1
|
Girase R, Gujarathi NA, Sukhia A, Kota SSN, Patil TS, Aher AA, Agrawal YO, Ojha S, Sharma C, Goyal SN. Targeted nanoliposomes for precision rheumatoid arthritis therapy: a review on mechanisms and in vivo potential. Drug Deliv 2025; 32:2459772. [PMID: 39891600 PMCID: PMC11789225 DOI: 10.1080/10717544.2025.2459772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory immune-triggered disease that causes synovitis, cartilage degradation, and joint injury. In nanotechnology, conventional liposomes were extensively investigated for RA. However, they frequently undergo rapid clearance, reducing circulation time and therapeutic efficacy. Additionally, their stability in the bloodstream is often compromised, resulting in premature drug release. The current review explores the potential of targeted liposomal-based nanosystems in the treatment of RA. It highlights the pathophysiology of RA, explores selective targeting sites, and elucidates diverse mechanisms of novel liposomal types and their applications. Furthermore, the targeting strategies of pH-sensitive, flexible, surface-modified, PEGylated, acoustic, ROS-mediated, and biofunctionalized liposomes are addressed. Targeted nanoliposomes showed potential in precisely delivering drugs to CD44, SR-A, FR-β, FLS, and toll-like receptors through the high affinity of ligands. In vitro studies interpreted stable release profiles and improved stability. Ex vivo studies on skin demonstrated that ultradeformable and glycerol-conjugated liposomes enhanced drug penetrability. In vivo experiments for liposomal types in the arthritis rat model depicted remarkable efficacy in reducing joint swelling, pro-inflammatory cytokines, and synovial hyperplasia. In conclusion, these targeted liposomes represented a significant leap forward in drug delivery, offering effective therapeutic options for RA. In the future, integrating these advanced liposomes with artificial intelligence, immunotherapy, and precision medicine holds great promise.
Collapse
Affiliation(s)
- Rushikesh Girase
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| | | | - Amey Sukhia
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sri Sai Nikitha Kota
- Department of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | | | - Abhijeet A. Aher
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| | | | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| |
Collapse
|
2
|
Yalamandala BN, Huynh TMH, Lien HW, Pan WC, Iao HM, Moorthy T, Chang YH, Hu SH. Advancing brain immunotherapy through functional nanomaterials. Drug Deliv Transl Res 2025:10.1007/s13346-024-01778-5. [PMID: 39789307 DOI: 10.1007/s13346-024-01778-5] [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] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
Glioblastoma (GBM), a highly aggressive brain tumor, poses significant treatment challenges due to its highly immunosuppressive microenvironment and the brain immune privilege. Immunotherapy activating the immune system and T lymphocyte infiltration holds great promise against GBM. However, the brain's low immunogenicity and the difficulty of crossing the blood-brain barrier (BBB) hinder therapeutic efficacy. Recent advancements in immune-actuated particles for targeted drug delivery have shown the potential to overcome these obstacles. These particles interact with the BBB by rapidly and reversibly disrupting its structure, thereby significantly enhancing targeting and penetrating delivery. The BBB targeting also minimizes potential long-term damage. At GBM, the particles demonstrated effective chemotherapy, chemodynamic therapy, photothermal therapy (PTT), photodynamic therapy (PDT), radiotherapy, or magnetotherapy, facilitating tumor disruption and promoting antigen release. Additionally, components of the delivery system retained autologous tumor-associated antigens and presented them to dendritic cells (DCs), ensuring prolonged immune activation. This review explores the immunosuppressive mechanisms of GBM, existing therapeutic strategies, and the role of nanomaterials in enhancing immunotherapy. We also discuss innovative particle-based approaches designed to traverse the BBB by mimicking innate immune functions to improve treatment outcomes for brain tumors.
Collapse
Affiliation(s)
- Bhanu Nirosha Yalamandala
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Thi My Hue Huynh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Hui-Wen Lien
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Wan-Chi Pan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Hoi Man Iao
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Thrinayan Moorthy
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Yun-Hsuan Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan.
| |
Collapse
|
3
|
Jin M, Wu H, Jin W, Zeng B, Liu Y, Wang N, Wang S, Chen L, Gao Z, Huang W. Transferrin Protein Corona-Targeted Codelivery of Tirapazamine and IR820 Facilitates Efficient PDT-Induced Hypoxic Chemotherapy on 4T1 Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:1892-1910. [PMID: 39699197 DOI: 10.1021/acsami.4c15045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Protein corona (PC) formation confers novel biological properties to the original nanomaterial, impeding its uptake and targeting efficacy in cells and tissues. Although many studies discussing PC formation have focused on inert proteins that may inhibit the function of nanomaterials, some functional plasma proteins with intrinsic targeting capabilities can also be adsorbed to the surface of nanomaterials, with active ligand properties to improve the targeting ability. In this approach, nanomaterials are surface-engineered to promote the adsorption of specific functional plasma proteins that are directly targeted to transport nanomaterials to the target site. In this study, T10 peptide-modified liposomes were employed to construct an in situ transferrin (Tf) PC-mediated liposome carrying a hypoxia-sensitive chemotherapy drug (tirapazamine, TPZ) and a photosensitizer (indocyanine green, IR820). The water-soluble drug TPZ was encapsulated in mesoporous silica nanoparticles (MSNs) and coated with IR820 (IR)-loaded liposome. Lipid-coated MSNs can inhibit aggregation in the body and significantly reduce the rapid release of water-soluble drugs, resulting in improved system stability and sustained release. Upon entering the in vivo circulation, T10 bound specifically to Tf in plasma to form an in situ Tf liposome-PC complex with enhanced targeting efficacy compared to traditional ligand-modified active-targeting strategies. However, large-sized PC particles faced challenges in penetrating deep into tumor tissues. IR could kill tumors through photodynamic therapy (PDT) and elicit complementary antitumor effects with the hypoxia-sensitive drug TPZ. This study demonstrates the novel design of in situ PC-mediated multifunctional liposomes for hypoxia-activated chemotherapy combined with PDT, a promising approach to cancer therapy.
Collapse
Affiliation(s)
- Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, Jilin Medical University, Jilin 132013, China
| | - Wenyu Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Dermatology, Yanbian University Hospital, Yanji 133000, China
| | - Bowen Zeng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Nuoya Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuangqing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
4
|
Wu E, Guan J, Yu Y, Lin S, Ding T, Chu Y, Pan F, Liu M, Yang Y, Zhang Z, Zhang J, Zhan C, Qian J. Exemplifying interspecies variation of liposome in vivo fate by the effects of anti-PEG antibodies. Acta Pharm Sin B 2024; 14:4994-5007. [PMID: 39664439 PMCID: PMC11628802 DOI: 10.1016/j.apsb.2024.07.009] [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: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 12/13/2024] Open
Abstract
The different fate of liposomes among species has been discovered and mentioned in many studies, but the underlying mechanisms have not been explored. In the present work, we concentrated on the in vivo fate of PEGylated liposomes (sLip) in three commonly used species (mice, rats, and dogs). It was exhibited that the accelerated blood clearance (ABC) phenomenon and hypersensitivity in large animals (beagle dogs) were much more significant than that in rodents. We demonstrated that anti-PEG IgM (partially) and complement (mostly) determined the elimination of sLip and linked the distinct interspecies performances with the diverse complement capacity among species. Based on the data from animals and clinical patients, it was revealed that the fate of sLip in large animals was closer to that in humans, for the sufficient complement capacity could expose the potential adverse reactions caused by anti-PEG antibodies. Our results suggested that the distinctive interspecies performances of sLip were highly related to the physiological variabilities among species, which should not be overlooked in the innovation and translation of nanomedicines.
Collapse
Affiliation(s)
- Ercan Wu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Juan Guan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Yifei Yu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Shiqi Lin
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Tianhao Ding
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Yuxiu Chu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Mengyuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Zui Zhang
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Jun Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| |
Collapse
|
5
|
Yang Y, Chu Y, Li C, Fan L, Lu H, Zhan C, Zhang Z. Brain-targeted drug delivery by in vivo functionalized liposome with stable D-peptide ligand. J Control Release 2024; 373:240-251. [PMID: 38977135 DOI: 10.1016/j.jconrel.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/17/2024] [Accepted: 07/05/2024] [Indexed: 07/10/2024]
Abstract
Brain-targeted drug delivery poses a great challenge due to the blood-brain barrier (BBB). In a previous study, we have developed a peptide-modified stealth liposome (SP-sLip) to enhance BBB penetration via the adsorption of apolipoproteins in plasma. SP is an 11-amino acid peptide derived from 25 to 35 of the Amyloid β peptide (Aβ1-42), which is a nature ligand of apolipoproteins. Although freshly prepared SP-sLip exhibited efficient brain targeting performance, it occured self-aggregation and instability in storage. In this study, we developed a D-peptide ligand according to the reverse sequence of SP with D-amino acids, known as DSP, to improve the stability in storage. Notably, DSP exhibited a reduced tendency for self-aggregation and improved stability in comparison to the SP peptide. Furthermore, compared to SP-sLip, DSP-modified sLip (DSP-sLip) demonstrated enhanced stability (>2 weeks), prolonged blood circulation (AUC increased 44.4%), reduced liver and spleen accumulation (reduced by 2.23 times and 1.86 times) with comparable brain-targeting efficiency. Similar to SP-sLip, DSP-sLip selectively adsorbed apolipoprotein A1, E, and J in the blood to form functionalized protein corona, thus crossing BBB via apolipoprotein receptor-mediated transcytosis. These findings underscored the importance of ligand stability in the in vitro and in vivo performance of brain-targeted liposomes, therefore paving the way for the design and optimization of efficient and stable nanocarriers.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Yuxiu Chu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Cheng Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Lianfeng Fan
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China.
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201399, China; Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| | - Zui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
6
|
Zhu J, Xu L, Wang W, Xiao M, Li J, Wang L, Jiang X. Molecular Dynamics Simulations Reveal Octanoylated Hyaluronic Acid Enhances Liposome Stability, Stealth and Targeting. ACS OMEGA 2024; 9:33833-33844. [PMID: 39130542 PMCID: PMC11307277 DOI: 10.1021/acsomega.4c03526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Liposome-based drug delivery systems have been widely used in drug and gene delivery. However, issues such as instability, immune clearance, and poor targeting have significantly limited their clinical utility. Consequently, there is an urgent need for innovative strategies to improve liposome performance. In this study, we explore the interaction mechanisms of hyaluronic acid (HA), a linear anionic polysaccharide composed of repeating disaccharide units of d-glucuronic acid and N-acetyl-d-glucosamine connected by alternating β-1,3 and β-1,4 glycosidic linkages, and its octanoylated derivates (OHA) with liposomes using extensive coarse-grained molecular dynamics simulations. The octyl moieties of OHA spontaneously inserted into the phospholipid bilayer of liposomes, leading to their effective coating onto the surface of liposome and enhancing their structural stability. Furthermore, encapsulating liposome with OHA neutralized their surface potential, interfering with the formation of a protein corona known to contribute to liposomal immune clearance. Importantly, the encapsulated OHA maintained its selectivity and therefore targeting ability for CD44, which is often overexpressed in tumor cells. These molecular-scale findings shed light on the interaction mechanisms between HA and liposomes and will be useful for the development of next-generation liposome-based drug delivery systems.
Collapse
Affiliation(s)
- Jingyi Zhu
- State
Key Laboratory of Microbial Technology, National Glycoengineering
Research Center, Shandong University, Qingdao 266237, China
| | - Limei Xu
- State
Key Laboratory of Microbial Technology, National Glycoengineering
Research Center, Shandong University, Qingdao 266237, China
| | - Wenxin Wang
- Shandong
Institute for Food and Drug Control, Jinan 250000, China
| | - Min Xiao
- State
Key Laboratory of Microbial Technology, National Glycoengineering
Research Center, Shandong University, Qingdao 266237, China
| | - Jian Li
- Biomedicine
Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Lushan Wang
- State
Key Laboratory of Microbial Technology, National Glycoengineering
Research Center, Shandong University, Qingdao 266237, China
| | - Xukai Jiang
- State
Key Laboratory of Microbial Technology, National Glycoengineering
Research Center, Shandong University, Qingdao 266237, China
| |
Collapse
|
7
|
Liu M, Wu E, Pan F, Tian K, Fu J, Yu Y, Guo Z, Ma Y, Wei A, Yu X, Zhan C, Qian J. Effects of drug-induced liver injury on the in vivo fate of liposomes. Eur J Pharm Biopharm 2024; 201:114389. [PMID: 38945407 DOI: 10.1016/j.ejpb.2024.114389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
Liposomes represent one of the most extensively studied nano-carriers due to their potential in targeted drug delivery. However, the complex in vivo fate, particularly under pathological conditions, presents challenges for clinical translation of liposomal therapeutics. Liver serves as the most important organ for liposome accumulation and metabolism. Unfortunately, the fate of liposomes under pathological liver conditions has been significantly overlooked. This study aimed to investigate the in vivo pharmacokinetic profile and biodistribution profile of liposomes under drug-induced liver injury (DILI) conditions. Two classic DILI animal models, i.e. acetaminophen-induced acute liver injury (AILI) and triptolide-induced subacute liver injury (TILI), were established to observe the effect of pathological liver conditions on the in vivo performance of liposomes. The study revealed significant changes in the in vivo fate of liposomes following DILI, including prolonged blood circulation and enhanced hepatic accumulation of liposomes. Changes in the composition of plasma proteins and mononuclear phagocyte system (MPS)-related cell subpopulations collectively led to the altered in vivo fate of liposomes under liver injury conditions. Despite liver injury, macrophages remained the primary cells responsible for liposomes uptake in liver, with the recruited monocyte-derived macrophages exhibiting enhanced ability to phagocytose liposomes under pathological conditions. These findings indicated that high capture of liposomes by the recruited hepatic macrophages not only offered potential solutions for targeted delivery, but also warned the clinical application of patients under pathological liver conditions.
Collapse
Affiliation(s)
- Mengyuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China
| | - Ercan Wu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China
| | - Kaisong Tian
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Jiaru Fu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yifei Yu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Zhiwei Guo
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Yinyu Ma
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Anqi Wei
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China
| | - Xiaoyue Yu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China.
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, PR China.
| | - Jun Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai 201203, PR China.
| |
Collapse
|
8
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
9
|
Ding T, Wang Y, Meng Y, Wu E, Shao Q, Lin S, Yu Y, Qian J, He Q, Zhang J, Wang J, Kohane DS, Zhan C. Reciprocal Interaction with Neutrophils Facilitates Cutaneous Accumulation of Liposomes. ACS NANO 2024; 18:18769-18784. [PMID: 38950189 DOI: 10.1021/acsnano.4c06638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Liposomes are versatile drug delivery systems in clinical use for cancer and many other diseases. Unfortunately, PEGylated liposomal doxorubicin (sLip/DOX) exhibits serious dose-limiting cutaneous toxicities, which are closely related to the extravascular accumulation of sLip/DOX in the dermis. No clinical interventions have been proposed for cutaneous toxicities due to the elusive transport pathways. Herein, we showed that the reciprocal interaction between liposomes and neutrophils played pivotal roles in liposome extravasation into the dermis. Neutrophils captured liposomes via the complement receptor 3 (CD11b/CD18) recognizing the fragment of complement component C3 (iC3b) deposited on the liposomal surface. Uptake of liposomes also activated neutrophils to induce CD11b upregulation and enhanced the ability of neutrophils to migrate outside the capillaries. Furthermore, inhibition of complement activation either by CRIg-L-FH (a C3b/iC3b targeted complement inhibitor) or blocking the phosphate negative charge in mPEG-DSPE could significantly reduce liposome uptake by neutrophils and alleviate the cutaneous accumulation of liposomes. These results validated the liposome extravasation pathway mediated by neutrophils and provided potential solutions to the devastating cutaneous toxicities occurring during sLip/DOX treatment.
Collapse
Affiliation(s)
- Tianhao Ding
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yang Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Ercan Wu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Qianwen Shao
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
| | - Shiqi Lin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yifei Yu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jun Qian
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
10
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
11
|
Sedghi Aminabad N, Saeedi Y, Adiban J, Nemati M, Shaterabadi D, Najafi F, Rahbarghazi R, Talebi M, Zarebkohan A. Discovery of a Novel Dual Targeting Peptide for Human Glioma: From In Silico Simulation to Acting as Targeting Ligand. Adv Pharm Bull 2024; 14:453-468. [PMID: 39206396 PMCID: PMC11347739 DOI: 10.34172/apb.2024.033] [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: 09/10/2023] [Revised: 01/14/2024] [Accepted: 03/03/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Receptor-mediated transcytosis (RMT) is a more specific, highly efficient, and reliable approach to crossing the blood-brain-barrier (BBB) and releasing the therapeutic cargos into the brain parenchyma. Methods Here, we introduced and characterized a human/mouse-specific novel leptin-derived peptide using in silico, in vitro and in vivo experiments. Results Based on the bioinformatics analysis and molecular dynamics (MD) simulation, a 14 amino acid peptide sequence (LDP 14) was introduced and its interaction with leptin-receptor (ObR) was analyzed in comparison with an well known leptin-derived peptide, Lep 30. MD simulation data revealed a significant stable interaction between ligand binding domains (LBD) of ObR with LDP 14. Analyses demonstrated suitable cellular uptake of LDP 14 alone and its derivatives (LDP 14-modified G4 PAMAM dendrimer and LDP 14-modified G4 PAMAM/pEGFP-N1 plasmid complexes) via ObR, energy and species dependent manner (preferred uptake by human/mouse cell lines compared to rat cell line). Importantly, our findings illustrated that the entry of LDP 14-modified dendrimers in hBCEC-D3 cells not only is not affected by protein corona (PC) formation, as the main reason for diminishing the cellular uptake, but also PC per se can enhance uptake rate. Finally, fluorescein labeled LDP 14-modified G4 PAMAM dendrimers efficiently accumulated in the mice brain with lower biodistribution in other organs, in our in vivo study. Conclusion LDP 14 introduced as a novel and highly efficient ligand, which can be used for drugs/genes delivery to brain tissue in different central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Negar Sedghi Aminabad
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Saeedi
- Department of Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Jamal Adiban
- Ministry of Health and Medical Education, Tehran, Iran
| | - Mahdieh Nemati
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Donya Shaterabadi
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhood Najafi
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Advanced Faculty of Medical Sciences, Tabriz University of Medical, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cell Sciences, Advanced Faculty of Medical Sciences, Tabriz University of Medical, Tabriz, Iran
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Chen S, Wang L, Hu Y, Liu S, Geng L, Li Y. High Drug Capacity of Nano-Levodopa-Liposomes: Preparation, In Vitro Release and Brain-Targeted Research. Appl Biochem Biotechnol 2024; 196:3317-3330. [PMID: 37646886 DOI: 10.1007/s12010-023-04673-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
In this work, nano-levodopa-liposomes (L-dopa-Lip) suspension was prepared by rotary-evaporated film-ultrasonic method, and freeze-drying powders of L-dopa-Lip were also obtained to improve the stability. The products were characterized by TEM, DLS, and TG-DSC, and the phase-transition temperature (Tm) and encapsulation efficiency were calculated. The brain-targeting and in vitro release of the drug was also studied. The results showed that L-dopa-Lip were well-formed spherical vesicles, and the sizes were about 100 nm, and the encapsulation efficiency was higher than 90%. The drug release temperature of L-dopa-Lip was 68 °C, and the in vitro release property and mathematical model were also studied. The brain targeting of L-dopa-Lip in vivo was explored by injecting the gold nanoparticles (AuNPs) labeled L-dopa-Lip (AuNPs-L-dopa-Lip) through the tail vein. ICP-MS and TEM showed that L-dopa-Lip had brain targeting, suggesting the potential treatment of L-dopa-Lip on brain dysfunction. The results of this work might be helpful for designing drug-loaded liposomes for the treatment of central nervous system (CNS) diseases and monitoring their distributions in vivo.
Collapse
Affiliation(s)
- Shenna Chen
- College of Chemistry and Material Science, Hebei Key Laboratory of Organic Functional Molecules, Hebei Normal University, Shijiazhuang, 050024, China
| | - Lin Wang
- College of Chemistry and Material Science, Hebei Key Laboratory of Organic Functional Molecules, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yue Hu
- College of Chemistry and Material Science, Hebei Key Laboratory of Organic Functional Molecules, Hebei Normal University, Shijiazhuang, 050024, China
| | - Sha Liu
- College of Chemistry and Material Science, Hebei Key Laboratory of Organic Functional Molecules, Hebei Normal University, Shijiazhuang, 050024, China
| | - Lina Geng
- College of Chemistry and Material Science, Hebei Key Laboratory of Organic Functional Molecules, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Yayong Li
- Department of Rehabilitation Medicine, Shijiazhuang People's Hospital, Shijiazhuang, 050000, China
| |
Collapse
|
13
|
Jang Y, Cho YS, Kim A, Zhou X, Kim Y, Wan Z, Moon JJ, Park H. CXCR4-Targeted Macrophage-Derived Biomimetic Hybrid Vesicle Nanoplatform for Enhanced Cancer Therapy through Codelivery of Manganese and Doxorubicin. ACS APPLIED MATERIALS & INTERFACES 2024; 16:17129-17144. [PMID: 38533538 PMCID: PMC11057903 DOI: 10.1021/acsami.3c18569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Immune-cell-derived membranes have garnered significant attention as innovative delivery modalities in cancer immunotherapy for their intrinsic immune-modulating functionalities and superior biocompatibilities. Integrating additional parental cell membranes or synthetic lipid vesicles into cellular vesicles can further potentiate their capacities to perform combinatorial pharmacological activities in activating antitumor immunity, thus providing insights into the potential of hybrid cellular vesicles as versatile delivery vehicles for cancer immunotherapy. Here, we have developed a macrophage-membrane-derived hybrid vesicle that has the dual functions of transporting immunotherapeutic drugs and shaping the polarization of tumor-associated macrophages for cancer immunotherapy. The platform combines M1 macrophage-membrane-derived vesicles with CXCR4-binding-peptide-conjugated liposomes loaded with manganese and doxorubicin. The hybrid nanovesicles exhibited remarkable macrophage-targeting capacity through the CXCR4-binding peptide, resulting in enhanced macrophage polarization to the antitumoral M1 phenotype characterized by proinflammatory cytokine release. The manganese/doxorubicin-loaded hybrid vesicles in the CXCR4-expressing tumor cells evoked potent cancer cytotoxicity, immunogenic cell death of tumor cells, and STING activation. Moreover, cotreatment with manganese and doxorubicin promoted dendritic cell maturation, enabling effective tumor growth inhibition. In murine models of CT26 colon carcinoma and 4T1 breast cancer, intravenous administration of the manganese/doxorubicin-loaded hybrid vesicles elicited robust tumor-suppressing activity at a low dosage without adverse systemic effects. Local administration of hybrid nanovesicles also induced an abscessive effect in a bilateral 4T1 tumor model. This study demonstrates a promising biomimetic manganese/doxorubicin-based hybrid nanovesicle platform for effective cancer immunotherapy tailored to the tumor microenvironment, which may offer an innovative approach to combinatorial immunotherapy.
Collapse
Affiliation(s)
- Yeonwoo Jang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Young Seok Cho
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - April Kim
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yujin Kim
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ziye Wan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
14
|
Marques C, Borchard G, Jordan O. Unveiling the challenges of engineered protein corona from the proteins' perspective. Int J Pharm 2024; 654:123987. [PMID: 38467206 DOI: 10.1016/j.ijpharm.2024.123987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
It is well known that protein corona affects the "biological identity" of nanoparticles (NPs), which has been seen as both a challenge and an opportunity. Approaches have moved from avoiding protein adsorption to trying to direct it, taking advantage of the formation of a protein corona to favorably modify the pharmacokinetic parameters of NPs. Although promising, the results obtained with engineered NPs still need to be completely understood. While much effort has been put into understanding how the surface of nanomaterials affects protein absorption, less is known about how proteins can affect corona formation due to their specific physicochemical properties. This review addresses this knowledge gap, examining key protein factors influencing corona formation, highlighting current challenges in studying protein-protein interactions, and discussing future perspectives in the field.
Collapse
Affiliation(s)
- Cintia Marques
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel Servet 1211, Geneva, Switzerland; Section of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet 1211, Geneva, Switzerland.
| | - Gerrit Borchard
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel Servet 1211, Geneva, Switzerland; Section of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet 1211, Geneva, Switzerland
| | - Olivier Jordan
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Rue Michel Servet 1211, Geneva, Switzerland; Section of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet 1211, Geneva, Switzerland
| |
Collapse
|
15
|
Zhang Y, Xiao W, He S, Xia X, Yang W, Yang Z, Hu H, Wang Y, Wang X, Li H, Huang Y, Gao H. Lipid-mediated protein corona regulation with increased apolipoprotein A-I recruitment for glioma targeting. J Control Release 2024; 368:42-51. [PMID: 38365180 DOI: 10.1016/j.jconrel.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Protein corona has long been a source of concern, as it might impair the targeting efficacy of targeted drug delivery systems. However, engineered up-regulating the adsorption of certain functional serum proteins could provide nanoparticles with specific targeting drug delivery capacity. Herein, apolipoprotein A-I absorption increased nanoparticles (SPC-PLGA NPs), composed with the Food and Drug Administration approved intravenously injectable soybean phosphatidylcholine (SPC) and poly (DL-lactide-co-glycolide) (PLGA), were fabricated for enhanced glioma targeting. Due to the high affinity of SPC and apolipoprotein A-I, the percentage of apolipoprotein A-I in the protein corona of SPC-PLGA NPs was 2.19-fold higher than that of nanoparticles without SPC, which made SPC-PLGA NPs have superior glioma targeting ability through binding to scavenger receptor class BI on blood-brain barrier and glioma cells both in vitro and in vivo. SPC-PLGA NPs loaded with paclitaxel could effectively reduce glioma invasion and prolong the survival time of glioma-bearing mice. In conclusion, we provided a good example of the direction of achieving targeting drug delivery based on protein corona regulation.
Collapse
Affiliation(s)
- Yiwei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wei Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Siqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wenqin Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhihang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Haili Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yushan Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaorong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
16
|
Cao X, Liu T, Wang T, Wang X, Xu Z, Zhou L, Tian C, Sun D. De Novo Screening and Mirror Image Isomerization of Linear Peptides Targeting α7 Nicotinic Acetylcholine Receptor. ACS Chem Biol 2024; 19:592-598. [PMID: 38380973 DOI: 10.1021/acschembio.3c00674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
As ligand-gated ion channels, nicotinic acetylcholine receptors (nAChRs) are widely distributed in the central and peripheral nervous systems and are associated with the pathogenesis of various degenerative neurological diseases. Here, we report the results of phage display-based de novo screening of an 11-residue linear peptide (named LKP1794) that targets the α7 nAChR, which is among the most abundant nAChR subtypes in the brain. Moreover, two d-peptides were generated through mirror image and/or primary sequence inverso isomerization (termed DRKP1794 and DKP1794) and displayed improved inhibitory effects (IC50 = 0.86 and 0.35 μM, respectively) on α7 nAChR compared with the parent l-peptide LKP1794 (IC50 = 2.48 μM), which markedly enhanced serum stability. A peptide-based fluorescence probe was developed using proteolytically resistant DKP1794 to specifically image the α7 nAChR in living cells. This work provides a new peptide tool to achieve inhibitory modulation and specifically image the α7 nAChR.
Collapse
Affiliation(s)
- Xiuxiu Cao
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Tianqi Liu
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Tao Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Xudong Wang
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Ziyan Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Li Zhou
- Anhui Provincial Peptide Drug Laboratory, Hefei 230026, P. R. China
| | - Changlin Tian
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
- Anhui Provincial Peptide Drug Laboratory, Hefei 230026, P. R. China
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, P. R. China
- Beijing Life Science Academy, Beijing 102200, P. R. China
| | - Demeng Sun
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, P. R. China
- Anhui Provincial Peptide Drug Laboratory, Hefei 230026, P. R. China
| |
Collapse
|
17
|
Yu YF, Wu EC, Lin SQ, Chu YX, Yang Y, Pan F, Ding TH, Qian J, Jiang K, Zhan CY. Reexamining the effects of drug loading on the in vivo performance of PEGylated liposomal doxorubicin. Acta Pharmacol Sin 2024; 45:646-659. [PMID: 37845342 PMCID: PMC10834505 DOI: 10.1038/s41401-023-01169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023] Open
Abstract
Higher drug loading employed in nanoscale delivery platforms is a goal that researchers have long sought after. But such viewpoint remains controversial because the impacts that nanocarriers bring about on bodies have been seriously overlooked. In the present study we investigated the effects of drug loading on the in vivo performance of PEGylated liposomal doxorubicin (PLD). We prepared PLDs with two different drug loading rates: high drug loading rate, H-Dox, 12.9% w/w Dox/HSPC; low drug loading rate, L-Dox, 2.4% w/w Dox/HSPC (L-Dox had about 5 folds drug carriers of H-Dox at the same Dox dose). The pharmaceutical properties and biological effects of H-Dox and L-Dox were compared in mice, rats or 4T1 subcutaneous tumor-bearing mice. We showed that the lowering of doxorubicin loading did not cause substantial shifts to the pharmaceutical properties of PLDs such as in vitro and in vivo stability (stable), anti-tumor effect (equivalent effective), as well as tissue and cellular distribution. Moreover, it was even more beneficial for mitigating the undesired biological effects caused by PLDs, through prolonging blood circulation and alleviating cutaneous accumulation in the presence of pre-existing anti-PEG Abs due to less opsonins (e.g. IgM and C3) deposition on per particle. Our results warn that the effects of drug loading would be much more convoluted than expected due to the complex intermediation between nanocarriers and bodies, urging independent investigation for each individual delivery platform to facilitate clinical translation and application.
Collapse
Affiliation(s)
- Yi-Fei Yu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Er-Can Wu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Shi-Qi Lin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Yu-Xiu Chu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Feng Pan
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Tian-Hao Ding
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Jun Qian
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China.
| | - Kuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China.
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Chang-You Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China.
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China.
| |
Collapse
|
18
|
Wang J, Xu Y, Zhou Y, Zhang J, Jia J, Jiao P, Liu Y, Su G. Modulating the toxicity of engineered nanoparticles by controlling protein corona formation: Recent advances and future prospects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169590. [PMID: 38154635 DOI: 10.1016/j.scitotenv.2023.169590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
With the rapid development and widespread application of engineered nanoparticles (ENPs), understanding the fundamental interactions between ENPs and biological systems is essential to assess and predict the fate of ENPs in vivo. When ENPs are exposed to complex physiological environments, biomolecules quickly and inevitably adsorb to ENPs to form a biomolecule corona, such as a protein corona (PC). The formed PC has a significant effect on the physicochemical properties of ENPs and gives them a brand new identity in the biological environment, which determines the subsequent ENP-cell/tissue/organ interactions. Controlling the formation of PCs is therefore of utmost importance to accurately predict and optimize the behavior of ENPs within living organisms, as well as ensure the safety of their applications. In this review, we provide an overview of the fundamental aspects of the PC, including the formation mechanism, composition, and frequently used characterization techniques. We comprehensively discuss the potential impact of the PC on ENP toxicity, including cytotoxicity, immune response, and so on. Additionally, we summarize recent advancements in manipulating PC formation on ENPs to achieve the desired biological outcomes. We further discuss the challenges and prospects, aiming to provide valuable insights for a better understanding and prediction of ENP behaviors in vivo, as well as the development of low-toxicity ENPs.
Collapse
Affiliation(s)
- Jiali Wang
- School of Pharmacy, Nantong University, Nantong 226019, China; School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Yuhang Xu
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Yun Zhou
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Jian Zhang
- Digestive Diseases Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 510001, China; Center for Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 510001 Guangzhou, China
| | - Jianbo Jia
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Peifu Jiao
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan 250200, China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong 226019, China.
| |
Collapse
|
19
|
Xun Z, Li T, Xue X. The application strategy of liposomes in organ targeting therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1955. [PMID: 38613219 DOI: 10.1002/wnan.1955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 04/14/2024]
Abstract
Liposomes-microscopic phospholipid bubbles with bilayered membrane structure-have been a focal point in drug delivery research for the past 30 years. Current liposomes possess a blend of biocompatibility, drug loading efficiency, prolonged circulation and targeted delivery. Tailored liposomes, varying in size, charge, lipid composition, and ratio, have been developed to address diseases in specific organs, thereby enhancing drug circulation, accumulation at lesion sites, intracellular delivery, and treatment efficacy for various organ-specific diseases. For further successful development of this field, this review summarized liposomal strategies for targeting different organs in series of major human diseases, including widely studied cardiovascular diseases, liver and spleen immune diseases, chronic or acute kidney injury, neurodegenerative diseases, and organ-specific tumors. It highlights recent advances of liposome-mediated therapeutic agent delivery for disease intervention and organ rehabilitation, offering practical guidelines for designing organ-targeted liposomes. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Zengyu Xun
- State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, People's Republic of China
| | - Tianqi Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, People's Republic of China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical Biology, College of Life Science, Nankai University, Tianjin, People's Republic of China
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
20
|
Mondal S, Ghosh S. Liposome-Mediated Anti-Viral Drug Delivery Across Blood-Brain Barrier: Can Lipid Droplet Target Be Game Changers? Cell Mol Neurobiol 2023; 44:9. [PMID: 38123863 DOI: 10.1007/s10571-023-01443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Lipid droplets (LDs) are subcellular organelles secreted from the endoplasmic reticulum (ER) that play a major role in lipid homeostasis. Recent research elucidates additional roles of LDs in cellular bioenergetics and innate immunity. LDs activate signaling cascades for interferon response and secretion of pro-inflammatory cytokines. Since balanced lipid homeostasis is critical for neuronal health, LDs play a crucial role in neurodegenerative diseases. RNA viruses enhance the secretion of LDs to support various phases of their life cycle in neurons which further leads to neurodegeneration. Targeting the excess LD formation in the brain could give us a new arsenal of antiviral therapeutics against neuroviruses. Liposomes are a suitable drug delivery system that could be used for drug delivery in the brain by crossing the Blood-Brain Barrier. Utilizing this, various pharmacological inhibitors and non-coding RNAs can be delivered that could inhibit the biogenesis of LDs or reduce their sizes, reversing the excess lipid-related imbalance in neurons. Liposome-Mediated Antiviral Drug Delivery Across Blood-Brain Barrier. Developing effective antiviral drug is challenging and it doubles against neuroviruses that needs delivery across the Blood-Brain Barrier (BBB). Lipid Droplets (LDs) are interesting targets for developing antivirals, hence targeting LD formation by drugs delivered using Liposomes can be game changers.
Collapse
Affiliation(s)
- Sourav Mondal
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sourish Ghosh
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
21
|
Rad ME, Soylukan C, Kulabhusan PK, Günaydın BN, Yüce M. Material and Design Toolkit for Drug Delivery: State of the Art, Trends, and Challenges. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55201-55231. [PMID: 37994836 DOI: 10.1021/acsami.3c10065] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
The nanomaterial and related toolkit have promising applications for improving human health and well-being. Nanobased drug delivery systems use nanoscale materials as carriers to deliver therapeutic agents in a targeted and controlled manner, and they have shown potential to address issues associated with conventional drug delivery systems. They offer benefits for treating various illnesses by encapsulating or conjugating biological agents, chemotherapeutic drugs, and immunotherapeutic agents. The potential applications of this technology are vast; however, significant challenges exist to overcome such as safety issues, toxicity, efficacy, and insufficient capacity. This article discusses the latest developments in drug delivery systems, including drug release mechanisms, material toolkits, related design molecules, and parameters. The concluding section examines the limitations and provides insights into future possibilities.
Collapse
Affiliation(s)
- Monireh Esmaeili Rad
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Caner Soylukan
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | | | - Beyza Nur Günaydın
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| | - Meral Yüce
- SUNUM Nanotechnology Research and Application Centre, Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
22
|
Liu H, Lv H, Duan X, Du Y, Tang Y, Xu W. Advancements in Macrophage-Targeted Drug Delivery for Effective Disease Management. Int J Nanomedicine 2023; 18:6915-6940. [PMID: 38026516 PMCID: PMC10680479 DOI: 10.2147/ijn.s430877] [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: 08/09/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophages play a crucial role in tissue homeostasis and the innate immune system. They perform essential functions such as presenting antigens, regulating cytokines, and responding to inflammation. However, in diseases like cancer, cardiovascular disorders, and autoimmune conditions, macrophages undergo aberrant polarization, which disrupts tissue regulation and impairs their normal behavior. To address these challenges, there has been growing interest in developing customized targeted drug delivery systems specifically designed for macrophage-related functions in different anatomical locations. Nanomedicine, utilizing nanoscale drug systems, offers numerous advantages including improved stability, enhanced pharmacokinetics, controlled release kinetics, and precise temporal drug delivery. These advantages hold significant promise in achieving heightened therapeutic efficacy, specificity, and reduced side effects in drug delivery and treatment approaches. This review aims to explore the roles of macrophages in major diseases and present an overview of current strategies employed in targeted drug delivery to macrophages. Additionally, this article critically evaluates the design of macrophage-targeted delivery systems, highlighting limitations and discussing prospects in this rapidly evolving field. By assessing the strengths and weaknesses of existing approaches, we can identify areas for improvement and refinement in macrophage-targeted drug delivery.
Collapse
Affiliation(s)
- Hanxiao Liu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, 261053, People’s Republic of China
- Department of Pharmacy, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, People’s Republic of China
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| | - Hui Lv
- Department of Pharmacy, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, People’s Republic of China
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| | - Xuehui Duan
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| | - Yan Du
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| | - Yixuan Tang
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| | - Wei Xu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, 261053, People’s Republic of China
- Department of Pharmacy, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, People’s Republic of China
- School of Pharmaceutical Sciences & Institute of Materia Medica, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Key Laboratory for Biotechnology Drugs of National Health Commission, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| |
Collapse
|
23
|
Wang Z, Wang X, Xu W, Li Y, Lai R, Qiu X, Chen X, Chen Z, Mi B, Wu M, Wang J. Translational Challenges and Prospective Solutions in the Implementation of Biomimetic Delivery Systems. Pharmaceutics 2023; 15:2623. [PMID: 38004601 PMCID: PMC10674763 DOI: 10.3390/pharmaceutics15112623] [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: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Biomimetic delivery systems (BDSs), inspired by the intricate designs of biological systems, have emerged as a groundbreaking paradigm in nanomedicine, offering unparalleled advantages in therapeutic delivery. These systems, encompassing platforms such as liposomes, protein-based nanoparticles, extracellular vesicles, and polysaccharides, are lauded for their targeted delivery, minimized side effects, and enhanced therapeutic outcomes. However, the translation of BDSs from research settings to clinical applications is fraught with challenges, including reproducibility concerns, physiological stability, and rigorous efficacy and safety evaluations. Furthermore, the innovative nature of BDSs demands the reevaluation and evolution of existing regulatory and ethical frameworks. This review provides an overview of BDSs and delves into the multifaceted translational challenges and present emerging solutions, underscored by real-world case studies. Emphasizing the potential of BDSs to redefine healthcare, we advocate for sustained interdisciplinary collaboration and research. As our understanding of biological systems deepens, the future of BDSs in clinical translation appears promising, with a focus on personalized medicine and refined patient-specific delivery systems.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; (Z.W.); (R.L.)
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Wanting Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Yongxiao Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Ruizhi Lai
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; (Z.W.); (R.L.)
| | - Xiaohui Qiu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Zhidong Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Meiying Wu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (X.W.); (W.X.); (Y.L.); (X.Q.); (X.C.); (Z.C.)
| |
Collapse
|
24
|
Jiang K, Yu Y, Qiu W, Tian K, Guo Z, Qian J, Lu H, Zhan C. Protein corona on brain targeted nanocarriers: Challenges and prospects. Adv Drug Deliv Rev 2023; 202:115114. [PMID: 37827336 DOI: 10.1016/j.addr.2023.115114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
Safe and efficient medical therapy for brain diseases is still an unmet clinical need due to various barriers represented by the blood-brain barrier. Well-designed brain targeted nanocarriers are potential solutions for enhanced brain drug delivery; however, the complicated in vivo process attenuates performance of nanocarriers, which severely hampers clinical translation. The formation of protein corona (PC) is inevitable for nanocarriers circulation and transport in biofluids, acting as an important factor to regulate in vivo performance of nanocarriers. In this review, the reported strategies have been retrospected for better understanding current situation in developing brain targeted nanocarriers. The interplay between brain targeted nanocarriers and plasma proteins is emphasized to comprehend how the nanocarriers adsorb proteins by certain synthetic identity, and following regulations on in vivo performance of nanocarriers. More importantly, the mainstream methods to promote efficiency of nanocarriers by regulating PC, defined as in vitro functionalization and in vivo functionalization strategies, are also discussed. Finally, viewpoints about future development of brain targeted nanocarriers according to the understanding on nanocarriers-PC interaction are proposed.
Collapse
Affiliation(s)
- Kuan Jiang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Yifei Yu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Wei Qiu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Kaisong Tian
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Zhiwei Guo
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China
| | - Jun Qian
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201399, PR China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201399, PR China.
| | - Changyou Zhan
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, PR China; Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201399, PR China.
| |
Collapse
|
25
|
Liu Q, Zou J, Chen Z, He W, Wu W. Current research trends of nanomedicines. Acta Pharm Sin B 2023; 13:4391-4416. [PMID: 37969727 PMCID: PMC10638504 DOI: 10.1016/j.apsb.2023.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 11/17/2023] Open
Abstract
Owing to the inherent shortcomings of traditional therapeutic drugs in terms of inadequate therapeutic efficacy and toxicity in clinical treatment, nanomedicine designs have received widespread attention with significantly improved efficacy and reduced non-target side effects. Nanomedicines hold tremendous theranostic potential for treating, monitoring, diagnosing, and controlling various diseases and are attracting an unfathomable amount of input of research resources. Against the backdrop of an exponentially growing number of publications, it is imperative to help the audience get a panorama image of the research activities in the field of nanomedicines. Herein, this review elaborates on the development trends of nanomedicines, emerging nanocarriers, in vivo fate and safety of nanomedicines, and their extensive applications. Moreover, the potential challenges and the obstacles hindering the clinical translation of nanomedicines are also discussed. The elaboration on various aspects of the research trends of nanomedicines may help enlighten the readers and set the route for future endeavors.
Collapse
Affiliation(s)
- Qiuyue Liu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Wu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
- Fudan Zhangjiang Institute, Shanghai 201203, China
| |
Collapse
|
26
|
Li L, Zhang M, Li J, Liu T, Bao Q, Li X, Long J, Fu L, Zhang Z, Huang S, Liu Z, Zhang L. Cholesterol removal improves performance of a model biomimetic system to co-deliver a photothermal agent and a STING agonist for cancer immunotherapy. Nat Commun 2023; 14:5111. [PMID: 37607938 PMCID: PMC10444796 DOI: 10.1038/s41467-023-40814-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023] Open
Abstract
Biological membranes often play important functional roles in biomimetic drug delivery systems. We discover that the circulation time and targeting capability of biological membrane coated nanovehicles can be significantly improved by reducing cholesterol level in the coating membrane. A proof-of-concept system using cholesterol-reduced and PD-1-overexpressed T cell membrane to deliver a photothermal agent and a STING agonist is thus fabricated. Comparing with normal membrane, this engineered membrane increases tumor accumulation by ~2-fold. In a melanoma model in male mice, tumors are eliminated with no recurrence in >80% mice after intravenous injection and laser irradiation; while in a colon cancer model in male mice, ~40% mice are cured without laser irradiation. Data suggest that the engineered membranes escape immune surveillance to avoid blood clearance while keeping functional surface molecules exposed. In summary, we develop a simple, effective, safe and widely-applicable biological membrane modification strategy. This "subtractive" strategy displays some advantages and is worth further development.
Collapse
Affiliation(s)
- Lin Li
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengxing Zhang
- Med-X center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Jing Li
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Tiantian Liu
- Med-X center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Qixue Bao
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Xi Li
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiaying Long
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Leyao Fu
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Shiqi Huang
- Med-X center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhenmi Liu
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ling Zhang
- Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
- Med-X center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
27
|
Wang H, Lin S, Wu X, Jiang K, Lu H, Zhan C. Interplay between Liposomes and IgM: Principles, Challenges, and Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301777. [PMID: 37150860 PMCID: PMC10369250 DOI: 10.1002/advs.202301777] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/14/2023] [Indexed: 05/09/2023]
Abstract
Liposomes have received tremendous attention as a class of versatile pharmaceutical vehicles of great potential over the past several decades. However, the application of liposomes encounters major challenges due to the knowledge gaps in their in vivo delivery process. Immunoglobulin M (IgM) displays both pervasiveness and complexity in regulating the biological functions as well as eliciting adverse effects of liposomes. Understanding, mitigating, and exploiting the duality of IgM are prerequisites for achieving various biomedical applications of liposomes. In this review, the intricate relationship between liposomes and their biological environments has been summarized, with an emphasis on the regulatory effects of IgM on in vivo performance of liposomes. Corresponding solutions have also been discussed to evade IgM-mediated opsonization for safe and efficient drug delivery.
Collapse
Affiliation(s)
- Huan Wang
- School of PharmacyNaval Medical UniversityShanghai200433P. R. China
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Shiqi Lin
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Xiying Wu
- Shanghai Skin Disease HospitalTongji University School of MedicineShanghai200443China
| | - Kuan Jiang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Huiping Lu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of PharmacologySchool of Basic Medical SciencesFudan UniversityShanghai201399P. R. China
- Shanghai Engineering Research Center for Synthetic ImmunologyFudan UniversityShanghai200032P. R. China
| |
Collapse
|
28
|
Han H, Jung JH, Lee H, Lee J, Jang SH, Goh U, Yoon J, Park JH. Targeted Fusogenic Liposomes for Effective Tumor Delivery and Penetration of Lipophilic Cargoes. ACS Biomater Sci Eng 2023; 9:1919-1927. [PMID: 36921244 DOI: 10.1021/acsbiomaterials.2c01490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Nanoparticle-based drug delivery has been widely used for effective anticancer treatment. However, a key challenge restricting the efficacy of nanotherapeutics is limited tissue penetration within solid tumors. Here, we report a targeted fusogenic liposome (TFL) that can selectively deliver lipophilic cargo to the plasma membranes of tumor cells. TFL is prepared by directly attaching tumor-targeting peptides to the surface of FL instead of the cationic moieties. The lipophilic cargo loaded in the membrane of TFL is transferred to the plasma membranes of tumor cells and subsequently packaged in the extracellular vesicles (EVs) released by the cells. Systemically administered TFL accumulates in the perivascular region of tumors, where the lipophilic cargo is unloaded to the tumor cell membranes and distributed autonomously throughout the tumor tissue via extracellular vesicle-mediated intercellular transfer. When loaded with a lipophilic pro-apoptotic drug, thapsigargin (Tg), TFL significantly inhibits tumor growth in a mouse colorectal cancer model. Furthermore, the combination treatment with TFL (Tg) potentiates the antitumor efficacy of FDA-approved liposomal doxorubicin, whose therapeutic effect is limited to perivascular regions without significant toxicity.
Collapse
Affiliation(s)
- Hyeonjeong Han
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jik-Han Jung
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyoungjin Lee
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junsung Lee
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seong-Hoon Jang
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Unbyeol Goh
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jooeun Yoon
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
29
|
Sepasi T, Ghadiri T, Ebrahimi-Kalan A, Bani F, Talebi M, Rahbarghazi R, Khodakarimi S, Beyrampour-Basmenj H, Seidi K, Abbaspour-Ravasjani S, Sadeghi MR, Zarebkohan A, Gao H. CDX-modified chitosan nanoparticles remarkably reduce therapeutic dose of fingolimod in the EAE model of mice. Int J Pharm 2023; 636:122815. [PMID: 36907279 DOI: 10.1016/j.ijpharm.2023.122815] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/20/2023] [Accepted: 03/05/2023] [Indexed: 03/13/2023]
Abstract
Fingolimod (Fin), an FDA-approved drug, is used to control relapsing-remitting multiple sclerosis (MS). This therapeutic agent faces crucial drawbacks like poor bioavailability rate, risk of cardiotoxicity, potent immunosuppressive effects, and high cost. Here, we aimed to assess the therapeutic efficacy of nano-formulated Fin in a mouse model of experimental autoimmune encephalomyelitis (EAE). Results showed the suitability of the present protocol in the synthesis of Fin-loaded CDX-modified chitosan (CS) nanoparticles (NPs) (Fin@CSCDX) with suitable physicochemical features. Confocal microscopy confirmed the appropriate accumulation of synthesized NPs within the brain parenchyma. Compared to the control EAE mice, INF-γ levels were significantly reduced in the group that received Fin@CSCDX (p < 0.05). Along with these data, Fin@CSCDX reduced the expression of TBX21, GATA3, FOXP3, and Rorc associated with the auto-reactivation of T cells (p < 0.05). Histological examination indicated a low-rate lymphocyte infiltration into the spinal cord parenchyma after the administration of Fin@CSCDX. Of note, HPLC data revealed that the concentration of nano-formulated Fin was about 15-fold less than Fin therapeutic doses (TD) with similar reparative effects. Neurological scores were similar in both groups that received nano-formulated fingolimod 1/15th of free Fin therapeutic amounts. Fluorescence imaging indicated that macrophages and especially microglia can efficiently uptake Fin@CSCDX NPs, leading to the regulation of pro-inflammatory responses. Taken together, current results indicated that CDX-modified CS NPs provide a suitable platform not only for the efficient reduction of Fin TD but also these NPs can target the brain immune cells during neurodegenerative disorders.
Collapse
Affiliation(s)
- Tina Sepasi
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tahereh Ghadiri
- Department of Neuroscience and Cognitive, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neuroscience and Cognitive, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Bani
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Khodakarimi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Neuroscience and Cognitive, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Beyrampour-Basmenj
- Department of Medical Biotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khaled Seidi
- Polymer Research Laboratory, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | | | - Mohammad-Reza Sadeghi
- Department of Medical Biotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Advanced Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
30
|
Chota A, George BP, Abrahamse H. Recent Advances in Green Metallic Nanoparticles for Enhanced Drug Delivery in Photodynamic Therapy: A Therapeutic Approach. Int J Mol Sci 2023; 24:4808. [PMID: 36902238 PMCID: PMC10003542 DOI: 10.3390/ijms24054808] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Globally, cancer is one of the leading causes of death among men and women, it is characterized by the unregulated proliferation of tumor cells. Some of the common risk factors associated with cancer development include the consistent exposure of body cells to carcinogenic agents such as alcohol, tobacco, toxins, gamma rays and alpha particles. Besides the above-mentioned risk factors, conventional therapies such as radiotherapy, and chemotherapy have also been linked to the development of cancer. Over the past decade, tremendous efforts have been invested in the synthesis of eco-friendly green metallic nanoparticles (NPs), and their medical application. Comparatively, metallic NPs have greater advantages over conventional therapies. Additionally, metallic NPs can be functionalized with different targeting moieties e.g., liposomes, antibodies, folic acid, transferrin, and carbohydrates. Herein, we review and discuss the synthesis, and therapeutic potential of green synthesized metallic NPs for enhanced cancer photodynamic therapy (PDT). Finally, the advantages of green hybridized activatable NPs over conventional photosensitizers (PSs) and the future perspectives of nanotechnology in cancer research are discussed in the review. Furthermore, we anticipate that the insights offered in this review will inspire the design and development of green nano-formulations for enhanced image-guided PDT in cancer treatment.
Collapse
Affiliation(s)
| | - Blassan P. George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Johannesburg 2028, South Africa
| | | |
Collapse
|
31
|
Zhao F, Li R, Liu Y, Chen H. Perspectives on lecithin from egg yolk: Extraction, physicochemical properties, modification, and applications. Front Nutr 2023; 9:1082671. [PMID: 36687715 PMCID: PMC9853391 DOI: 10.3389/fnut.2022.1082671] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Egg yolk lecithin has physiological activities as an antioxidant, antibacterial, anti-inflammatory, and neurologic, cardiovascular, and cerebrovascular protectant. There are several methods for extracting egg yolk lecithin, including solvent extraction and supercritical extraction. However, changes in extraction methods and functional activity of egg yolk lecithin are a matter of debate. In this review we summarized the molecular structure, extraction method, and functional activity of egg yolk lecithin to provide a good reference for the development of egg yolk lecithin products in the future.
Collapse
Affiliation(s)
- Feng Zhao
- College of Food Science and Engineering, Jilin Agriculture University, Changchun, Jilin, China
| | - Rongji Li
- College of Food Science and Engineering, Jilin Agriculture University, Changchun, Jilin, China
| | - Yun Liu
- College of Life Sciences, Beijing University of Chemical Technology, Beijing, China
| | - Haiyan Chen
- College of Food Science and Engineering, Changchun Sci-Tech University, Changchun, Jilin, China,College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, China,*Correspondence: Haiyan Chen ✉
| |
Collapse
|
32
|
Zahednezhad F, Zakeri-Milani P, Mojarrad JS, Sarfraz M, Mahmoudian M, Baradaran B, Valizadeh H. Acetyl carnitine modified liposomes elevate cisplatin uptake in macrophage and cancer cells. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
33
|
Cheng D, Wen Z, Chen H, Lin S, Zhang W, Tang X, Wu W. Hepatocyte-targeting and tumor microenvironment-responsive liposomes for enhanced anti-hepatocarcinoma efficacy. Drug Deliv 2022; 29:2995-3008. [PMID: 36104946 PMCID: PMC9487930 DOI: 10.1080/10717544.2022.2122635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
To increase the antitumor drug concentration in the liver tumor site and improve the therapeutic effects, a functionalized liposome (PPP-LIP) with tumor targetability and enhanced internalization after matrix metalloproteinase-2 (MMP2)-triggered cell-penetrating peptide (TATp) exposure was modified with myrcludex B (a synthetic HBV preS-derived lipopeptide endowed with compelling liver tropism) for liver tumor-specific delivery. After intravenous administration, PPP-LIP was mediated by myrcludex B to reach the hepatocyte surface. The MMP2-overexpressing tumor microenvironment deprotected PEG, exposing it to TATp, facilitating tumor penetration and subsequent efficient destruction of tumor cells. In live imaging of small animals and cellular uptake, PPP-LIP was taken up much more than typical unmodified liposomes in the ICR mouse liver and liver tumor cells. Hydroxycamptothecin (HCPT)-loaded PPP-LIP showed a better antitumor effect than commercially available HCPT injections among MTT, three-dimensional (3 D) tumor ball, and tumor-bearing nude mouse experiments. Our findings indicated that PPP-LIP nanocarriers could be a promising tumor-targeted medication delivery strategy for treating liver cancers with elevated MMP2 expression.
Collapse
Affiliation(s)
- Dongliang Cheng
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Hui Chen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Shiyuan Lin
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Xin Tang
- School of Public Health, Guilin Medical University, Guilin, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin, China
| |
Collapse
|
34
|
Xu W, Ye C, Qing X, Liu S, Lv X, Wang W, Dong X, Zhang Y. Multi-target tyrosine kinase inhibitor nanoparticle delivery systems for cancer therapy. Mater Today Bio 2022; 16:100358. [PMID: 35880099 PMCID: PMC9307458 DOI: 10.1016/j.mtbio.2022.100358] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
Multi-target Tyrosine Kinase Inhibitors (MTKIs) have drawn substantial attention in tumor therapy. MTKIs could inhibit tumor cell proliferation and induce apoptosis by blocking the activity of tyrosine kinase. However, the toxicity and drug resistance of MTKIs severely restrict their further clinical application. The nano pharmaceutical technology based on MTKIs has attracted ever-increasing attention in recent years. Researchers deliver MTKIs through various types of nanocarriers to overcome drug resistance and improve considerably therapeutic efficiency. This review intends to summarize comprehensive applications of MTKIs nanoparticles in malignant tumor treatment. Firstly, the mechanism and toxicity were introduced. Secondly, various nanocarriers for MTKIs delivery were outlined. Thirdly, the combination treatment schemes and drug resistance reversal strategies were emphasized to improve the outcomes of cancer therapy. Finally, conclusions and perspectives were summarized to guide future research.
Collapse
Affiliation(s)
- Wenjing Xu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chunping Ye
- Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Qing
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shengli Liu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| | - Xinyi Lv
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Wenjun Wang
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng, 252059, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China
| |
Collapse
|
35
|
Youden B, Wang F, Zhang X, Curry D, Majtenyi N, Shaaer A, Bingham K, Nguyen Q, Bragg L, Liu J, Servos M, Zhang X, Jiang R. Degradable Multifunctional Gold-Liposomes as an All-in-One Theranostic Platform for Image-Guided Radiotherapy. Int J Pharm 2022; 629:122413. [DOI: 10.1016/j.ijpharm.2022.122413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/07/2022] [Accepted: 11/13/2022] [Indexed: 11/23/2022]
|
36
|
Tian Y, Gao Z, Wang N, Hu M, Ju Y, Li Q, Caruso F, Hao J, Cui J. Engineering Poly(ethylene glycol) Nanoparticles for Accelerated Blood Clearance Inhibition and Targeted Drug Delivery. J Am Chem Soc 2022; 144:18419-18428. [PMID: 36166420 DOI: 10.1021/jacs.2c06877] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Surface modification with poly(ethylene glycol) (PEGylation) is an effective strategy to improve the colloidal stability of nanoparticles (NPs) and is often used to minimize cellular uptake and clearance of NPs by the immune system. However, PEGylation can also trigger the accelerated blood clearance (ABC) phenomenon, which is known to reduce the circulation time of PEGylated NPs. Herein, we report the engineering of stealth PEG NPs that can avoid the ABC phenomenon and, when modified with hyaluronic acid (HA), show specific cancer cell targeting and drug delivery. PEG NPs cross-linked with disulfide bonds are prepared by using zeolitic imidazolate framework-8 NPs as templates. The reported templating strategy enables the simultaneous removal of the template and formation of PEG NPs under mild conditions (pH 5.5 buffer). Compared to PEGylated liposomes, PEG NPs avoid the secretion of anti-PEG antibodies and the presence of anti-PEG IgM and IgG did not significantly accelerate the blood clearance of PEG NPs, indicating the inhibition of the ABC effect for the PEG NPs. Functionalization of the PEG NPs with HA affords PEG NPs that retain their stealth properties against macrophages, target CD44-expressed cancer cells and, when loaded with the anticancer drug doxorubicin, effectively inhibit tumor growth. The innovation of this study lies in the engineering of PEG NPs that can circumvent the ABC phenomenon and that can be functionalized for the improved and targeted delivery of drugs.
Collapse
Affiliation(s)
- Yuan Tian
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Zhiliang Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Ning Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Ming Hu
- School of Physics and Electronic Science, East China Normal University, Shanghai 200241, China
| | - Yi Ju
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.,Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Qiang Li
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China.,State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
37
|
Harnessing Protein Corona for Biomimetic Nanomedicine Design. Biomimetics (Basel) 2022; 7:biomimetics7030126. [PMID: 36134930 PMCID: PMC9496170 DOI: 10.3390/biomimetics7030126] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are usually treated as multifunctional agents combining several therapeutical applications, like imaging and targeting delivery. However, clinical translation is still largely hindered by several factors, and the rapidly formed protein corona on the surface of NPs is one of them. The formation of protein corona is complicated and irreversible in the biological environment, and protein corona will redefine the “biological identity” of NPs, which will alter the following biological events and therapeutic efficacy. Current understanding of protein corona is still limited and incomplete, and in many cases, protein corona has adverse impacts on nanomedicine, for instance, losing targeting ability, activating the immune response, and rapid clearance. Due to the considerable role of protein corona in NPs’ biological fate, harnessing protein corona to achieve some therapeutic effects through various methods like biomimetic approaches is now treated as a promising way to meet the current challenges in nanomedicine such as poor pharmacokinetic properties, off-target effect, and immunogenicity. This review will first introduce the current understanding of protein corona and summarize the investigation process and technologies. Second, the strategies of harnessing protein corona with biomimetic approaches for nanomedicine design are reviewed. Finally, we discuss the challenges and future outlooks of biomimetic approaches to tune protein corona in nanomedicine.
Collapse
|
38
|
Wang H, Lin S, Wang S, Jiang Z, Ding T, Wei X, Lu Y, Yang F, Zhan C. Folic Acid Enables Targeting Delivery of Lipodiscs by Circumventing IgM-Mediated Opsonization. NANO LETTERS 2022; 22:6516-6522. [PMID: 35943299 DOI: 10.1021/acs.nanolett.2c01509] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Folic acid (FA) is one of the most widely utilized small-molecule ligands for cancer targeted drug delivery. Natural IgM was recently found to avidly absorb on the surface of FA-functionalized liposomes (FA-sLip), negatively regulating the in vivo performance by efficiently activating complement. Herein, FA-functionalized lipodiscs (FA-Disc) were constructed to successfully circumvent IgM-mediated opsonization and retained binding activity with folate receptors in vivo. The FA moiety along with the bound IgM was restricted to the highly curved rim of lipodiscs, leading to IgM incapability of presenting the membrane-bound conformation to trigger complement activation. The C1q docking, C3 binding, and C5a release were blocked and accelerated blood clearance phenomenon was mitigated of FA-Disc. FA-Disc retained folate binding activity and could effectively target folate receptor positive tumors in vivo. The present study provides a useful solution to avoid the negative regulation by IgM and achieve FA-enabled targeting by exploring disc-shaped nanocarriers.
Collapse
Affiliation(s)
- Huan Wang
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Shiqi Lin
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Songli Wang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Zhuxuan Jiang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Tianhao Ding
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Xiaoli Wei
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| | - Ying Lu
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
| | - Feng Yang
- School of Pharmacy, Naval Medical University, Shanghai 200433, P.R. China
| | - Changyou Zhan
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University Shanghai Engineering Research Center for Synthetic Immunology, Shanghai 201399, P.R. China
| |
Collapse
|
39
|
Wang X, Li C, Wang Y, Chen H, Zhang X, Luo C, Zhou W, Li L, Teng L, Yu H, Wang J. Smart drug delivery systems for precise cancer therapy. Acta Pharm Sin B 2022; 12:4098-4121. [DOI: 10.1016/j.apsb.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
|
40
|
Xiao Q, Zoulikha M, Qiu M, Teng C, Lin C, Li X, Sallam MA, Xu Q, He W. The effects of protein corona on in vivo fate of nanocarriers. Adv Drug Deliv Rev 2022; 186:114356. [PMID: 35595022 DOI: 10.1016/j.addr.2022.114356] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
With the emerging advances in utilizing nanocarriers for biomedical applications, a molecular-level understanding of the in vivo fate of nanocarriers is necessary. After administration into human fluids, nanocarriers can attract proteins onto their surfaces, forming an assembled adsorption layer called protein corona (PC). The formed PC can influence the physicochemical properties and subsequently determine nanocarriers' biological behaviors. Therefore, an in-depth understanding of the features and effects of the PC on the nanocarriers' surface is the first and most important step towards controlling their in vivo fate. This review introduces fundamental knowledge such as the definition, formation, composition, conformation, and characterization of the PC, emphasizing the in vivo environmental factors that control the PC formation. The effect of PC on the physicochemical properties and thus biological behaviors of nanocarriers was then presented and thoroughly discussed. Finally, we proposed the design strategies available for engineering PC onto nanocarriers to manipulate them with the desired surface properties and achieve the best biomedical outcomes.
Collapse
|
41
|
Taghipour YD, Zarebkohan A, Salehi R, Rahimi F, Torchilin VP, Hamblin MR, Seifalian A. An update on dual targeting strategy for cancer treatment. J Control Release 2022; 349:67-96. [PMID: 35779656 DOI: 10.1016/j.jconrel.2022.06.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/04/2022] [Accepted: 06/24/2022] [Indexed: 12/18/2022]
Abstract
The key issue in the treatment of solid tumors is the lack of efficient strategies for the targeted delivery and accumulation of therapeutic cargoes in the tumor microenvironment (TME). Targeting approaches are designed for more efficient delivery of therapeutic agents to cancer cells while minimizing drug toxicity to normal cells and off-targeting effects, while maximizing the eradication of cancer cells. The highly complicated interrelationship between the physicochemical properties of nanoparticles, and the physiological and pathological barriers that are required to cross, dictates the need for the success of targeting strategies. Dual targeting is an approach that uses both purely biological strategies and physicochemical responsive smart delivery strategies to increase the accumulation of nanoparticles within the TME and improve targeting efficiency towards cancer cells. In both approaches, either one single ligand is used for targeting a single receptor on different cells, or two different ligands for targeting two different receptors on the same or different cells. Smart delivery strategies are able to respond to triggers that are typical of specific disease sites, such as pH, certain specific enzymes, or redox conditions. These strategies are expected to lead to more precise targeting and better accumulation of nano-therapeutics. This review describes the classification and principles of dual targeting approaches and critically reviews the efficiency of dual targeting strategies, and the rationale behind the choice of ligands. We focus on new approaches for smart drug delivery in which synthetic and/or biological moieties are attached to nanoparticles by TME-specific responsive linkers and advanced camouflaged nanoparticles.
Collapse
Affiliation(s)
- Yasamin Davatgaran Taghipour
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Fariborz Rahimi
- Department of Electrical Engineering, University of Bonab, Bonab, Iran
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine and Department of Chemical Engineering, Northeastern University, Boston, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Laser Research Centre, Faculty of Health Science, University of Johannesburg, South Africa
| | - Alexander Seifalian
- Nanotechnology & Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
42
|
A Narrative Review of the Potential Roles of Lipid-Based Vesicles (Vesiculosomes) in Burn Management. Sci Pharm 2022. [DOI: 10.3390/scipharm90030039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Burn injuries can have a lasting effect on people’s quality of life, as they negatively impact their physical and mental health. Then, they are likely to suffer psychological problems as a result. A serious problem is that deep burns are more challenging to treat due to their slow healing rate and susceptibility to microbial infection. Conventional topical medications used for burn treatment are sometimes ineffective because they cannot optimize their ability of transcutaneous absorption at the targeted site and accelerate healing. However, nanotechnology offers excellent prospects for developing current medical wound therapies and is capable of addressing issues such as low drug stability, water solubility, permeability, and bioavailability. The current review focuses on lipid-based vesicles (vesiculosomes) as an example of advanced delivery systems, showing their potential clinical applications in burn wound management. Vesiculosomes may help overcome impediments including the low bioavailability of active agents, offering the controlled release of drugs, increased drug stability, fewer side effects, and reduced dosing frequency, which will ultimately improve therapeutic efficacy and patient compliance. We discuss the application of various types of vesiculosomes such as liposomes, niosomes, ethosomes, cubosomes, transfersomes, and phytosomes in burn healing therapy, as these demonstrate superior skin penetration compared to conventional burn topical treatment. We also highlight their noteworthy uses in the formulation of natural products and discuss the current status as well as future perspectives of these carriers in burn management. Furthermore, the burn treatment options currently available in the market are also summarized.
Collapse
|
43
|
Jiang Z, Chu Y, Zhan C. Protein corona: challenges and opportunities for targeted delivery of nanomedicines. Expert Opin Drug Deliv 2022; 19:833-846. [PMID: 35738018 DOI: 10.1080/17425247.2022.2093854] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Targeted drug delivery has been widely explored as a promising way to improve the performance of nanomedicines. However, protein corona formed on the nano-surface represents a major issue that has great impacts on the in vivo fate of targeting nanomedicines, which has been overlooked in the past. With the increasing understanding of protein corona in the recent decade, many efforts have been made to improve targeting efficacy. AREAS COVERED In this review, we briefly summarize insights of targeted delivery systems inspired by protein corona, and discuss the promising strategies to regulate protein corona for better targeting. EXPERT OPINION The interaction between nanomedicines and endogenous proteins brings great uncertainty and challenges, but it also provides great opportunities for the development of targeting nanomedicines at the same time. With increasing understanding of protein corona, the strategies to regulate protein corona pave new avenues for the development of targeting nanomedicines.
Collapse
Affiliation(s)
- Zhuxuan Jiang
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, P.R. China
| | - Yuxiu Chu
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, P.R. China
| | - Changyou Zhan
- Center of Medical Research and Innovation, Shanghai Pudong Hospital & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, P.R. China.,Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, P.R. China.,Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, P.R. China
| |
Collapse
|
44
|
Mojarad-Jabali S, Mahdinloo S, Farshbaf M, Sarfraz M, Fatahi Y, Atyabi F, Valizadeh H. Transferrin receptor-mediated liposomal drug delivery: recent trends in targeted therapy of cancer. Expert Opin Drug Deliv 2022; 19:685-705. [PMID: 35698794 DOI: 10.1080/17425247.2022.2083106] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Compared to normal cells, malignant cancer cells require more iron for their growth and rapid proliferation, which can be supplied by a high expression level of transferrin receptor (TfR). It is well known that the expression of TfR on the tumor cells is considerably higher than that of normal cells, which makes TfR an attractive target in cancer therapy. AREAS COVERED In this review, the primary focus is on the role of TfR as a valuable tool for cancer-targeted drug delivery, followed by the full coverage of available TfR ligands and their conjugation chemistry to the surface of liposomes. Finally, the most recent studies investigating the potential of TfR-targeted liposomes as promising drug delivery vehicles to different cancer cells are highlighted with emphasis on their improvement possibilities to become a part of future cancer medicines. EXPERT OPINION Liposomes as a valuable class of nanocarriers have gained much attention toward cancer therapy. From all the studies that have exploited the therapeutic and diagnostic potential of TfR on cancer cells, it can be realized that the systematic assessment of TfR ligands applied for liposomal targeted delivery has yet to be entirely accomplished.
Collapse
Affiliation(s)
- Solmaz Mojarad-Jabali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Mahdinloo
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Farshbaf
- Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - Yousef Fatahi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Valizadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
45
|
Gao X, Xu J, Yao T, Liu X, Zhang H, Zhan C. Peptide-decorated nanocarriers penetrating the blood-brain barrier for imaging and therapy of brain diseases. Adv Drug Deliv Rev 2022; 187:114362. [PMID: 35654215 DOI: 10.1016/j.addr.2022.114362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Blood-Brain Barrier (BBB) is one of the most important physiological barriers strictly restricting the substance exchange between blood and brain tissues. While the BBB protects the brain from infections and toxins and maintains brain homeostasis, it is also recognized as the main obstacle to the penetration of therapeutics and imaging agents into the brain. Due to high specificity and affinity, peptides are frequently exploited to decorate nanocarriers across the BBB for diagnosis and/or therapy purposes. However, there are still some challenges that restrict their clinical application, such as stability, safety and immunocompatibility. In this review, we summarize the biological and pathophysiological characteristics of the BBB, strategies across the BBB, and recent progress on peptide decorated nanocarriers for brain diseases diagnosis and therapy. The challenges and opportunities for their translation are also discussed.
Collapse
|
46
|
Zhang L, Cao C, Kaushik N, Lai RY, Liao J, Wang G, Ariotti N, Jin D, Stenzel MH. Controlling the Biological Behaviors of Polymer-Coated Upconverting Nanoparticles by Adjusting the Linker Length of Estrone Ligands. Biomacromolecules 2022; 23:2572-2585. [PMID: 35584062 DOI: 10.1021/acs.biomac.2c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The estrone ligand is used for modifying nanoparticle surfaces to improve their targeting effect on cancer cell lines. However, to date, there is no common agreement on the ideal linker length to be used for the optimum targeting performance. In this study, we aimed to investigate the impact of poly(poly ethylene glycol methyl ether methacrylate) (PPEGMEMA) linker length on the cellular uptake behavior of polymer-coated upconverting nanoparticles (UCNPs). Different triblock terpolymers, poly(poly (ethylene glycol) methyl ether methacrylate)-block-polymethacrylic acid-block-polyethylene glycol methacrylate phosphate (PPEGMEMAx-b-PMAAy-b-PEGMP3: x = 7, 15, 33, and 80; y = 16, 20, 18, and 18), were synthesized with different polymer linker chain lengths between the surface and the targeting ligand by reversible addition-fragmentation chain transfer polymerization. The estrone ligand was attached to the polymer via specific terminal conjugation. The cellular association of polymer-coated UCNPs with linker chain lengths was evaluated in MCF-7 cells by flow cytometry. Our results showed that the bioactivity of ligand modification is dependent on the length of the polymer linker. The shortest polymer PPEGMEMA7-b-PMAA16-b-PEGMP3 with estrone at the end of the polymer chain was found to have the best cellular association behavior in the estrogen receptor (ER)α-positive expression cell line MCF-7. Additionally, the anticancer drug doxorubicin•HCl was encapsulated in the nanocarrier to evaluate the 2D and 3D cytotoxicity. The results showed that estrone modification could efficiently improve the cellular uptake in ERα-positive expression cell lines and in 3D spheroid models.
Collapse
Affiliation(s)
- Lin Zhang
- Cluster for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Sydney 2052, New South Wales, Australia
| | - Cheng Cao
- Cluster for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Sydney 2052, New South Wales, Australia
| | - Natasha Kaushik
- Electron Microscope Unit, University of New South Wales, Sydney 2052, New South Wales, Australia
| | - Rebecca Y Lai
- Cluster for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Sydney 2052, New South Wales, Australia
| | - Jiayan Liao
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney 2006 New South Wales, Australia
| | - Guannan Wang
- Cluster for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Sydney 2052, New South Wales, Australia
| | - Nicholas Ariotti
- Electron Microscope Unit, University of New South Wales, Sydney 2052, New South Wales, Australia
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney 2006 New South Wales, Australia
| | - Martina H Stenzel
- Cluster for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Sydney 2052, New South Wales, Australia
| |
Collapse
|
47
|
Evaluation of CTB-sLip for Targeting Lung Metastasis of Colorectal Cancer. Pharmaceutics 2022; 14:pharmaceutics14040868. [PMID: 35456702 PMCID: PMC9032673 DOI: 10.3390/pharmaceutics14040868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Lung metastasis of colorectal cancer is common in the clinic; however, precise targeting for the diagnosis and therapy purposes of those lung metastases remains challenging. Herein, cholera toxin subunit b (CTB) protein was chemically conjugated on the surface of PEGylated liposomes (CTB-sLip). Both human-derived colorectal cancer cell lines, HCT116 and HT-29, demonstrated high binding affinity and cellular uptake with CTB-sLip. In vivo, CTB-sLip exhibited elevated targeting capability to the lung metastasis of colorectal cancer in the model nude mice in comparison to PEGylated liposomes (sLip) without CTB modification. CTB conjugation induced ignorable effects on the interaction between liposomes and plasma proteins but significantly enhanced the uptake of liposomes by numerous blood cells and splenic cells, leading to relatively rapid blood clearance in BALB/c mice. Even though repeated injections of CTB-sLip induced the production of anti-CTB antibodies, our results suggested CTB-sLip as promising nanocarriers for the diagnosis of lung metastasis of colorectal cancer.
Collapse
|
48
|
Lu L, Xu Q, Wang J, Wu S, Luo Z, Lu W. Drug Nanocrystals for Active Tumor-Targeted Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14040797. [PMID: 35456631 PMCID: PMC9026472 DOI: 10.3390/pharmaceutics14040797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 12/17/2022] Open
Abstract
Drug nanocrystals, which are comprised of active pharmaceutical ingredients and only a small amount of essential stabilizers, have the ability to improve the solubility, dissolution and bioavailability of poorly water-soluble drugs; in turn, drug nanocrystal technology can be utilized to develop novel formulations of chemotherapeutic drugs. Compared with passive targeting strategy, active tumor-targeted drug delivery, typically enabled by specific targeting ligands or molecules modified onto the surface of nanomedicines, circumvents the weak and heterogeneous enhanced permeability and retention (EPR) effect in human tumors and overcomes the disadvantages of nonspecific drug distribution, high administration dosage and undesired side effects, thereby contributing to improving the efficacy and safety of conventional nanomedicines for chemotherapy. Continuous efforts have been made in the development of active tumor-targeted drug nanocrystals delivery systems in recent years, most of which are encouraging and also enlightening for further investigation and clinical translation.
Collapse
Affiliation(s)
- Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China
- Correspondence:
| |
Collapse
|
49
|
Tang Y, Gao J, Wang T, Zhang Q, Wang A, Huang M, Yu R, Chen H, Gao X. The effect of drug loading and multiple administration on the protein corona formation and brain delivery property of PEG-PLA nanoparticles. Acta Pharm Sin B 2022; 12:2043-2056. [PMID: 35847504 PMCID: PMC9279712 DOI: 10.1016/j.apsb.2021.09.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/23/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
The presence of protein corona on the surface of nanoparticles modulates their physiological interactions such as cellular association and targeting property. It has been shown that α-mangostin (αM)-loaded poly(ethylene glycol)-poly(l-lactide) (PEG-PLA) nanoparticles (NP-αM) specifically increased low density lipoprotein receptor (LDLR) expression in microglia and improved clearance of amyloid beta (Aβ) after multiple administration. However, how do the nanoparticles cross the blood‒brain barrier and access microglia remain unknown. Here, we studied the brain delivery property of PEG-PLA nanoparticles under different conditions, finding that the nanoparticles exhibited higher brain transport efficiency and microglia uptake efficiency after αM loading and multiple administration. To reveal the mechanism, we performed proteomic analysis to characterize the composition of protein corona formed under various conditions, finding that both drug loading and multiple dosing affect the composition of protein corona and subsequently influence the cellular uptake of nanoparticles in b.End3 and BV-2 cells. Complement proteins, immunoglobulins, RAB5A and CD36 were found to be enriched in the corona and associated with the process of nanoparticles uptake. Collectively, we bring a mechanistic understanding about the modulator role of protein corona on targeted drug delivery, and provide theoretical basis for engineering brain or microglia-specific targeted delivery system.
Collapse
|
50
|
Luo Z, Lu L, Xu W, Meng N, Wu S, Zhou J, Xu Q, Xie C, Liu Y, Lu W. In vivo self-assembled drug nanocrystals for metastatic breast cancer all-stage targeted therapy. J Control Release 2022; 346:32-42. [PMID: 35378211 DOI: 10.1016/j.jconrel.2022.03.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/11/2022] [Accepted: 03/29/2022] [Indexed: 12/22/2022]
Abstract
Chemotherapy is still the mainstay treatment for metastatic triple-negative breast cancers (TNBC) currently in clinical practice. The unmet needs of chemotherapy for metastatic TNBC are mainly from the insufficient drug delivery and unavailable targeting strategy that thwart the whole progression of metastatic TNBC. The in vivo ligands-mediated active targeting efficiency is usually affected by protein corona. While, the protein corona-bridged natural targeting, in turn, provides a new way for specific drug delivery. Herein, we develop a novel metastatic progression-oriented in vivo self-assembled Cabazitaxel nanocrystals (CNC) delivery system (PC/CNC) through the CNC automatically absorbing functional plasma proteins (transferrin, apolipoprotein A-IV and apolipoprotein E) in vivo, aiming to achieve the simultaneously targeted delivery to primary tumors, circulating tumor cells and metastatic lesions. With the unique advantages of superhigh drug-loading and protein corona empowered active targeting properties to tumor cells, HUVECs, active-platelets and blood-brain barrier/blood-tumor barrier, the PC/CNC exhibits a significantly improved therapeutic effect in metastatic TNBC therapy compared with free drug and CNC-loaded liposomes.
Collapse
Affiliation(s)
- Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Nana Meng
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China; Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System, Minghang Hospital, Fudan University, Shanghai 201199, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|