivery nanoplatforms.
Collapse
Affiliation(s)
- Wenjing Yin
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Hongyin Ma
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Siji Wang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Ruoyu Zhao
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
- Neuroscience Research Center, Department of Neurology, The First Hospital of Jilin University, Chang Chun 130021, People's Republic of China
| |
Collapse
|
22
|
Liu J, You Q, Liang F, Ma L, Zhu L, Wang C, Yang Y. Ultrasound-nanovesicles interplay for theranostics. Adv Drug Deliv Rev 2024; 205:115176. [PMID: 38199256 DOI: 10.1016/j.addr.2023.115176] [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: 09/26/2023] [Revised: 12/04/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Nanovesicles (NVs) are widely used in the treatment and diagnosis of diseases due to their excellent vascular permeability, good biocompatibility, high loading capacity, and easy functionalization. However, their yield and in vivo penetration depth limitations and their complex preparation processes still constrain their application and development. Ultrasound, as a fundamental external stimulus with deep tissue penetration, concentrated energy sources, and good safety, has been proven to be a patient-friendly and highly efficient strategy to overcome the restrictions of traditional clinical medicine. Recent research has shown that ultrasound can drive the generation of NVs, increase their yield, simplify their preparation process, and provide direct therapeutic effects and intelligent control to enhance the therapeutic effect of NVs. In addition, NVs, as excellent drug carriers, can enhance the targeting efficiency of ultrasound-based sonodynamic therapy or sonogenetic regulation and improve the accuracy of ultrasound imaging. This review provides a detailed introduction to the classification, generation, and modification strategies of NVs, emphasizing the impact of ultrasound on the formation of NVs and summarizing the enhanced treatment and diagnostic effects of NVs combined with ultrasound for various diseases.
Collapse
Affiliation(s)
- Jingyi Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing You
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Fuming Liang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lilusi Ma
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
23
|
Wang X, Sun H, Mu T. Materials and structure of polysaccharide-based delivery carriers for oral insulin: A review. Carbohydr Polym 2024; 323:121364. [PMID: 37940264 DOI: 10.1016/j.carbpol.2023.121364] [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: 06/18/2023] [Revised: 08/24/2023] [Accepted: 09/02/2023] [Indexed: 11/10/2023]
Abstract
Diabetes mellitus is a chronic metabolic disease that affects >500 million patients worldwide. Subcutaneous injection of insulin is the most effective treatment at present. However, regular needle injections will cause pain, inflammation, and other adverse consequences. In recent years, significant progress has been made in non-injectable insulin preparations. Oral administration is the best way of administration due to its simplicity, convenience, and good patient compliance. However, oral insulin delivery is hindered by many physiological barriers in the gastrointestinal tract, resulting in the low relative bioavailability of direct oral insulin delivery. To improve the relative bioavailability, a variety of insulin delivery vectors have been developed. Polysaccharides are used to achieve safe and effective insulin loading due to their excellent biocompatibility and protein affinity. The functional characteristics of polysaccharide-based delivery carriers, such as pH responsiveness, mucosal adhesion, and further functionalization modifications, enhance the gastrointestinal absorption and bioavailability of insulin. This paper reviews the materials and structures of oral insulin polysaccharide-based carriers, providing ideas for further improving the relative bioavailability of oral insulin.
Collapse
Affiliation(s)
- Xinran Wang
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China
| | - Hongnan Sun
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China.
| | - Taihua Mu
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, No. 2 Yuan Ming Yuan West Road, Haidian District, P.O. Box 5109, Beijing 100193, China.
| |
Collapse
|
24
|
Faustino C, Duarte N, Pinheiro L. Triterpenes Drug Delivery Systems, a Modern Approach for Arthritis Targeted Therapy. Pharmaceuticals (Basel) 2023; 17:54. [PMID: 38256888 PMCID: PMC10819636 DOI: 10.3390/ph17010054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Arthritis is a major cause of disability. Currently available anti-arthritic drugs, such as disease-modifying anti-rheumatic drugs (DMARDs), have serious side-effects associated with long-term use. Triterpenoids are natural products with known anti-inflammatory properties, and many have revealed efficiency against arthritis both in vitro and in vivo in several animal models, with negligible cytotoxicity. However, poor bioavailability due to low water solubility and extensive metabolism upon oral administration hinder the therapeutic use of anti-arthritic triterpenoids. Therefore, drug delivery systems (DDSs) able to improve the pharmacokinetic profile of triterpenoids and achieve sustained drug release are useful alternatives for targeted delivery in arthritis treatment. Several DDSs have been described in the literature for triterpenoid delivery, including microparticulate and nanoparticulate DDSs, such as polymeric micro and nanoparticles (NPs), polymeric micelles, liposomes, micro and nanoemulsions, and hydrogels. These systems have shown superior therapeutic effects in arthritis compared to the free drugs and are similar to currently available anti-arthritic drugs without significant side-effects. This review focuses on nanocarriers for triterpenoid delivery in arthritis therapy, including osteoarthritis (OA), rheumatoid arthritis (RA) and gout that appeared in the literature in the last ten years.
Collapse
Affiliation(s)
| | - Noélia Duarte
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Lídia Pinheiro
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| |
Collapse
|
25
|
Xiang T, Liu Y, Xu S, Zhong W, Sha Z, Zhang J, Chen L, Li Y, Li W, Yan Z, Chen Z, Xu L. Construction of a novel amphiphilic peptide paclitaxel rod micelle: Demonstrating that the nano-delivery system shape can affect the cellular uptake efficiency of paclitaxel and improve the therapeutic efficacy for breast cancer. BIOMATERIALS ADVANCES 2023; 155:213673. [PMID: 39491929 DOI: 10.1016/j.bioadv.2023.213673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/05/2024]
Abstract
Recent studies have shown that the morphology of nano-delivery systems has become a key factor affecting their anti-tumor effects. Although it has been demonstrated that rod-like nanoparticles are more easily absorbed by tumor cells, the application of rod-like nanoparticles is still limited by the lack of safe vector in vivo. In this study, a biocompatible amphiphilic peptide (IIQQQQ, I2Q4), was designed to form rod-like micelles. The key forces of the self-assembly mechanism were investigated. Driven by hydrogen bonds, the hydrophilic segment of the peptide formed a β-sheet structure, and the molecules accumulated and extended along the side chain direction to form a rod-like structure. Using paclitaxel (PTX) as the model drug, a PTX rod-like nano-drug delivery system, PTX@I2Q4, was constructed. PTX exists in a randomly coiled state in the hydrophobic cavity formed by the peptide. Compared to PTX and spherical PTX albumin nanoparticles, PTX@I2Q4 showed higher entry efficiency and better antitumor effects in vivo and in vitro. This was mainly because PTX@I2Q4 not only allowed more efficient entry into cells via macro-pinocytosis, but also significantly prolonged the t1/2 of PTX. The results confirmed the feasibility of regulating the morphology of nanoparticles to improve the efficacy of PTX and provide a reference for further research on the influence of the morphology of the nano-drug delivery system on the efficacy of antitumor effects.
Collapse
Affiliation(s)
- Tangyong Xiang
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Yu Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Shan Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Weixi Zhong
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Zhengzhou Sha
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Jian Zhang
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Linwei Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Yarong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Weidong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China
| | - Zheng Yan
- Jiangyin Hospital of Traditional Chinese Medicine, Jiangsu, Jiangyin, 214400, PR China.
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China.
| | - Liu Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, 210023, PR China.
| |
Collapse
|
26
|
Pires PC, Paiva-Santos AC, Veiga F. Liposome-Derived Nanosystems for the Treatment of Behavioral and Neurodegenerative Diseases: The Promise of Niosomes, Transfersomes, and Ethosomes for Increased Brain Drug Bioavailability. Pharmaceuticals (Basel) 2023; 16:1424. [PMID: 37895895 PMCID: PMC10610493 DOI: 10.3390/ph16101424] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Psychiatric and neurodegenerative disorders are amongst the most prevalent and debilitating diseases, but current treatments either have low success rates, greatly due to the low permeability of the blood-brain barrier, and/or are connected to severe side effects. Hence, new strategies are extremely important, and here is where liposome-derived nanosystems come in. Niosomes, transfersomes, and ethosomes are nanometric vesicular structures that allow drug encapsulation, protecting them from degradation, and increasing their solubility, permeability, brain targeting, and bioavailability. This review highlighted the great potential of these nanosystems for the treatment of Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, anxiety, and depression. Studies regarding the encapsulation of synthetic and natural-derived molecules in these systems, for intravenous, oral, transdermal, or intranasal administration, have led to an increased brain bioavailability when compared to conventional pharmaceutical forms. Moreover, the developed formulations proved to have neuroprotective, anti-inflammatory, and antioxidant effects, including brain neurotransmitter level restoration and brain oxidative status improvement, and improved locomotor activity or enhancement of recognition and working memories in animal models. Hence, albeit being relatively new technologies, niosomes, transfersomes, and ethosomes have already proven to increase the brain bioavailability of psychoactive drugs, leading to increased effectiveness and decreased side effects, showing promise as future therapeutics.
Collapse
Affiliation(s)
- Patrícia C. Pires
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Cláudia Paiva-Santos
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Faculty of Pharmacy, Faculty of Pharmacy of the University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
27
|
Peng X, Li X, Xie B, Lai Y, Sosnik A, Boucetta H, Chen Z, He W. Gout therapeutics and drug delivery. J Control Release 2023; 362:728-754. [PMID: 37690697 DOI: 10.1016/j.jconrel.2023.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Gout is a common inflammatory arthritis caused by persistently elevated uric acid levels. With the improvement of people's living standards, the consumption of processed food and the widespread use of drugs that induce elevated uric acid, gout rates are increasing, seriously affecting the human quality of life, and becoming a burden to health systems worldwide. Since the pathological mechanism of gout has been elucidated, there are relatively effective drug treatments in clinical practice. However, due to (bio)pharmaceutical shortcomings of these drugs, such as poor chemical stability and limited ability to target the pathophysiological pathways, traditional drug treatment strategies show low efficacy and safety. In this scenario, drug delivery systems (DDS) design that overcome these drawbacks is urgently called for. In this review, we initially describe the pathological features, the therapeutic targets, and the drugs currently in clinical use and under investigation to treat gout. We also comprehensively summarize recent research efforts utilizing lipid, polymeric and inorganic carriers to develop advanced DDS for improved gout management and therapy.
Collapse
Affiliation(s)
- Xiuju Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
28
|
Zarenezhad E, Marzi M, Abdulabbas HT, Jasim SA, Kouhpayeh SA, Barbaresi S, Ahmadi S, Ghasemian A. Bilosomes as Nanocarriers for the Drug and Vaccine Delivery against Gastrointestinal Infections: Opportunities and Challenges. J Funct Biomater 2023; 14:453. [PMID: 37754867 PMCID: PMC10531812 DOI: 10.3390/jfb14090453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 09/28/2023] Open
Abstract
The gastrointestinal tract (GIT) environment has an intricate and complex nature, limiting drugs' stability, oral bioavailability, and adsorption. Additionally, due to the drugs' toxicity and side effects, renders are continuously seeking novel delivery systems. Lipid-based drug delivery vesicles have shown various loading capacities and high stability levels within the GIT. Indeed, most vesicular platforms fail to efficiently deliver drugs toward this route. Notably, the stability of vesicular constructs is different based on the different ingredients added. A low GIT stability of liposomes and niosomes and a low loading capacity of exosomes in drug delivery have been described in the literature. Bilosomes are nonionic, amphiphilic, flexible surfactant vehicles that contain bile salts for the improvement of drug and vaccine delivery. The bilosomes' stability and plasticity in the GIT facilitate the efficient carriage of drugs (such as antimicrobial, antiparasitic, and antifungal drugs), vaccines, and bioactive compounds to treat infectious agents. Considering the intricate and harsh nature of the GIT, bilosomal formulations of oral substances have a remarkably enhanced delivery efficiency, overcoming these conditions. This review aimed to evaluate the potential of bilosomes as drug delivery platforms for antimicrobial, antiviral, antifungal, and antiparasitic GIT-associated drugs and vaccines.
Collapse
Affiliation(s)
- Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa P.O. Box 7461686688, Iran; (E.Z.); (M.M.); (S.A.)
| | - Mahrokh Marzi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa P.O. Box 7461686688, Iran; (E.Z.); (M.M.); (S.A.)
| | - Hussein T. Abdulabbas
- Department of Medical Microbiology, Medical College, Al Muthanna University, Al Muthanna P.O. Box 07835544777, Iraq;
| | | | - Seyed Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa P.O. Box 7461686688, Iran;
| | - Silvia Barbaresi
- Department of Movement and Sports Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Shiva Ahmadi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa P.O. Box 7461686688, Iran; (E.Z.); (M.M.); (S.A.)
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa P.O. Box 7461686688, Iran; (E.Z.); (M.M.); (S.A.)
| |
Collapse
|
29
|
Kwong KWY, Xin Y, Lai NCY, Sung JCC, Wu KC, Hamied YK, Sze ETP, Lam DMK. Oral Vaccines: A Better Future of Immunization. Vaccines (Basel) 2023; 11:1232. [PMID: 37515047 PMCID: PMC10383709 DOI: 10.3390/vaccines11071232] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Oral vaccines are gaining more attention due to their ease of administration, lower invasiveness, generally greater safety, and lower cost than injectable vaccines. This review introduces certified oral vaccines for adenovirus, recombinant protein-based, and transgenic plant-based oral vaccines, and their mechanisms for inducing an immune response. Procedures for regulatory approval and clinical trials of injectable and oral vaccines are also covered. Challenges such as instability and reduced efficacy in low-income countries associated with oral vaccines are discussed, as well as recent developments, such as Bacillus-subtilis-based and nanoparticle-based delivery systems that have the potential to improve the effectiveness of oral vaccines.
Collapse
Affiliation(s)
- Keith Wai-Yeung Kwong
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Ying Xin
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
| | - Nelson Cheuk-Yin Lai
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Johnny Chun-Chau Sung
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Kam-Chau Wu
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
| | | | - Eric Tung-Po Sze
- School of Science and Technology, Hong Kong Metropolitan University, Hong Kong, China
| | - Dominic Man-Kit Lam
- DrD Novel Vaccines Limited, Hong Kong, China
- Torsten Wiesel International Research Institute, Sichuan University, Chengdu 610064, China
| |
Collapse
|
30
|
Li JD, Yin J. Interleukin-10-alveolar macrophage cell membrane-coated nanoparticles alleviate airway inflammation and regulate Th17/regulatory T cell balance in a mouse model. Front Immunol 2023; 14:1186393. [PMID: 37275919 PMCID: PMC10235466 DOI: 10.3389/fimmu.2023.1186393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/27/2023] [Indexed: 06/07/2023] Open
Abstract
Background Allergic airway disease (AAD) is a chronic disease characterized by airway inflammation, bronchoconstriction, and hyperresponsiveness. Although exogenous interleukin-10 (IL-10) alleviates allergic inflammation, it has a short half-life in vivo. Cell membrane-coated nanomaterials have been shown to protect therapeutic payloads and increase therapeutic efficacy. Objective This study was aimed at investigating the efficacy of a novel macrophage-based nanoparticle drug for the treatment of house dust mite (HDM)-induced allergic airway diseases. Methods IL-10-poly (lactic-co-glycolic acid (PLGA) nanoparticles were encapsulated in alveolar macrophage cell membranes. An allergic airway disease mouse model was established by repeated inhalation of HDM extracts. The mice were treated with free IL-10, IL-10-PLGA nanoparticles (IL10-NP), or IL-10-alveolar macrophage cell membrane-coated nanoparticles (IL10-AMNP). The therapeutic effects were evaluated by measuring airway hyperresponsiveness, lung inflammation, cytokine levels, and regulatory T cells (Treg)- T-helper 17 (Th17) cell balance. Results Compared to free IL-10, IL10-AMNP significantly reduced airway hyperresponsiveness and T-helper 2 (Th2)/Th17 cytokines and inhibited neutrophilia and eosinophilia recruitment into the airways of HDM-induced mouse models. Additionally, the balance between Tregs and Th17 cells was significantly improved in groups treated with IL10-AMNP. Conclusion This study demonstrated that PLGA nanoparticle cores coated with alveolar macrophage cell membranes can effectively deliver therapeutic cytokines to the lungs and improve the homeostatic balance between Tregs and Th17 cells. These findings suggest that macrophage-based nanoparticle drugs represent a promising approach for treating allergic airway diseases.
Collapse
Affiliation(s)
- Jun-Da Li
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Allergy, Peking Union Medical College Hospital, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, China
- Department of Allergy, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Disease, Beijing, China
| | - Jia Yin
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Allergy, Peking Union Medical College Hospital, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Beijing, China
- Department of Allergy, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Disease, Beijing, China
| |
Collapse
|
31
|
Zafar A, Arshad R, Ur.Rehman A, Ahmed N, Akhtar H. Recent Developments in Oral Delivery of Vaccines Using Nanocarriers. Vaccines (Basel) 2023; 11:490. [PMID: 36851367 PMCID: PMC9964829 DOI: 10.3390/vaccines11020490] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
As oral administration of vaccines is the preferred route due to its high patient compliance and ability to stimulate both cellular and humoral immune responses, it is also associated with several challenges that include denaturation of vaccine components in the acidic environment of the stomach, degradation from proteolytic enzymes, and poor absorption through the intestinal membrane. To achieve effective delivery of such biomolecules, there is a need to investigate novel strategies of formulation development that can overcome the barriers associated with conventional vaccine delivery systems. Nanoparticles are advanced drug delivery carriers that provide target-oriented delivery by encapsulating vaccine components within them, thus making them stable against unfavorable conditions. This review provides a detailed overview of the different types of nanocarriers and various approaches that can enhance oral vaccine delivery.
Collapse
Affiliation(s)
- Amna Zafar
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Raffia Arshad
- Yusra Institute of Pharmaceutical Sciences, Yusra Medical and Dental College, Islamabad 45730, Pakistan
| | - Asim Ur.Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Naveed Ahmed
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Hashaam Akhtar
- Yusra Institute of Pharmaceutical Sciences, Yusra Medical and Dental College, Islamabad 45730, Pakistan
| |
Collapse
|
32
|
Fu X, Chen K, Li Z, Fan H, Xu B, Liu M, Guo L, Xie Z, Liu K, Zhang S, Kou L. Pharmacokinetics and Oral Bioavailability of Panax Notoginseng Saponins Administered to Rats Using a Validated UPLC-MS/MS Method. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:469-479. [PMID: 36576094 DOI: 10.1021/acs.jafc.2c06312] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Panax notoginseng saponins (PNS) are the most important bioactive components of P. Notoginseng. In this paper, an evaluation of the pharmacokinetics and oral absolute bioavailability of PNS was carried out following intravenous and oral administration of PNS to Sprague-Dawley rats. The plasma concentration of 28 PNS was determined using a validated UPLC-MS/MS system. The results demonstrated that Rb1(32.8%), Rg1(41.4%), R1(9.4%), Re(4.5%), and Rd(3.5%) are the five main ingredients of PNS for administration. After oral administration, it was found that the area under the curve (AUC0-72 h) for these five major saponins was significantly different. AUC0-72 h of Rb1 and Rd accounted for about 60% of all PNS exposure, while AUC0-72 h of Rg1 and R1 only accounted for 0.7%, and Re was undetectable in plasma. Also, PPD, PPT, and CK were detected as the major PNS metabolites in vivo. Furthermore, it was shown that the total oral bioavailability of PNS was only 1.2%.
Collapse
Affiliation(s)
- Xinzhen Fu
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| | - Kun Chen
- School of Pharmacy, Yantai University, Yantai264003, China
| | - Zhi Li
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| | - Huaying Fan
- School of Pharmacy, Yantai University, Yantai264003, China
| | - Bo Xu
- School of Pharmacy, Yantai University, Yantai264003, China
| | - Ming Liu
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| | - Lin Guo
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| | - Zeping Xie
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| | - Ke Liu
- Shandong Boyuan Biomedical Co., Ltd., Yantai264003, China
| | - Shumin Zhang
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| | - Lijuan Kou
- School of Pharmacy, Binzhou Medical University, Yantai264003, China
| |
Collapse
|