1
|
Kim M, Choi R, Kim L, Kim YC, Noh I. Cell membrane nanoparticles in cancer therapy: From basic structure to surface functionalization. J Control Release 2025; 382:113752. [PMID: 40254140 DOI: 10.1016/j.jconrel.2025.113752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cell membrane nanoparticles (CNPs) have recently garnered significant attention as effective drug-delivery vehicles. Beyond their simple function of encapsulating cargo within a lipid bilayer structure, the cell membrane is a complex entity derived from biological materials, presenting a variety of surface proteins and glycans. Notable features that enhance their effectiveness as delivery vehicles include the inhibition of protein corona formation in the plasma and the suppression of macrophage phagocytosis, both of which contribute to prolonged blood circulation. Furthermore, CNPs exhibit homotypic targeting effects toward their cells of origin, resulting in reduced side effects, and because they are not xenobiotics, the likelihood of nonspecific immune activation is also minimized. This review focuses on various applications of CNPs in cancer therapeutic studies, examining their structural evolution and surface engineering developments. We introduce studies that leverage the inherent functionality of cell membranes and recent research in functional CNPs synthesized through genetic or chemical engineering methods. Through this review, we aim to trace the progression of CNP research, explore potential directions for their use in biomedical applications, and assess the prospects for clinical trials.
Collapse
Affiliation(s)
- Munsik Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Rohbin Choi
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Lian Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| | - Ilkoo Noh
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
2
|
Yang J, Li X, Li T, Mei J, Chen Y. Recent advances in biomimetic nanodelivery systems for cancer Immunotherapy. Mater Today Bio 2025; 32:101726. [PMID: 40270890 PMCID: PMC12017925 DOI: 10.1016/j.mtbio.2025.101726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/26/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Tumor immunotherapy is a developing and promising therapeutic method. However, the mechanism of tumor immune microenvironment and individual differences of patients make the clinical application of immunotherapy still very limited. The resulting targeting of the tumor environment and immune system is a suitable strategy for tumor therapy. Biomimetic nanodelivery systems (BNDS) coated with nanoparticles has brought new hope for tumor immunotherapy. Due to its high targeting, maximum drug delivery efficiency and immune escape, BNDS has become one of the options for tumor immunotherapy in the future. BNDS combines the advantages of natural cell membranes and nanoparticles and has good targeting properties. This review summarizes the relationship between tumor and immune microenvironment, classification of immunotherapy, engineering modification of cell membrane, and a comprehensive overview of different types of membrane BNDS in immunotherapy. Furthermore, the prospects and challenges of biomimetic nanoparticles coated with membranes in tumor immunotherapy are further discussed.
Collapse
Affiliation(s)
- Jiawei Yang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
| | - Xueqi Li
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
| | - Tongyu Li
- Department of Hematology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| | - Jin Mei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China, No. 508 North Second Ring East Road, Ningbo, 315302, Zhejiang, China
- Institute of Engineering Medicine, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| | - Ying Chen
- Institute of Engineering Medicine, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, 315010, China
| |
Collapse
|
3
|
Yang L, Li W, Huang Z, Zhao Y, Sun Z, Wang H, Cao L, Lu J, Sun R, Ma X, Shao T, Wu X, He S, Liu Z. Engineered Macrophage Membrane-Coated Nanoparticles for Hepatic Ischemia-Reperfusion Injury Therapeutics. Biomater Res 2025; 29:0212. [PMID: 40416938 PMCID: PMC12099054 DOI: 10.34133/bmr.0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/20/2025] [Accepted: 04/26/2025] [Indexed: 05/27/2025] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a common perioperative complication occurring after liver transplantation and can lead to further problems such as early allograft dysfunction (EAD). Currently, the treatment options for HIRI are extremely limited. In this study, we used bioinformatics analysis to elucidate the critical role of neutrophil chemokines (CXC chemokines) in HIRI. By analyzing sequencing data from the hepatic tissue of posttransplant patients with EAD and the reperfused animal model, we discovered that hepatocytes and macrophages are the primary cells secreting CXC chemokines, and the activation of the nuclear factor kappa B (NF-κB) signaling pathway is the main driver of their secretion. Melatonin (MT) can protect cells from oxidative harm while also inhibiting NF-κB signaling, suggesting its potential to ameliorate HIRI. Accordingly, we designed a nanoparticle platform coated with genetically engineered macrophage membranes-called CXCR2-MM@PLGA/MT-to target the cells secreting CXC chemokines. CXCR2 overexpression on the macrophage membranes not only enhanced the targeting capacity of the nanoparticles but also prevented neutrophil infiltration via the scavenging of CXC chemokines. Meanwhile, the MT delivered to the site of injury successfully attenuated CXC chemokine release after macrophage polarization and hepatocyte necrosis by inhibiting NF-κB phosphorylation and inducing antioxidant effects. Through the synergistic effects of MT and the CXCL/CXCR axis-blocking function of the engineered nanoparticles, CXCR2-MM@PLGA/MT attenuated the aggregation of neutrophils at the site of injury, markedly reducing local inflammation and cellular damage following HIRI. This engineered cellular nanoparticle-based therapy could thus serve as a safe, effective, and cost-efficient strategy for treating HIRI.
Collapse
Affiliation(s)
- Long Yang
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiwei Li
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhen Huang
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yinping Zhao
- College of Biomedical Engineering,
Chongqing Medical University, Chongqing, China
| | - Zhenwen Sun
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haoyu Wang
- Department of Orthopedic Oncology, Shanghai General Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai, China
| | - Longpo Cao
- College of Biomedical Engineering,
Chongqing Medical University, Chongqing, China
| | - Jiao Lu
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruirui Sun
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Ma
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tianxin Shao
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xixi Wu
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siqi He
- College of Biomedical Engineering,
Chongqing Medical University, Chongqing, China
| | - Zuojin Liu
- Hepatobiliary Surgery,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Wang S, Zhou H, Mao J, Zhang Y, Qi Y, Pang M, Jin X, Zhang J, Luo L, You J. Precision nanomedicine for pneumonocyte-targeting: Emerging strategies and clinical prospects in refractory pulmonary disease therapy. Biomaterials 2025; 323:123420. [PMID: 40424832 DOI: 10.1016/j.biomaterials.2025.123420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 05/11/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Refractory pulmonary diseases, including chronic obstructive pulmonary disease (COPD) and tuberculosis (TB), pose a critical global health challenge due to the limitations of conventional therapies in advanced stages, such as poor drug penetration, systemic side effects, and inability to eradicate pathogens in protected microenvironments. While the lung's complex structure is essential for respiratory function, it also facilitates persistent damage from environmental and infectious agents. Nanomedicine provides a transformative approach by utilizing customizable carriers (e.g., ligand-gated targeting, stimuli-responsive payload release) to bypass physiological barriers through both passive mechanisms such as enhanced vascular permeability and active-targeting. Such platforms achieve hierarchical drug deposition-from organ-level accumulation to pneumonocyte-targeting-thereby addressing the spatial heterogeneity of therapy-resistant lesions. Besides, A unique advantage of nanomedicine lies in its intrinsic interactions with lung immune cells (e.g., macrophages), allowing dual-functional systems that not only deliver therapeutics to disease sites but also modulate local immune responses-such as reducing inflammation in COPD or enhancing bacterial clearance in TB. This targeted approach improves treatment efficacy while minimizing systemic toxicity. Furthermore, nanomedicine ensures the stability of encapsulated drugs, particularly nucleic acid therapeutics (siRNA, mRNA), which are crucial for treating genetic defect-related pulmonary diseases. Building on the relationship between malignant pulmonary conditions and lung cells, this review summarizes nanoplatform-based strategies for precise targeting and examines ongoing clinical trials. By bridging the gap between preclinical research and clinical application, this review aims to guide the development of novel therapeutic approaches and accelerate the clinical translation of nanomedicines for refractory pulmonary diseases.
Collapse
Affiliation(s)
- Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Huanli Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Yitao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Yuxin Qi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Mei Pang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
5
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
6
|
Liu X, Luo Y, Huang Y, Li M, Guo M, Dong Z, Wu J, Cai G, Zhu H, Wang K, Chen X, Li P, Li Q. Novel Biomarkers as Non-Invasive Diagnostic Tools in IgA Nephropathy: A Comparative Study with Lupus Nephritis and Membranous Nephropathy. J Inflamm Res 2025; 18:4627-4639. [PMID: 40191091 PMCID: PMC11972577 DOI: 10.2147/jir.s512916] [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: 12/18/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
Rationale The diagnostic value of endothelial-associated biomarkers in IgAN and their ability to differentiate it from other kidney diseases have not yet been clarified. Objective This study aimed to investigate the diagnostic value of endothelial-associated biomarkers in IgAN patients. Methods and Results This is a cross-sectional study involving 96 participants, with IgAN, LN, MN, and healthy subjects recruited in a 1:1:1:1 ratio. Seventy-five percent of the sample was used for developing a classification model, and the remaining 25% was used for constructing a validation cohort. Plasma levels of 12 endothelial-associated biomarkers were detected using multiplex immunoassay technology. Among all the biomarkers evaluated, VLA-4 and VEGFD were prioritized for distinguishing IgAN from other groups (p<0.001), with 85% classification accuracy. These two biomarkers also showed significant correlation with eGFR (VLA-4: r = - 0.291, P = 0.021; VEGFD: r = - 0.271, P = 0.031) and Gd-IgA1 (VLA-4: r = 0.403, P = 0.003; VEGFD: r = 0.412, P = 0.002). These two biomarkers also showed superior diagnostic efficacy (AUC=0.952 and 0.945) compared to Gd-IgA1 (AUC=0.736). Subgroup analysis of IgAN patients revealed clinically relevant effect sizes for the IgA and IgA/C3 ratios between high- and low-VLA-4 and VEGFD groups, with Hedges' g values of 0.962 and 0.819, respectively. The diagnostic efficacy of VLA-4 and VEGFD levels in IgAN was further validated in an independent cohort comprising 24 participants. Conclusion VLA-4 and VEGFD emerge as robust, non-invasive biomarkers for IgAN diagnosis and may play significant roles in the pathogenesis of IgAN.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yushuang Luo
- Department of Mathematics and Statistics, Southwest University, Chongqing, People’s Republic of China
| | - Yiyu Huang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Mengfei Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Ming Guo
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Zheyi Dong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Jie Wu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Kaifa Wang
- Department of Mathematics and Statistics, Southwest University, Chongqing, People’s Republic of China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
7
|
Ma L, Chen F, Chen S, Hu Y, Nie S, He X, Li J, Wang Y, Dong Z, Luo H, Guo R, Li M, Xiang B, He Q. Inflammation-targeting nanoparticles impede neutrophil infiltration and scavenge ROS for acute lung injury alleviation. Int J Pharm 2025; 673:125359. [PMID: 39954974 DOI: 10.1016/j.ijpharm.2025.125359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Acute lung injury (ALI) is a severe acute diffuse lung disease caused by noncardiac factors, primarily associated with uncontrolled inflammation and excessive reactive oxygen species (ROS). In severe cases, ALI develops into acute respiratory distress syndrome (ARDS) with high mortality. The current clinical treatments of ALI have shown limited efficacy and fail to reduce mortality. The development of targeted therapeutic strategies presents significant clinical value and demand. Inspired by the fact that the neutrophils adhere to inflammatory endothelium and enter the inflammation site through PSGL-1/P-selectin interaction, we developed a P-selectin-binding platform termed LA NPs. Curcumin-loaded LA NPs (LA/Cur NPs) with multiple therapeutic modules bound to elevated P-selectin to target the inflammatory endothelium in the lung with ALI. Moreover, LA/Cur NPs reduced the infiltration of neutrophils by interfering with the PSGL-1/P-selectin interaction, suppressed the inflammation via the NF-κB pathway, and scavenged excessive reactive oxygen species (ROS), which eventually alleviated ALI in vivo. We provide a promising inflammation-targeting and ROS/inflammation suppression strategy for the treatment of ALI and other inflammatory lung diseases.
Collapse
Affiliation(s)
- Ling Ma
- 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
| | - Fang Chen
- 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
| | - Shuang Chen
- 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
| | - Yeyi Hu
- 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
| | - Shipei Nie
- 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
| | - Xuan He
- 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
| | - Jiaxin Li
- 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
| | - Yashi Wang
- 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
| | - Ziyan Dong
- 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
| | - Haoyuan Luo
- 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
| | - Rong Guo
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041 PR China
| | - Man Li
- 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
| | - Bing Xiang
- Department of Hematology, Hematology Research Laboratory, West China Hospital of Sichuan University, Chengdu 610041, PR China.
| | - Qin He
- 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
|
8
|
Wang H, Gou R, Li W, Chen Z, Gu C, Shi S, Zou L, Li H. Targeting Delivery of Dexamethasone to Inflamed Joints by Albumin-Binding Peptide Modified Liposomes for Rheumatoid Arthritis Therapy. Int J Nanomedicine 2025; 20:3789-3802. [PMID: 40162333 PMCID: PMC11953054 DOI: 10.2147/ijn.s486488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/15/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Delivering the anti-inflammatory dexamethasone in nanoformulations is important for reducing off-target effects when treating rheumatoid arthritis. Nanoformulations can be targeted to sites of inflammation by modifying the nanoparticles with albumin before administration, but such particles can be unstable in vivo. Methods Here, we have developed and validated an alternative targeting in which dexamethasone-loaded liposomes were modified with a 46-residue peptide called "albumin-binding domain", such that the liposomes would adsorb endogenous albumin after administration. Results The resulting liposomes were small (90 nm diameter) and uniformly dispersed, and both X-ray diffraction and differential scanning calorimetry confirmed efficient dexamethasone encapsulation. Functionalizing the liposomes with albumin-binding peptide strongly increased not only their binding to albumin in vitro but also their uptake by RAW264.7 cells in culture. After injection into rats with adjuvant-induced arthritis, the liposomes accumulated and persisted at sites of inflammation, effectively inhibiting joint swelling and reducing the level of the inflammatory factors TNF-α and IL-1β in joints. The liposomes decorated with the albumin-binding peptide did not display obvious hepatotoxicity and did not reduce red and white blood cells number. Discussion Our results validate modifying liposomes with albumin-binding domain as a way to target them to sites of inflammation for efficient drug delivery against rheumatoid arthritis.
Collapse
Affiliation(s)
- Huan Wang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Rui Gou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Wei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Zhoujiang Chen
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Chun Gu
- Department of Hepatobiliary & Pancreatic Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, People’s Republic of China
| | - Hanmei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, People’s Republic of China
| |
Collapse
|
9
|
Samanta R, Haldar N, Pamecha A, Gajbhiye V. Cell membrane-camouflaged nanocarriers: A cutting-edge biomimetic technology to develop cancer immunotherapy. Int J Pharm 2025; 672:125336. [PMID: 39947362 DOI: 10.1016/j.ijpharm.2025.125336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
The development and growth of many diseases are significantly influenced by immune dysregulation. Similarly, uncontrolled tumor growth occurs in cancer because the immune system is unable to identify and eradicate cancer cells. Therefore, to address this issue, cancer immunotherapy plays a crucial role in detecting tumors and inhibiting their growth. This immune-oncotherapy has gained significant interest over the last decade because of its relevant success in biomedical applications. The fundamental goal of immunotherapy in the war against cancer is to develop potent immunotherapies that have minimal side effects and excellent tumor selectivity. To develop these characteristics, nanotechnology offered promising opportunities for cancer immunotherapy. Cell membrane-coated nanoparticles (CMNPs) have recently evolved, which has a tremendous advantage over other nanoparticles (NPs). The CMNPs can be formed by wrapping cell membranes, which can camouflage the specific cell type, allowing these NPs to survive like "self" during blood circulation and escape immune cell capture. These provide NPs with increased biocompatibility, minimal immunogenicity, longer circulation, and targeted tumor therapy. These advantages have made CMNPs a potential delivery vehicle for immunostimulatory drugs, which can induce immunological responses and lead to cancer immunotherapy. Surface modification of CMNPs using cutting-edge genetic engineering techniques revolutionizes cancer immunotherapy to produce new nano-formulations with greater effectiveness. In this review, we briefly discuss the relationship between cancer and the immune system, various techniques of CMNPs synthesis, and the use of naturally occurring and genetically modified CMNPs for cancer immunotherapy.
Collapse
Affiliation(s)
- Rajkumar Samanta
- Nanobioscience, Agharkar Research Institute, Pune 411004 India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007 India
| | - Niladri Haldar
- Nanobioscience, Agharkar Research Institute, Pune 411004 India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007 India
| | - Anchal Pamecha
- Place of Work, Nanobioscience Group, Agharkar Research Institute, Pune 411004 India
| | - Virendra Gajbhiye
- Nanobioscience, Agharkar Research Institute, Pune 411004 India; Savitribai Phule Pune University, Ganeshkhind, Pune 411007 India.
| |
Collapse
|
10
|
Zhu H, Cai F, Li Z, Zhang L, Zhou X, Yao J, Wang W, Zhou L, Jiang X, Xi K, Gu Y, Chen L, Zhou Y. Neutrophil membrane-coated multifunctional biomimetic nanoparticles for spinal cord injuries. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:415-439. [PMID: 39298153 DOI: 10.1080/09205063.2024.2404760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 09/11/2024] [Indexed: 02/25/2025]
Abstract
Spinal cord injury (SCI) is one of the most complex diseases. After SCI, severe secondary injuries can cause intense inflammatory storms and oxidative stress responses, leading to extensive neuronal apoptosis. Effective regulation of inflammation and oxidative stress after SCI remains an unresolved challenge. In this study, resveratrol-loaded nanoparticles coated with neutrophil membranes (NMR) were prepared using the emulsion-solvent evaporation method and membrane encapsulation technology. Multifunctional biomimetic nanoparticles retain neutrophil membrane-related receptors and possess a strong adsorption capacity for inflammatory factors. As a drug carrier, NMR can sustainably release resveratrol for >72 h. Moreover, co-culture studies in vitro show that the NMR help regulate macrophage polarization to relieve inflammatory response, reduce intracellular reactive oxygen species by approximately 50%, and improve mitochondrial membrane potential to alleviate oxidative stress. After injecting NMR into the injury site, it reduces early apoptosis, inhibit scar formation, and promote neural network recovery to improve motor function. This study demonstrates the anti-inflammatory, antioxidant, and neuroprotective effects of NMR, thus providing a novel therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Hongyi Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Feng Cai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Lichen Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Jiapei Yao
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, P.R. China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Liang Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Xinzhao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| | - Yidi Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
| |
Collapse
|
11
|
Mathavan S, Tam YJ, Mustaffa KMF, Tye GJ. Aptamer based immunotherapy: a potential solid tumor therapeutic. Front Immunol 2025; 16:1536569. [PMID: 40034705 PMCID: PMC11873091 DOI: 10.3389/fimmu.2025.1536569] [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: 11/29/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Aptamer-based immunotherapy can be a new hope for treating solid tumors with personalized and specific approaches toward cancer therapies. Aptamers are small synthetic single-stranded nucleic acids that may bring in a paradigm shift in treating solid tumors. These are highly selective drugs applied in cellular immunotherapy, cytokine modulation, and immune checkpoint suppression. This review provides an overview of the recent advances in aptamer-based technologies with specific key clinical trials involving AON-D21 and AM003. Aptamers are potently active in immune regulation and tumor targeting. However, aptamer stability and bioavailability are seriously compromised by the issues relating to renal clearance and rapid degradation through nucleases. The latter are reviewed here along with novel improvements, some of which involve chemical modifications that greatly enhance stability and prolong the circulation time; exemplary such modifications are PEGylation, cholesterol conjugation, and the synthesis of circular nucleic acids. The regulatory aspect is also crucial. For example, in addition to specific strategies to prevent drug-drug interactions (DDIs) in cancer remediation medications, this paper underscores the need of risk assessment, particularly because of immunogenicity and organ failure. The use of aptamers is expanded by the development of SOMAmers, X-aptamers, and bioinformatics. To make aptamer-based drugs a major part of cancer treatment, future research should concentrate more on resolving existing issues and expanding their beneficial uses.
Collapse
Affiliation(s)
- Sarmilah Mathavan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Minden, Pulau Pinang, Malaysia
- Biogenes Technologies Sdn Bhd, Jalan Maklumat, Universiti Putra Malaysia, Serdang, Malaysia
| | - Yew Joon Tam
- Biogenes Technologies Sdn Bhd, Jalan Maklumat, Universiti Putra Malaysia, Serdang, Malaysia
| | | | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Minden, Pulau Pinang, Malaysia
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, National Institutes of Biotechnology Malaysia, Gelugor, Pulau Pinang, Malaysia
| |
Collapse
|
12
|
Hou H, Liu X, Liu J, Wang Y. Carbohydrate polymer-based nanoparticles with cell membrane camouflage for cancer therapy: A review. Int J Biol Macromol 2025; 289:138620. [PMID: 39674458 DOI: 10.1016/j.ijbiomac.2024.138620] [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: 10/08/2024] [Revised: 11/21/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Recent developments in biomimetic nanoparticles, specifically carbohydrate polymer-coated cell membrane nanoparticles, have demonstrated considerable promise in treating cancer. These systems improve drug delivery by imitating natural cell actions, enhancing biocompatibility, and decreasing immune clearance. Conventional drug delivery methods frequently face challenges with non-specific dispersal and immune detection, which can hinder their efficiency and safety. These biomimetic nanoparticles improve target specificity, retention times, and therapeutic efficiency by using biological components like chitosan, hyaluronic acid, and alginate. Chitosan-based nanoparticles, which come from polysaccharides found in nature, have self-assembly abilities that make them better drug carriers. Hyaluronic acid helps target tissues more effectively, especially in cancer environments where there are high levels of hyaluronic acid receptors. Alginate-based systems also enhance drug delivery by being biocompatible and degradable, making them ideal choices for advanced therapeutic uses. Moreover, these particles hold potential for overcoming resistance to multiple drugs and boosting the body's immune reaction to tumors through precise delivery and decreased side effects of chemotherapy drugs. This review delves into the possibilities of using carbohydrate polymer-functionalized nanoparticles and their impact on enhancing the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Haijia Hou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuejian Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Liu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yudong Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
13
|
Zhang S, Zhao X, Lv Y, Niu J, Wei X, Luo Z, Wang X, Chen XL. Exosomes of different cellular origins: prospects and challenges in the treatment of acute lung injury after burns. J Mater Chem B 2025; 13:1531-1547. [PMID: 39704476 DOI: 10.1039/d4tb02351j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Acute lung injury (ALI) is a critical clinical disease caused by direct factors (inhalation injury, gastroesophageal reflux, etc.) or indirect factors (including infection, sepsis, burn, shock, trauma, acute pancreatitis, fat embolism, drug overdose, etc.). ALI is characterized mainly by diffuse interstitial and alveolar edema caused by an uncontrolled inflammatory response and damage to the alveoli-capillary barrier and has very high morbidity and mortality rates. Currently, there is no effective treatment strategy other than mechanical ventilation, fluid management or other supportive treatments. Exosomes are nanovesicle-like vesicles with double-membrane structures detached from the cell membrane or secreted by cells. These vesicles can be used as drug carriers because of their unique biological properties, such as anti-inflammatory, anti-apoptotic, pro-cell growth and immunomodulatory functions, and have been applied in the treatment of ALI in recent years. In this study, the mechanism and pathophysiological characteristics of ALI were first systematically described. The different cellular sources and characteristics of exosomes are summarized, and their functions and value as drug carriers in the treatment of ALI are discussed, as are the challenges that may be faced in the treatment of ALI with exosomes.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Xinyu Zhao
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Yang Lv
- Plastic Surgery Department, The Second Affiliated Hospital of Anhui Medical University, 230061, P. R. China
| | - Jianguo Niu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Xiaolong Wei
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P. R. China.
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230022, China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
14
|
Ciftci F, Özarslan AC, Kantarci İC, Yelkenci A, Tavukcuoglu O, Ghorbanpour M. Advances in Drug Targeting, Drug Delivery, and Nanotechnology Applications: Therapeutic Significance in Cancer Treatment. Pharmaceutics 2025; 17:121. [PMID: 39861768 PMCID: PMC11769154 DOI: 10.3390/pharmaceutics17010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
In the 21st century, thanks to advances in biotechnology and developing pharmaceutical technology, significant progress is being made in effective drug design. Drug targeting aims to ensure that the drug acts only in the pathological area; it is defined as the ability to accumulate selectively and quantitatively in the target tissue or organ, regardless of the chemical structure of the active drug substance and the method of administration. With drug targeting, conventional, biotechnological and gene-derived drugs target the body's organs, tissues, and cells that can be selectively transported to specific regions. These systems serve as drug carriers and regulate the timing of release. Despite having many advantageous features, these systems have limitations in thoroughly treating complex diseases such as cancer. Therefore, combining these systems with nanoparticle technologies is imperative to treat cancer at both local and systemic levels effectively. The nanocarrier-based drug delivery method involves encapsulating target-specific drug molecules into polymeric or vesicular systems. Various drug delivery systems (DDS) were investigated and discussed in this review article. The first part discussed active and passive delivery systems, hydrogels, thermoplastics, microdevices and transdermal-based drug delivery systems. The second part discussed drug carrier systems in nanobiotechnology (carbon nanotubes, nanoparticles, coated, pegylated, solid lipid nanoparticles and smart polymeric nanogels). In the third part, drug targeting advantages were discussed, and finally, market research of commercial drugs used in cancer nanotechnological approaches was included.
Collapse
Affiliation(s)
- Fatih Ciftci
- Department of Biomedical Engineering, Faculty of Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
- Department of Technology Transfer Office, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
| | - Ali Can Özarslan
- Department of Metallurgical and Materials Engineering, Istanbul University-Cerrahpasa, Istanbul 34320, Turkey;
| | - İmran Cagri Kantarci
- Department of Bioengineering, Faculty of Chemistry-Metallurgy, Yildiz Technical University, Istanbul 34210, Turkey;
| | - Aslihan Yelkenci
- Department of Pediatric Dentistry, Faculty of Dentistry, University of Health Sciences, Istanbul 34668, Turkey;
| | - Ozlem Tavukcuoglu
- Department of Biochemistry, Faculty of Hamidiye Pharmacy, University of Health Sciences, Istanbul 34668, Turkey;
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-8-8349, Iran;
| |
Collapse
|
15
|
Wu H, Chen C, Li J, Yu D, Wu X, Huang H, Tang Z, Wu Q, Yan S, Wang N, Wang M, Wei F, Yu Y, Wang D, Shi M, Yue X, Cao P, Zheng Z, Li X, Guo B, Shi L, Guo Z. Engineered Magneto-Piezoelectric Nanoparticles-Enhanced Scaffolds Disrupt Biofilms and Activate Oxidative Phosphorylation in Icam1 + Macrophages for Infectious Bone Defect Regeneration. ACS NANO 2024; 18:35575-35594. [PMID: 39690461 DOI: 10.1021/acsnano.4c13562] [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: 12/19/2024]
Abstract
Infectious bone defects pose significant clinical challenges due to persistent infection and impaired bone healing. Icam1+ macrophages were identified as crucial and previously unrecognized regulators in the repair of bone defects, where impaired oxidative phosphorylation within this macrophage subset represents a significant barrier to effective bone regeneration. To address this challenge, dual-responsive iron-doped barium titanate (BFTO) nanoparticles were synthesized with magnetic and ultrasonic properties. These nanoparticles were further loaded with the anti-inflammatory agent curcumin and coated with engineered mesenchymal stem cell membranes (EMM) modified with γ3 peptide, creating BFTO-Cur@EMM nanoparticles specifically designed to target Icam1+ macrophages. These nanoparticles were shown to disrupt bacterial biofilms under alternating magnetic fields (AMF) and to activate oxidative phosphorylation and osteogenic immune responses in Icam1+ macrophages via low-intensity pulsed ultrasound (LIPUS). Transcriptomic sequencing and validation experiments demonstrated that this approach activates oxidative phosphorylation (OXPHOS) by stimulating the JAK2-STAT3 pathway and inhibiting the MAPK-JNK pathway, thereby promoting the polarization of Icam1+ macrophages toward a pro-reparative phenotype and enhancing the secretion of pro-angiogenic and osteogenic cytokines. These nanoparticles were subsequently integrated into quaternized chitosan (QCS) and tricalcium phosphate (TCP) to create a bioink for three-dimensional (3D) printing anti-infection QT/BFTO-Cur@EMM bone repair scaffolds. In vivo studies indicated that these scaffolds significantly improved the healing of infectious bone defects without causing thermal damage to surrounding tissues. This work highlights the potential of this material and the targeting of Icam1+ macrophages as an effective strategy for simultaneously controlling infection and promoting bone regeneration.
Collapse
Affiliation(s)
- Hao Wu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Changcheng Chen
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Jiangfeng Li
- Institute of Burn Research, Southwest Hospital & State key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Dongmei Yu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Hai Huang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Zhen Tang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Qi Wu
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Shichao Yan
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Ning Wang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Mo Wang
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Feilong Wei
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Yunlong Yu
- Institute of Burn Research, Southwest Hospital & State key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, P. R. China
| | - Duan Wang
- Orthopaedic Research Institute, Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Mengting Shi
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Xusong Yue
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Pengfei Cao
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Zenghui Zheng
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Xiaokang Li
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| | - Baolin Guo
- State Key Laboratory for Mechanical Behavior of Materials, and Frontier Institute of Science and Technology, and Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Lei Shi
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P. R. China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P. R. China
| |
Collapse
|
16
|
Liu J, Yan Y, Zhang Y, Pan X, Xia H, Zhou J, Wan F, Huang X, Zhang W, Zhang Q, Chen B, Wang Y. Lysosome-Mitochondria Cascade Targeting Nanoparticle Drives Robust Pyroptosis for Cancer Immunotherapy. J Am Chem Soc 2024; 146:34568-34582. [PMID: 39639594 DOI: 10.1021/jacs.4c12264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The subcellular distribution of cargoes plays a crucial role in determining cell fate and therapeutic efficacy. However, achieving the precise delivery of therapeutics to specific intracellular targets remains a significant challenge. Here, we present a trimodular and acid/enzyme-gated nanoplatform (TAEN) that undergoes disassembly within acidic endosomes and then is cleaved by lysosomal cathepsin B to facilitate efficient and targeted transport of released cargoes into mitochondria compartments. By utilizing this nanovehicle, we successfully achieve selective sorting of photosensitizer molecules into mitochondria with a colocalization coefficient of up to 0.98, leading to the generation of reactive oxygen species stress specifically within the mitochondria for potent pyroptosis-based cancer therapy. The induction of mitochondrial stress triggers the intrinsic apoptotic pathway as well as caspase-3/gasdermin-E (GSDME) cascade, resulting in an enhanced cancer cell killing efficacy by nearly 2 orders of magnitude as compared to lysosomal stress. Furthermore, due to its superior capability to stimulate both innate and adaptive immune responses, our mitochondria-sorted nanophotosensitizer exhibits robust antitumor immune efficacy in multiple tumor-bearing mice models. This study not only provides insights into engineering nanomedicines for subcellular targeted delivery but also offers a valuable toolkit for advanced research in the field of nanobiology at subcellular resolution.
Collapse
Affiliation(s)
- Jianxiong Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yue Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yimeng Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xingquan Pan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiayi Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fangjie Wan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinyu Huang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Weiwei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Chemical Biology Center, Peking University, Beijing 100191, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo 315832, China
| |
Collapse
|
17
|
Naskar A, Kilari S, Baranwal G, Kane J, Misra S. Nanoparticle-Based Drug Delivery for Vascular Applications. Bioengineering (Basel) 2024; 11:1222. [PMID: 39768040 PMCID: PMC11673055 DOI: 10.3390/bioengineering11121222] [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: 08/07/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 01/05/2025] Open
Abstract
Nanoparticle (NP)-based drug delivery systems have received widespread attention due to the excellent physicochemical properties of nanomaterials. Different types of NPs such as lipid NPs, poly(lactic-co-glycolic) acid (PLGA) NPs, inorganic NPs (e.g., iron oxide and Au), carbon NPs (graphene and carbon nanodots), 2D nanomaterials, and biomimetic NPs have found favor as drug delivery vehicles. In this review, we discuss the different types of customized NPs for intravascular drug delivery, nanoparticle behaviors (margination, adhesion, and endothelium uptake) in blood vessels, and nanomaterial compatibility for successful drug delivery. Additionally, cell surface protein targets play an important role in targeted drug delivery, and various vascular drug delivery studies using nanoparticles conjugated to these proteins are reviewed. Finally, limitations, challenges, and potential solutions for translational research regarding NP-based vascular drug delivery are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (A.N.); (S.K.); (G.B.); (J.K.)
| |
Collapse
|
18
|
Lu H, Wang J, Chen Z, Wang J, Jiang Y, Xia Z, Hou Y, Shang P, Li R, Liu Y, Xie J. Engineered Macrophage Membrane-Coated S100A9-siRNA for Ameliorating Myocardial Ischemia-Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403542. [PMID: 39264262 PMCID: PMC11538685 DOI: 10.1002/advs.202403542] [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: 04/04/2024] [Revised: 09/03/2024] [Indexed: 09/13/2024]
Abstract
Despite the widespread adoption of emergency coronary reperfusion therapy, reperfusion-induced myocardial injury remains a challenging issue in clinical practice. Following myocardial reperfusion, S100A8/A9 molecules are considered pivotal in initiating and regulating tissue inflammatory damage. Effectively reducing the S100A8/A9 level in ischemic myocardial tissue holds significant therapeutic value in salvaging damaged myocardium. In this study, HA (hemagglutinin)- and RAGE (receptor for advanced glycation end products)- comodified macrophage membrane-coated siRNA nanoparticles (MMM/RNA NPs) with siRNA targeting S100A9 (S100A9-siRNA) are successfully prepared. This nanocarrier system is able to target effectively the injured myocardium in an inflammatory environment while evading digestive damage by lysosomes. In vivo, migration of MMM/RNA NPs to myocardial injury lesions is confirmed in a myocardial ischemia-reperfusion injury (MIRI) mouse model. Intravenous injection of MMM/RNA NPs significantly reduced S100A9 levels in serum and myocardial tissues, further decreasing myocardial infarction area and improving cardiac function. Targeted reduction of S100A8/A9 by genetically modified macrophage membrane-coated nanoparticles may represent a new therapeutic intervention for MIRI.
Collapse
Affiliation(s)
- He Lu
- Nanjing Drum Tower HospitalDrum Tower Clinical CollegeNanjing University of Chinese MedicineNo. 321 Zhongshan RoadNanjing210008China
| | - Junzhuo Wang
- Nanjing Drum Tower HospitalDrum Tower Clinical CollegeNanjing University of Chinese MedicineNo. 321 Zhongshan RoadNanjing210008China
| | - Ziwei Chen
- Department of CardiologyAffiliated Hospital of Nantong UniversityNantong226001China
| | - Jing Wang
- Nanjing Drum Tower HospitalDrum Tower Clinical CollegeNanjing University of Chinese MedicineNo. 321 Zhongshan RoadNanjing210008China
| | - Yaohui Jiang
- Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNo. 321 Zhongshan RoadNanjing210008China
| | - Zequn Xia
- Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNo. 321 Zhongshan RoadNanjing210008China
| | - Ya Hou
- Nanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNo. 321 Zhongshan RoadNanjing210008China
| | - Pingping Shang
- Department of CardiologyThe People's Hospital of Jiawang District of XuzhouXuzhou221011China
| | - Rutian Li
- Department of OncologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNo. 321 Zhongshan RoadNanjing210008China
| | - Yuyong Liu
- Department of Cardiac SurgeryNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Beijing Institute of HeartLung, and Blood Vessel DiseasesBeijing Anzhen Hospital Affiliated to Capital Medical UniversityBeijing100029China
| | - Jun Xie
- Nanjing Drum Tower HospitalDrum Tower Clinical CollegeNanjing University of Chinese MedicineNo. 321 Zhongshan RoadNanjing210008China
- Department of Cardiac SurgeryNational Cardiovascular Disease Regional Center for Anhuithe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| |
Collapse
|
19
|
Cong X, Zhang Z, Li H, Yang YG, Zhang Y, Sun T. Nanocarriers for targeted drug delivery in the vascular system: focus on endothelium. J Nanobiotechnology 2024; 22:620. [PMID: 39396002 PMCID: PMC11470712 DOI: 10.1186/s12951-024-02892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
Endothelial cells (ECs) are pivotal in maintaining vascular health, regulating hemodynamics, and modulating inflammatory responses. Nanocarriers hold transformative potential for precise drug delivery within the vascular system, particularly targeting ECs for therapeutic purposes. However, the complex interactions between vascular ECs and nanocarriers present significant challenges for the development and clinical translation of nanotherapeutics. This review assesses recent advancements and key strategies in employing nanocarriers for drug delivery to vascular ECs. It suggested that through precise physicochemical design and surface modifications, nanocarriers can enhance targeting specificity and improve drug internalization efficiency in ECs. Additionally, we elaborated on the applications of nanocarriers specifically designed for targeting ECs in the treatment of cardiovascular diseases, cancer metastasis, and inflammatory disorders. Despite these advancements, safety concerns, the complexity of in vivo processes, and the challenge of achieving subcellular drug delivery remain significant obstacles to the effective targeting of ECs with nanocarriers. A comprehensive understanding of endothelial cell biology and its interaction with nanocarriers is crucial for realizing the full potential of targeted drug delivery systems.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - Zebin Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, 130012, Jilin, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China.
| |
Collapse
|
20
|
Funahashi Y, Park SH, Hebert JF, Eiwaz MB, Munhall AC, Groat T, Zeng L, Kim J, Choi HS, Hutchens MP. Nanotherapeutic kidney cell-specific targeting to ameliorate acute kidney injury. Kidney Int 2024; 106:597-610. [PMID: 39067856 DOI: 10.1016/j.kint.2024.06.021] [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: 07/12/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
Acute kidney injury (AKI) increases the risk of in-hospital death, adds to expense of care, and risk of early chronic kidney disease. AKI often follows an acute event such that timely treatment could ameliorate AKI and potentially reduce the risk of additional disease. Despite therapeutic success of dexamethasone in animal models, clinical trials have not demonstrated broad success. To improve the safety and efficacy of dexamethasone for AKI, we developed and characterized a novel, kidney-specific nanoparticle enabling specific within-kidney targeting to proximal tubular epithelial cells provided by the megalin ligand cilastatin. Cilastatin and dexamethasone were complexed to H-Dot nanoparticles, which were constructed from generally recognized as safe components. Cilastatin/Dexamethasone/H-Dot nanotherapeutics were found to be stable at plasma pH and demonstrated salutary release kinetics at urine pH. In vivo, they were specifically biodistributed to the kidney and bladder, with 75% recovery in the urine and with reduced systemic toxicity compared to native dexamethasone. Cilastatin complexation conferred proximal tubular epithelial cell specificity within the kidney in vivo and enabled dexamethasone delivery to the proximal tubular epithelial cell nucleus in vitro. The Cilastatin/Dexamethasone/H-Dot nanotherapeutic improved kidney function and reduced kidney cellular injury when administered to male C57BL/6 mice in two translational models of AKI (rhabdomyolysis and bilateral ischemia reperfusion). Thus, our design-based targeting and therapeutic loading of a kidney-specific nanoparticle resulted in preservation of the efficacy of dexamethasone, combined with reduced off-target disposition and toxic effects. Hence, our study illustrates a potential strategy to target AKI and other diseases of the kidney.
Collapse
Affiliation(s)
- Yoshio Funahashi
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Seung Hun Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica F Hebert
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Mahaba B Eiwaz
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Adam C Munhall
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Tahnee Groat
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Lingxue Zeng
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Jonghan Kim
- Department of Biomedical & Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael P Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA; Operative Care Division, Portland VA Medical Center, Portland, Oregon, USA.
| |
Collapse
|
21
|
Sun Y, Liu Y, Li R, Zhang C, Wu M, Zhang X, Zheng A, Liao N, Zheng Y, Xu H, Zeng R, Zeng Y, Liu X. Multifunctional Biomimetic Nanocarriers for Dual-Targeted Immuno-Gene Therapy Against Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400951. [PMID: 38973319 PMCID: PMC11425963 DOI: 10.1002/advs.202400951] [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: 01/25/2024] [Revised: 05/22/2024] [Indexed: 07/09/2024]
Abstract
Growing evidences have proved that tumors evade recognition and attack by the immune system through immune escape mechanisms, and PDL1/Pbrm1 genes have a strong correlation with poor response or resistance to immune checkpoint blockade (ICB) therapy. Herein, a multifunctional biomimetic nanocarrier (siRNA-CaP@PD1-NVs) is developed, which can not only enhance the cytotoxic activity of immune cells by blocking PD1/PDL1 axis, but also reduce tumor immune escape via Pbrm1/PDL1 gene silencing, leading to a significant improvement in tumor immunosuppressive microenvironment. Consequently, the nanocarrier promotes DC cell maturation, enhances the infiltration and activity of CD8+ T cells, and forms long-term immune memory, which can effectively inhibit tumor growth or even eliminate tumors, and prevent tumor recurrence and metastasis. Overall, this study presents a powerful strategy for co-delivery of siRNA drugs, immune adjuvant, and immune checkpoint inhibitors, and holds great promise for improving the effectiveness and safety of current immunotherapy regimens.
Collapse
Affiliation(s)
- Yupeng Sun
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Yan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- College of Biological Science and EngineeringFuzhou UniversityFuzhou350116P. R. China
| | - Rui Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- College of Biological Science and EngineeringFuzhou UniversityFuzhou350116P. R. China
| | - Cuilin Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Xiaolong Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Aixian Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Haipo Xu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
| | - Rui Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
- Liver Disease CenterThe First Affiliated Hospital of Fujian Medical UniversityFuzhou350005P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian ProvinceMengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhou350025P. R. China
- Mengchao Med‐X CenterFuzhou UniversityFuzhou350116P. R. China
- CAS Key Laboratory of Design and Assembly of Functional NanostructuresFujian Institute of Research on the Structure of Matter Chinese Academy of SciencesFuzhou350002P. R. China
| |
Collapse
|
22
|
Wang Z, Chen F, Cao Y, Zhang F, Sun L, Yang C, Xie X, Wu Z, Sun M, Ma F, Shao D, Leong KW, Pei R. An Engineered Nanoplatform with Tropism Toward Irradiated Glioblastoma Augments Its Radioimmunotherapy Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314197. [PMID: 38713519 DOI: 10.1002/adma.202314197] [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: 12/26/2023] [Revised: 03/27/2024] [Indexed: 05/09/2024]
Abstract
Combining radiotherapy with immune checkpoint blockade therapy offers a promising approach to treat glioblastoma multiforme (GBM), yet challenges such as limited effectiveness and immune-related adverse events (irAEs) persist. These issues are largely due to the failure in targeting immunomodulators directly to the tumor microenvironment. To address this, a biomimetic nanoplatform that combines a genetically modified mesenchymal stem cell (MSC) membrane with a bioactive nanoparticle core for chemokine-directed radioimmunotherapy of GBM is developed. The CC chemokine receptor 2 (CCR2)-overexpressing MSC membrane acts as a tactical tentacle to achieve radiation-induced tropism toward the abundant chemokine (CC motif) ligand 2 (CCL2) in irradiated gliomas. The nanoparticle core, comprising diselenide-bridged mesoporous silica nanoparticles (MSNs) and PD-L1 antibodies (αPD-L1), enables X-ray-responsive drug release and radiosensitization. In two murine models with orthotopic GBM tumors, this nanoplatform reinvigorated immunogenic cell death, and augmented the efficacy and specificity of GBM radioimmunotherapy, with reduced occurrence of irAEs. This study suggests a promising radiation-induced tropism strategy for targeted drug delivery, and presents a potent nanoplatform that enhances the efficacy and safety of radio-immunotherapy.
Collapse
Affiliation(s)
- Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Fan Zhang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Lina Sun
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Chao Yang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Ziping Wu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Madi Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Fanshu Ma
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
23
|
Ma J, Ding L, Peng X, Jiang L, Liu G. Recent Advances of Engineered Cell Membrane-Based Nanotherapeutics to Combat Inflammatory Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308646. [PMID: 38334202 DOI: 10.1002/smll.202308646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/20/2024] [Indexed: 02/10/2024]
Abstract
An immune reaction known as inflammation serves as a shield from external danger signals, but an overactive immune system may additionally lead to tissue damage and even a variety of inflammatory disorders. By inheriting biological functionalities and serving as both a therapeutic medication and a drug carrier, cell membrane-based nanotherapeutics offer the potential to treat inflammatory disorders. To further strengthen the anti-inflammatory benefits of natural cell membranes, researchers alter and optimize the membranes using engineering methods. This review focuses on engineered cell membrane-based nanotherapeutics (ECMNs) and their application in treating inflammation-related diseases. Specifically, this article discusses the methods of engineering cell membranes for inflammatory diseases and examines the progress of ECMNs in inflammation-targeted therapy, inflammation-neutralizing therapy, and inflammation-immunomodulatory therapy. Additionally, the article looks into the perspectives and challenges of ECMNs in inflammatory treatment and offers suggestions as well as guidance to encourage further investigations and implementations in this area.
Collapse
Affiliation(s)
- Jiaxin Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Linyu Ding
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xuqi Peng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Lai Jiang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Gang Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
24
|
Puccetti M, Pariano M, Schoubben A, Ricci M, Giovagnoli S. Engineering carrier nanoparticles with biomimetic moieties for improved intracellular targeted delivery of mRNA therapeutics and vaccines. J Pharm Pharmacol 2024; 76:592-605. [PMID: 38092697 DOI: 10.1093/jpp/rgad089] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/12/2023] [Indexed: 06/07/2024]
Abstract
Biological membrane-engineered lipid nanoparticles (LNP) have shown enormous potential as vehicles for drug delivery due to their outstanding biomimetic properties. To make these nanoparticles more adaptable to complex biological systems, several methods and cellular sources have been adopted to introduce biomembrane-derived moieties onto LNP and provide the latter with more functions while preserving their intrinsic nature. In this review, we focus on LNP decoration with specific regard to mRNA therapeutics and vaccines. The bio-engineering approach exploits a variety of biomembranes for functionalization, such as those derived from red blood cells, white blood cells, cancer cells, platelets, exosomes, and others. Biomembrane engineering could greatly enhance efficiency in targeted drug delivery, treatment, and diagnosis of cancer, inflammation, immunological diseases, and a variety of pathologic conditions. These membrane-modification techniques are expected to advance biomembrane-derived LNP into wider applications in the future.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aurelie Schoubben
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
25
|
Gao R, Lin P, Fang Z, Yang W, Gao W, Wang F, Pan X, Yu W. Cell-derived biomimetic nanoparticles for the targeted therapy of ALI/ARDS. Drug Deliv Transl Res 2024; 14:1432-1457. [PMID: 38117405 DOI: 10.1007/s13346-023-01494-6] [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] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common clinical critical diseases with high morbidity and mortality. Especially since the COVID-19 outbreak, the mortality rates of critically ill patients with ARDS can be as high as 60%. Therefore, this problem has become a matter of concern to respiratory critical care. To date, the main clinical measures for ALI/ARDS are mechanical ventilation and drug therapy. Although ventilation treatment reduces mortality, it increases the risk of hyperxemia, and drug treatment lacks safe and effective delivery methods. Therefore, novel therapeutic strategies for ALI/ARDS are urgently needed. Developments in nanotechnology have allowed the construction of a safe, efficient, precise, and controllable drug delivery system. However, problems still encounter in the treatment of ALI/ARDS, such as the toxicity, poor targeting ability, and immunogenicity of nanomaterials. Cell-derived biomimetic nanodelivery drug systems have the advantages of low toxicity, long circulation, high targeting, and high bioavailability and show great therapeutic promises for ALI/ARDS owing to their acquired cellular biological features and some functions. This paper reviews ALI/ARDS treatments based on cell membrane biomimetic technology and extracellular vesicle biomimetic technology, aiming to achieve a significant breakthrough in ALI/ARDS treatments.
Collapse
Affiliation(s)
- Rui Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Peihong Lin
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhengyu Fang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Wenjing Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Wenyan Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| | - Fangqian Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Xuwang Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China.
| | - Wenying Yu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China.
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China.
| |
Collapse
|
26
|
Sun L, Wang D, Feng K, Zhang JA, Gao W, Zhang L. Cell membrane-coated nanoparticles for targeting carcinogenic bacteria. Adv Drug Deliv Rev 2024; 209:115320. [PMID: 38643841 DOI: 10.1016/j.addr.2024.115320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
The etiology of cancers is multifactorial, with certain bacteria established as contributors to carcinogenesis. As the understanding of carcinogenic bacteria deepens, interest in cancer treatment through bacterial eradication is growing. Among emerging antibacterial platforms, cell membrane-coated nanoparticles (CNPs), constructed by enveloping synthetic substrates with natural cell membranes, exhibit significant promise in overcoming challenges encountered by traditional antibiotics. This article reviews recent advancements in developing CNPs for targeting carcinogenic bacteria. It first summarizes the mechanisms of carcinogenic bacteria and the status of cancer treatment through bacterial eradication. Then, it reviews engineering strategies for developing highly functional and multitasking CNPs and examines the emerging applications of CNPs in combating carcinogenic bacteria. These applications include neutralizing virulence factors to enhance bacterial eradication, exploiting bacterium-host binding for precise antibiotic delivery, and modulating antibacterial immunity to inhibit bacterial growth. Overall, this article aims to inspire technological innovations in developing CNPs for effective cancer treatment through oncogenic bacterial targeting.
Collapse
Affiliation(s)
- Lei Sun
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Dan Wang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Kailin Feng
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Jiayuan Alex Zhang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
27
|
Jia Y, Zhang L, Xu J, Xiang L. Recent advances in cell membrane camouflaged nanotherapeutics for the treatment of bacterial infection. Biomed Mater 2024; 19:042006. [PMID: 38697197 DOI: 10.1088/1748-605x/ad46d4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 05/01/2024] [Indexed: 05/04/2024]
Abstract
Infectious diseases caused by bacterial infections are common in clinical practice. Cell membrane coating nanotechnology represents a pioneering approach for the delivery of therapeutic agents without being cleared by the immune system in the meantime. And the mechanism of infection treatment should be divided into two parts: suppression of pathogenic bacteria and suppression of excessive immune response. The membrane-coated nanoparticles exert anti-bacterial function by neutralizing exotoxins and endotoxins, and some other bacterial proteins. Inflammation, the second procedure of bacterial infection, can also be suppressed through targeting the inflamed site, neutralization of toxins, and the suppression of pro-inflammatory cytokines. And platelet membrane can affect the complement process to suppress inflammation. Membrane-coated nanoparticles treat bacterial infections through the combined action of membranes and nanoparticles, and diagnose by imaging, forming a theranostic system. Several strategies have been discovered to enhance the anti-bacterial/anti-inflammatory capability, such as synthesizing the material through electroporation, pretreating with the corresponding pathogen, membrane hybridization, or incorporating with genetic modification, lipid insertion, and click chemistry. Here we aim to provide a comprehensive overview of the current knowledge regarding the application of membrane-coated nanoparticles in preventing bacterial infections as well as addressing existing uncertainties and misconceptions.
Collapse
Affiliation(s)
- Yinan Jia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Zhang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
28
|
Huang D, Wang X, Wang W, Li J, Zhang X, Xia B. Cell-membrane engineering strategies for clinic-guided design of nanomedicine. Biomater Sci 2024; 12:2865-2884. [PMID: 38686665 DOI: 10.1039/d3bm02114a] [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: 05/02/2024]
Abstract
Cells are the fundamental units of life. The cell membrane primarily composed of two layers of phospholipids (a bilayer) structurally defines the boundary of a cell, which can protect its interior from external disturbances and also selectively exchange substances and conduct signals from the extracellular environment. The complexity and particularity of transmembrane proteins provide the foundation for versatile cellular functions. Nanomedicine as an emerging therapeutic strategy holds tremendous potential in the healthcare field. However, it is susceptible to recognition and clearance by the immune system. To overcome this bottleneck, the technology of cell membrane coating has been extensively used in nanomedicines for their enhanced therapeutic efficacy, attributed to the favorable fluidity and biocompatibility of cell membranes with various membrane-anchored proteins. Meanwhile, some engineering strategies of cell membranes through various chemical, physical and biological ways have been progressively developed to enable their versatile therapeutic functions against complex diseases. In this review, we summarized the potential clinical applications of four typical cell membranes, elucidated their underlying therapeutic mechanisms, and outlined their current engineering approaches. In addition, we further discussed the limitation of this technology of cell membrane coating in clinical applications, and possible solutions to address these challenges.
Collapse
Affiliation(s)
- Di Huang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Xiaoyu Wang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Wentao Wang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Jiachen Li
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen/University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Xiaomei Zhang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Bing Xia
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, P. R. China
| |
Collapse
|
29
|
Qin X, Zhu L, Zhong Y, Wang Y, Luo X, Li J, Yan F, Wu G, Qiu J, Wang G, Qu K, Zhang K, Wu W. Universal cell membrane camouflaged nano-prodrugs with right-side-out orientation adapting for positive pathological vascular remodeling in atherosclerosis. Chem Sci 2024; 15:7524-7544. [PMID: 38784734 PMCID: PMC11110172 DOI: 10.1039/d4sc00761a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 05/25/2024] Open
Abstract
A right-side-out orientated self-assembly of cell membrane-camouflaged nanotherapeutics is crucial for ensuring their biological functionality inherited from the source cells. In this study, a universal and spontaneous right-side-out coupling-driven ROS-responsive nanotherapeutic approach, based on the intrinsic affinity between phosphatidylserine (PS) on the inner leaflet and PS-targeted peptide modified nanoparticles, has been developed to target foam cells in atherosclerotic plaques. Considering the increased osteopontin (OPN) secretion from foam cells in plaques, a bioengineered cell membrane (OEM) with an overexpression of integrin α9β1 is integrated with ROS-cleavable prodrugs, OEM-coated ETBNPs (OEM-ETBNPs), to enhance targeted drug delivery and on-demand drug release in the local lesion of atherosclerosis. Both in vitro and in vivo experimental results confirm that OEM-ETBNPs are able to inhibit cellular lipid uptake and simultaneously promote intracellular lipid efflux, regulating the positive cellular phenotypic conversion. This finding offers a versatile platform for the biomedical applications of universal cell membrane camouflaging biomimetic nanotechnology.
Collapse
Affiliation(s)
- Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University Chongqing 400016 China
| | - Xiaoshan Luo
- Guizhou Information Engineering University Bijie 551700 China
| | - Jiawei Li
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Fei Yan
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
| | - Guicheng Wu
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- JinFeng Laboratory Chongqing 401329 China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- JinFeng Laboratory Chongqing 401329 China
| |
Collapse
|
30
|
Zhong Z, Deng W, Wu J, Shang H, Tong Y, He Y, Huang Q, Ba X, Chen Z, Tang K. Cell membrane coated nanoparticles as a biomimetic drug delivery platform for enhancing cancer immunotherapy. NANOSCALE 2024; 16:8708-8738. [PMID: 38634521 DOI: 10.1039/d4nr00284a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Cancer immunotherapy, a burgeoning modality for cancer treatment, operates by activating the autoimmune system to impede the growth of malignant cells. Although numerous immunotherapy strategies have been employed in clinical cancer therapy, the resistance of cancer cells to immunotherapeutic medications and other apprehensions impede the attainment of sustained advantages for most patients. Recent advancements in nanotechnology for drug delivery hold promise in augmenting the efficacy of immunotherapy. However, the efficacy is currently constrained by the inadequate specificity of delivery, low rate of response, and the intricate immunosuppressive tumor microenvironment. In this context, the investigation of cell membrane coated nanoparticles (CMNPs) has revealed their ability to perform targeted delivery, immune evasion, controlled release, and immunomodulation. By combining the advantageous features of natural cell membranes and nanoparticles, CMNPs have demonstrated their unique potential in the realm of cancer immunotherapy. This review aims to emphasize recent research progress and elucidate the underlying mechanisms of CMNPs as an innovative drug delivery platform for enhancing cancer immunotherapy. Additionally, it provides a comprehensive overview of the current immunotherapeutic strategies involving different cell membrane types of CMNPs, with the intention of further exploration and optimization.
Collapse
Affiliation(s)
- Zichen Zhong
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Wen Deng
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Qiu Huang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji medical college, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
31
|
Liao J, Lu L, Chu X, Xiong Y, Zhou W, Cao F, Cheng P, Shahbazi MA, Liu G, Mi B. Cell membrane coated nanoparticles: cutting-edge drug delivery systems for osteoporosis therapy. NANOSCALE 2024; 16:8236-8255. [PMID: 38584466 DOI: 10.1039/d3nr06264c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Osteoporosis, characterized by a reduction in bone mineral density, represents a prevalent skeletal disorder with substantial global health implications. Conventional therapeutic strategies, exemplified by bisphosphonates and hormone replacement regimens, though effective, encounter inherent limitations and challenges. Recent years have witnessed the surge of cell-membrane-coated nanoparticles (CMNPs) as a promising intervention for osteoporosis, leveraging their distinct attributes including refined biocompatibility, heightened pharmaceutical payload capacity, as well as targeted drug release kinetics. However, a comprehensive review consolidating the application of CMNPs-based therapy for osteoporosis remains absent within the existing literature. In this review, we provide a concise overview of the distinctive pathogenesis associated with osteoporosis, alongside an in-depth exploration of the physicochemical attributes intrinsic to CMNPs derived from varied cellular sources. Subsequently, we explore the potential utility of CMNPs, elucidating emerging trends in their deployment for osteoporosis treatment through multifaceted therapeutic approaches. By linking the notable attributes of CMNPs with their roles in mitigating osteoporosis, this review serves as a catalyst for further advances in the design of advanced CMNPs tailored for osteoporosis management. Ultimately, such progress is promising for enhancing outcomes in anti-bone loss interventions, paving the way for clinical translation in the near future.
Collapse
Affiliation(s)
- Jiewen Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Li Lu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiangyu Chu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
32
|
Hu N, Xue H, Zhang T, Fan Y, Guo F, Li Z, Huo M, Guan X, Chen G. Harnessing PD-1 cell membrane-coated paclitaxel dimer nanoparticles for potentiated chemoimmunotherapy. Biomed Pharmacother 2024; 174:116482. [PMID: 38520866 DOI: 10.1016/j.biopha.2024.116482] [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: 01/31/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024] Open
Abstract
Chemoimmunotherapy has emerged as a promising strategy for improving the efficacy of cancer treatment. Herein, we present PD-1 receptor-presenting membrane-coated paclitaxel dimers nanoparticles (PD-1@PTX2 NPs) for enhanced treatment efficacy. PD-1 cell membrane-cloaked PTX dimer exhibited effective cellular uptake and increased cytotoxicity against cancer cells. PD-1@PTX2 NPs could selectively bind with PD-L1 ligands expressed on breast cancer cells. Our nanoparticles exhibit a remarkable tumor growth inhibition rate of 71.3% in mice bearing 4T1 xenografts and significantly prolong survival in mouse models of breast cancer. Additionally, our nanoparticles promoted a significant 3.2-fold increase in CD8+ T cell infiltration and 73.7% regulatory T cell (Treg) depletion within tumors, boosting a robust antitumor immune response. These findings underscore the potential of utilizing immune checkpoint receptor-presented PTX nanoparticles to enhance the efficacy of chemoimmunotherapy, providing an alternative approach for improving cancer treatment.
Collapse
Affiliation(s)
- Nannan Hu
- Department of General Surgery, The Affiliated Wenling Hospital of Taizhou University Taizhou 317500, PR China; College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Han Xue
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Ting Zhang
- Department of General Surgery, The Affiliated Wenling Hospital of Taizhou University Taizhou 317500, PR China
| | - Yuxin Fan
- Department of General Surgery, The Affiliated Wenling Hospital of Taizhou University Taizhou 317500, PR China; College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Fenglin Guo
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Zhimin Li
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Mingge Huo
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Xingang Guan
- Medical School, Taizhou University, Taizhou 318001, PR China.
| | - Guofu Chen
- Department of General Surgery, The Affiliated Wenling Hospital of Taizhou University Taizhou 317500, PR China.
| |
Collapse
|
33
|
Xu Y, Lv L, Wang Q, Yao Q, Kou L, Zhang H. Emerging application of nanomedicine-based therapy in acute respiratory distress syndrome. Colloids Surf B Biointerfaces 2024; 237:113869. [PMID: 38522285 DOI: 10.1016/j.colsurfb.2024.113869] [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: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are serious lung injuries caused by various factors, leading to increased permeability of the alveolar-capillary barrier, reduced stability of the alveoli, inflammatory response, and hypoxemia. Despite several decades of research since ARDS was first formally described in 1967, reliable clinical treatment options are still lacking. Currently, supportive therapy and mechanical ventilation are prioritized, and there is no medication that can be completely effective in clinical treatment. In recent years, nanomedicine has developed rapidly and has exciting preclinical treatment capabilities. Using a drug delivery system based on nanobiotechnology, local drugs can be continuously released in lung tissue at therapeutic levels, reducing the frequency of administration and improving patient compliance. Furthermore, this novel drug delivery system can target specific sites and reduce systemic side effects. Currently, many nanomedicine treatment options for ARDS have demonstrated efficacy. This review briefly introduces the pathophysiology of ARDS, discusses various research progress on using nanomedicine to treat ARDS, and anticipates future developments in related fields.
Collapse
Affiliation(s)
- Yitianhe Xu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Leyao Lv
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Qian Wang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Hailin Zhang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Department of Children's Respiration Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
34
|
Pan H, Yang S, Gao L, Zhou J, Cheng W, Chen G, Shuhang W, Li N, Veranič P, Musiol R, Cai Q, Shubhra QT. At the crossroad of nanotechnology and cancer cell membrane coating: Expanding horizons with engineered nanoplatforms for advanced cancer therapy harnessing homologous tumor targeting. Coord Chem Rev 2024; 506:215712. [DOI: 10.1016/j.ccr.2024.215712] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Peng Z, Qi B, Luo Z, Sun Y, Zhang X, Lin J, Pang J, Zhang P, Zhao Z, Wang X, Chen J. Agomir-122-loaded nanoparticles coated with cell membrane of activated fibroblasts to treat frozen shoulder based on homologous targeting. J Nanobiotechnology 2024; 22:165. [PMID: 38600567 PMCID: PMC11008019 DOI: 10.1186/s12951-024-02403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
As a common musculoskeletal disorder, frozen shoulder is characterized by thickened joint capsule and limited range of motion, affecting 2-5% of the general population and more than 20% of patients with diabetes mellitus. Pathologically, joint capsule fibrosis resulting from fibroblast activation is the key event. The activated fibroblasts are proliferative and contractive, producing excessive collagen. Albeit high prevalence, effective anti-fibrosis modalities, especially fibroblast-targeting therapies, are still lacking. In this study, microRNA-122 was first identified from sequencing data as a potential therapeutic agent to antagonize fibroblast activation. Then, Agomir-122, an analog of microRNA-122, was loaded into poly(lactic-co-glycolic acid) (PLGA) nanoparticles (Agomir-122@NP), a carrier with excellent biocompatibility for the agent delivery. Moreover, relying on the homologous targeting effect, we coated Agomir-122@NP with the cell membrane derived from activated fibroblasts (Agomir-122@MNP), with an attempt to inhibit the proliferation, contraction, and collagen production of abnormally activated fibroblasts. After confirming the targeting effect of Agomir-122@MNP on activated fibroblasts in vitro, we proved that Agomir-122@MNP effectively curtailed fibroblasts activation, ameliorated joint capsule fibrosis, and restored range of motion in mouse models both prophylactically and therapeutically. Overall, an effective targeted delivery method was developed with promising translational value against frozen shoulder.
Collapse
Affiliation(s)
- Zhen Peng
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiaotong University, 85# Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Beijie Qi
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Shanghai Medicine College, Fudan University, Shanghai, 201399, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200080, China
| | - Yaying Sun
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiaotong University, 85# Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Xingyu Zhang
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiaotong University, 85# Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Jinrong Lin
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200080, China
| | - Jinhui Pang
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiaotong University, 85# Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Peng Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200080, China
| | - Zhihu Zhao
- Department of Orthopaedics, Tianjin Hospital, No. 406, Jiefangnan Road, Hexi District, Tianjin, 300000, China.
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China.
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiaotong University, 85# Wujin Road, Hongkou District, Shanghai, 200080, China.
| |
Collapse
|
36
|
Liu Y, Lin Z, Wang Y, Chen L, Wang Y, Luo C. Nanotechnology in inflammation: cutting-edge advances in diagnostics, therapeutics and theranostics. Theranostics 2024; 14:2490-2525. [PMID: 38646646 PMCID: PMC11024862 DOI: 10.7150/thno.91394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/14/2024] [Indexed: 04/23/2024] Open
Abstract
Inflammatory dysregulation is intimately associated with the occurrence and progression of many life-threatening diseases. Accurate detection and timely therapeutic intervention on inflammatory dysregulation are crucial for the effective therapy of inflammation-associated diseases. However, the clinical outcomes of inflammation-involved disorders are still unsatisfactory. Therefore, there is an urgent need to develop innovative anti-inflammatory strategies by integrating emerging technological innovations with traditional therapeutics. Biomedical nanotechnology is one of the promising fields that can potentially transform the diagnosis and treatment of inflammation. In this review, we outline recent advances in biomedical nanotechnology for the diagnosis and treatment of inflammation, with special attention paid to nanosensors and nanoprobes for precise diagnosis of inflammation-related diseases, emerging anti-inflammatory nanotherapeutics, as well as nanotheranostics and combined anti-inflammatory applications. Moreover, the prospects and challenges for clinical translation of nanoprobes and anti-inflammatory nanomedicines are highlighted.
Collapse
Affiliation(s)
- Yuting Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Ziqi Lin
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Yuting Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Liuhui Chen
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, P. R. China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| |
Collapse
|
37
|
Tan Y, Wang X, Gu Y, Bao X, Lu H, Sun X, Kang L, Xu B. Neutrophil and endothelial cell membranes coassembled roflumilast nanoparticles attenuate myocardial ischemia/reperfusion injury. Nanomedicine (Lond) 2024; 19:779-797. [PMID: 38426485 DOI: 10.2217/nnm-2023-0313] [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] [Indexed: 03/02/2024] Open
Abstract
Aim: This study aimed to develop biomimetic nanoparticles (NPs) of roflumilast (ROF) for attenuating myocardial ischemia/reperfusion (MI/R) injury. Materials & methods: We synthesized biomimetic ROF NPs and assembled ROF NPs in neutrophil and endothelial cell membranes (NE/ROF NPs). The physical properties of NE/ROF NPs were characterized and biological functions of NE/ROF NPs were tested in vitro. Targeting characteristics, therapeutic efficacy and safety of NE/ROF NPs were examined in mice model of MI/R. Results: NE/ROF NPs exhibited significant anti-inflammatory and antiadhesion effects. Meanwhile, they was effective in reducing MI/R injury in mice. Furthermore, NE/ROF NPs exhibited stronger targeting capabilities and demonstrated good safety. Conclusion: NE/ROF NPs may be a versatile biomimetic drug-delivery system for attenuating MI/R injury.
Collapse
Affiliation(s)
- Ying Tan
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Xun Wang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Yu Gu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Xue Bao
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| | - He Lu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210000, China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210000, China
| |
Collapse
|
38
|
Gao P, Liu Y, Wang X, Feng X, Liu H, Liu S, Huang X, Wu X, Xiong F, Jia X, Hui H, Jiang J, Tian J. Adhesion molecule-targeted magnetic particle imaging nanoprobe for visualization of inflammation in acute lung injury. Eur J Nucl Med Mol Imaging 2024; 51:1233-1245. [PMID: 38095676 DOI: 10.1007/s00259-023-06550-4] [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: 08/26/2023] [Accepted: 11/27/2023] [Indexed: 03/22/2024]
Abstract
PURPOSE Uncontrolled intra-alveolar inflammation is a central pathogenic feature, and its severity translates into a valid prognostic indicator of acute lung injury (ALI). Unfortunately, current clinical imaging approaches are unsuitable for visualizing and quantifying intra-alveolar inflammation. This study aimed to construct a small-sized vascular cell adhesion molecule-1 (VCAM-1)-targeted magnetic particle imaging (MPI) nanoprobe (ESPVPN) to visualize and accurately quantify intra-alveolar inflammation at the molecular level. METHODS ESPVPN was engineered by conjugating a peptide (VHPKQHRGGSK(Cy7)GC) onto a polydopamine-functionalized superparamagnetic iron oxide core. The MPI performance, targeting, and biosafety of the ESPVPN were characterized. VCAM-1 expression in HUVECs and mouse models was evaluated by western blot. The degree of inflammation and distribution of VCAM-1 in the lungs were assessed using histopathology. The expression of pro-inflammatory markers and VCAM-1 in lung tissue lysates was measured using ELISA. After intravenous administration of ESPVPN, MPI and CT imaging were used to analyze the distribution of ESPVPN in the lungs of the LPS-induced ALI models. RESULTS The small-sized (~10 nm) ESPVPN exhibited superior MPI performance compared to commercial MagImaging® and Vivotrax, and ESPVPN had effective targeting and biosafety. VCAM-1 was highly expressed in LPS-induced ALI mice. VCAM-1 expression was positively correlated with the LPS-induced dose (R = 0.9381). The in vivo MPI signal showed positive correlations with both VCAM-1 expression (R = 0.9186) and representative pro-inflammatory markers (MPO, TNF-α, IL-6, IL-8, and IL-1β, R > 0.7). CONCLUSION ESPVPN effectively targeted inflammatory lungs and combined the advantages of MPI quantitative imaging to visualize and evaluate the degree of ALI inflammation.
Collapse
Affiliation(s)
- Pengli Gao
- School of Biological Science and Medicine Engineering & School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, No. 37, Xueyuan Road, Beijing, 100191, China
- School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yu Liu
- School of Biological Science and Medicine Engineering & School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, No. 37, Xueyuan Road, Beijing, 100191, China
- School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaoli Wang
- School of Medical Imaging, Weifang Medical University, Weifang, 261053, China
| | - Xin Feng
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Heng Liu
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, No. 16 Xinjiekou Outer Street, Beijing, 100088, China
| | - Songlu Liu
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiazi Huang
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiangjun Wu
- School of Biological Science and Medicine Engineering & School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, No. 37, Xueyuan Road, Beijing, 100191, China
- School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Fei Xiong
- School of Biological Science and Medicine Engineering & School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, No. 37, Xueyuan Road, Beijing, 100191, China
- School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaohua Jia
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Jingying Jiang
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, No. 37, Xueyuan Road, Beijing, 100191, China.
- School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China.
| | - Jie Tian
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, No. 37, Xueyuan Road, Beijing, 100191, China.
- School of Engineering Medicine, Beihang University, No. 37, Xueyuan Road, Beijing, 100191, China.
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
39
|
Song L, Gao Y, Wang Z, Shi Y. Serum Levels of Intercellular Adhesion Molecule 1 and Vascular Cell Adhesion Molecule 1 as Biomarkers to Predict Radiotherapy Sensitivity in Cervical Cancer. Geburtshilfe Frauenheilkd 2024; 84:370-377. [PMID: 38618575 PMCID: PMC11006555 DOI: 10.1055/a-2275-0717] [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: 11/25/2023] [Accepted: 02/22/2024] [Indexed: 04/16/2024] Open
Abstract
Background Cervical cancer is a significant global health burden, and individualized treatment approaches are necessary due to its heterogeneity. Radiotherapy is a common treatment modality; however, the response varies among patients. The identification of reliable biomarkers to predict radiotherapy sensitivity is crucial. Methods A cohort of 189 patients with stage IB2-IVA cervical cancer, treated with radiotherapy alone or concurrent chemoradiotherapy, was included. Serum samples were collected before treatment, and intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) concentrations were determined. Patients were categorized into radiotherapy-sensitive (RS) and radiotherapy-resistant (RR) groups based on treatment response. Clinicopathological characteristics and survival rates were analyzed. Results The analysis of clinicopathological characteristics showed that age, family history of cervical cancer and post-menopausal status did not significantly differ between RS and RR groups. Tumor size demonstrated a borderline significant association with radiotherapy response, while differentiation degree was significantly associated. Serum ICAM-1 and VCAM-1 concentrations were significantly higher in the RR group compared to the RS group. Combined detection of ICAM-1 and VCAM-1 improved the predictive ability for radiotherapy sensitivity. Higher serum ICAM-1 and VCAM-1 levels were observed in patients with lower tumor differentiation. Five-year overall survival rates differed significantly between patients with high and low ICAM-1 and VCAM-1 levels. Conclusion Serum ICAM-1 and VCAM-1 levels show potential as predictive biomarkers for radiotherapy sensitivity in cervical cancer.
Collapse
Affiliation(s)
- Lina Song
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, China
| | - Yali Gao
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, China
| | - Zhicong Wang
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, China
| | - Yufeng Shi
- Department of Radiation Therapy, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
40
|
Guo H, Guo M, Xia Z, Shao Z. Membrane-coated nanoparticles as a biomimetic targeted delivery system for tumour therapy. BIOMATERIALS TRANSLATIONAL 2024; 5:33-45. [PMID: 39220664 PMCID: PMC11362346 DOI: 10.12336/biomatertransl.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/05/2023] [Accepted: 12/18/2023] [Indexed: 09/04/2024]
Abstract
Drug therapy towards tumours often causes adverse effects because of their non-specific nature. Membrane-coated technology and membrane-coated nanoparticles provide an advanced and promising platform of targeted and safe delivery. By camouflaging the nanoparticles with natural derived or artificially modified cell membranes, the nano-payloads are bestowed with properties from cell membranes such as longer circulation, tumour or inflammation-targeting, immune stimulation, augmenting the performance of traditional therapeutics. In this review, we review the development of membrane coating technology, and summarise the technical details, physicochemical properties, and research status of membrane-coated nanoparticles from different sources in tumour treatment. Finally, we also look forward to the prospects and challenges of transforming membrane coating technology from experiment into clinical use. Taken together, membrane-coated nanoparticles are bound to become one of the most potential anti-tumour strategies in the future.
Collapse
Affiliation(s)
- Haoyu Guo
- Department of Orthopaedic, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Department of Orthopaedic, Beijing Jishuitan Hospital, Fourth Medical College of Peking University, Beijing, China
| | - Mingke Guo
- Department of Orthopaedics, Affiliated Hospital of NCO School of Army Medical University, Shijiazhuang, Hebei Province, China
| | - Zhidao Xia
- Centre for Nanohealth, ILS2, Medical School, Swansea University, Swansea, UK
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
41
|
Huang S, Li YJ, Wu JY, Hao XY, Xu WJ, Tang YC, Zhou M, Zhang JC, Luo S, Xiang DX. Biomimetic nanodecoys deliver cholesterol-modified heteroduplex oligonucleotide to target dopaminergic neurons for the treatment of Parkinson's disease. Acta Biomater 2024; 177:316-331. [PMID: 38244661 DOI: 10.1016/j.actbio.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder characterized by the accumulation of α-synuclein (α-syn) aggregates called Lewy bodies leading to the gradual loss of dopaminergic (DA) neurons in the substantia nigra. Although α-syn expression can be attenuated by antisense oligonucleotides (ASOs) and heteroduplex oligonucleotide (HDO) by intracerebroventricular (ICV) injection, the challenge to peripheral targeted delivery of oligonucleotide safely and effectively into DA neurons remains unresolved. Here, we designed a new DNA/DNA double-stranded (complementary DNA, coDNA) molecule with cholesterol conjugation (Chol-HDO (coDNA)) based on an α-syn-ASO sequence and evaluated its silence efficiency. Further, Chol-HDO@LMNPs, Chol-HDO-loaded, cerebrovascular endothelial cell membrane with DSPE-PEG2000-levodopa modification (L-DOPA-CECm)-coated nanoparticles (NPs), were developed for the targeted treatment of PD by tail intravenous injection. CECm facilitated the blood-brain barrier (BBB) penetration of NPs, together with cholesterol escaped from reticuloendothelial system uptake, as well as L-DOPA was decarboxylated into dopamine which promoted the NPs toward the PD site for DA neuron regeneration. The behavioral tests demonstrated that the nanodecoys improved the efficacy of HDO on PD mice. These findings provide insights into the development of biomimetic nanodecoys loading HDO for precise therapy of PD. STATEMENT OF SIGNIFICANCE: The accumulation of α-synuclein (α-syn) aggregates is a hallmark of PD. Our previous study designed a specific antisense oligonucleotide (ASO) targeting human SNCA, but the traumatic intracerebroventricular (ICV) is not conducive to clinical application. Here, we further optimize the ASO by creating a DNA/DNA double-stranded molecule with cholesterol-conjugated, named Chol-HDO (coDNA), and develop a DA-targeted biomimetic nanodecoy Chol-HDO@LMNPs by engineering cerebrovascular endothelial cells membranes (CECm) with DSPE-PEG2000 and L-DOPA. The in vivo results demonstrated that tail vein injection of Chol-HDO@LMNPs could target DA neurons in the brain and ameliorate motor deficits in a PD mouse model. This investigation provides a promising peripheral delivery platform of L-DOPA-CECm nanodecoy loaded with a new Chol-HDO (coDNA) targeting DA neurons in PD therapy.
Collapse
Affiliation(s)
- Si Huang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Yong-Jiang Li
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Jun-Yong Wu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Xin-Yan Hao
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Wen-Jie Xu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Yu-Cheng Tang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Min Zhou
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Ji-Chun Zhang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital of Central South University, Changsha, 410008, PR China.
| | - Da-Xiong Xiang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China.
| |
Collapse
|
42
|
Wang Z, Li W, Jiang Y, Park J, Gonzalez KM, Wu X, Zhang QY, Lu J. Cholesterol-modified sphingomyelin chimeric lipid bilayer for improved therapeutic delivery. Nat Commun 2024; 15:2073. [PMID: 38453918 PMCID: PMC10920917 DOI: 10.1038/s41467-024-46331-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Cholesterol (Chol) fortifies packing and reduces fluidity and permeability of the lipid bilayer in vesicles (liposomes)-mediated drug delivery. However, under the physiological environment, Chol is rapidly extracted from the lipid bilayer by biomembranes, which jeopardizes membrane stability and results in premature leakage for delivered payloads, yielding suboptimal clinic efficacy. Herein, we report a Chol-modified sphingomyelin (SM) lipid bilayer via covalently conjugating Chol to SM (SM-Chol), which retains membrane condensing ability of Chol. Systemic structure activity relationship screening demonstrates that SM-Chol with a disulfide bond and longer linker outperforms other counterparts and conventional phospholipids/Chol mixture systems on blocking Chol transfer and payload leakage, increases maximum tolerated dose of vincristine while reducing systemic toxicities, improves pharmacokinetics and tumor delivery efficiency, and enhances antitumor efficacy in SU-DHL-4 diffuse large B-cell lymphoma xenograft model in female mice. Furthermore, SM-Chol improves therapeutic delivery of structurally diversified therapeutic agents (irinotecan, doxorubicin, dexamethasone) or siRNA targeting multi-drug resistant gene (p-glycoprotein) in late-stage metastatic orthotopic KPC-Luc pancreas cancer, 4T1-Luc2 triple negative breast cancer, lung inflammation, and CT26 colorectal cancer animal models in female mice compared to respective FDA-approved nanotherapeutics or lipid compositions. Thus, SM-Chol represents a promising platform for universal and improved drug delivery.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jonghan Park
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Xiangmeng Wu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Qing-Yu Zhang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, USA.
- Clinical and Translational Oncology Program (CTOP), The University of Arizona Cancer Center, Tucson, AZ, 85721, USA.
- BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
43
|
Krishnan N, Jiang Y, Zhou J, Mohapatra A, Peng FX, Duan Y, Holay M, Chekuri S, Guo Z, Gao W, Fang RH, Zhang L. A modular approach to enhancing cell membrane-coated nanoparticle functionality using genetic engineering. NATURE NANOTECHNOLOGY 2024; 19:345-353. [PMID: 37903891 PMCID: PMC10954421 DOI: 10.1038/s41565-023-01533-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 09/22/2023] [Indexed: 11/01/2023]
Abstract
Since their initial development, cell membrane-coated nanoparticles (CNPs) have become increasingly popular in the biomedical field. Despite their inherent versatility and ability to enable complex biological applications, there is considerable interest in augmenting the performance of CNPs through the introduction of additional functionalities. Here we demonstrate a genetic-engineering-based modular approach to CNP functionalization that can encompass a wide range of ligands onto the nanoparticle surface. The cell membrane coating is engineered to express a SpyCatcher membrane anchor that can readily form a covalent bond with any moiety modified with SpyTag. To demonstrate the broad utility of this technique, three unique targeted CNP formulations are generated using different classes of targeting ligands, including a designed ankyrin repeat protein, an affibody and a single-chain variable fragment. In vitro, the modified nanoparticles exhibit enhanced affinity towards cell lines overexpressing the cognate receptors for each ligand. When formulated with a chemotherapeutic payload, the modularly functionalized nanoparticles display strong targeting ability and growth suppression in a murine tumour xenograft model of ovarian cancer. Our data suggest genetic engineering offers a feasible approach for accelerating the development of multifunctional CNPs for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Animesh Mohapatra
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Fei-Xing Peng
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Yaou Duan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sanam Chekuri
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Zhongyuan Guo
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
44
|
Puccetti M, Pariano M, Schoubben A, Giovagnoli S, Ricci M. Biologics, theranostics, and personalized medicine in drug delivery systems. Pharmacol Res 2024; 201:107086. [PMID: 38295917 DOI: 10.1016/j.phrs.2024.107086] [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: 12/26/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
The progress in human disease treatment can be greatly advanced through the implementation of nanomedicine. This approach involves targeted and cell-specific therapy, controlled drug release, personalized dosage forms, wearable drug delivery, and companion diagnostics. By integrating cutting-edge technologies with drug delivery systems, greater precision can be achieved at the tissue and cellular levels through the use of stimuli-responsive nanoparticles, and the development of electrochemical sensor systems. This precision targeting - by virtue of nanotechnology - allows for therapy to be directed specifically to affected tissues while greatly reducing side effects on healthy tissues. As such, nanomedicine has the potential to transform the treatment of conditions such as cancer, genetic diseases, and chronic illnesses by facilitating precise and cell-specific drug delivery. Additionally, personalized dosage forms and wearable devices offer the ability to tailor treatment to the unique needs of each patient, thereby increasing therapeutic effectiveness and compliance. Companion diagnostics further enable efficient monitoring of treatment response, enabling customized adjustments to the treatment plan. The question of whether all the potential therapeutic approaches outlined here are viable alternatives to current treatments is also discussed. In general, the application of nanotechnology in the field of biomedicine may provide a strong alternative to existing treatments for several reasons. In this review, we aim to present evidence that, although in early stages, fully merging advanced technology with innovative drug delivery shows promise for successful implementation across various disease areas, including cancer and genetic or chronic diseases.
Collapse
Affiliation(s)
- Matteo Puccetti
- Department of Pharmaceutical Sciences, University of Perugia, Italy,.
| | | | | | | | - Maurizio Ricci
- Department of Pharmaceutical Sciences, University of Perugia, Italy,.
| |
Collapse
|
45
|
Guan X, Xing S, Liu Y. Engineered Cell Membrane-Camouflaged Nanomaterials for Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:413. [PMID: 38470744 DOI: 10.3390/nano14050413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Recent strides in nanomaterials science have paved the way for the creation of reliable, effective, highly accurate, and user-friendly biomedical systems. Pioneering the integration of natural cell membranes into sophisticated nanocarrier architectures, cell membrane camouflage has emerged as a transformative approach for regulated drug delivery, offering the benefits of minimal immunogenicity coupled with active targeting capabilities. Nevertheless, the utility of nanomaterials with such camouflage is curtailed by challenges like suboptimal targeting precision and lackluster therapeutic efficacy. Tailored cell membrane engineering stands at the forefront of biomedicine, equipping nanoplatforms with the capacity to conduct more complex operations. This review commences with an examination of prevailing methodologies in cell membrane engineering, spotlighting strategies such as direct chemical modification, lipid insertion, membrane hybridization, metabolic glycan labeling, and genetic engineering. Following this, an evaluation of the unique attributes of various nanomaterials is presented, delivering an in-depth scrutiny of the substantial advancements and applications driven by cutting-edge engineered cell membrane camouflage. The discourse culminates by recapitulating the salient influence of engineered cell membrane camouflage within nanomaterial applications and prognosticates its seminal role in transformative healthcare technologies. It is envisaged that the insights offered herein will catalyze novel avenues for the innovation and refinement of engineered cell membrane camouflaged nanotechnologies.
Collapse
Affiliation(s)
- Xiyuan Guan
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Kay Lab of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Simin Xing
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Kay Lab of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yang Liu
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Kay Lab of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
46
|
Deng R, Zhao R, Zhang Z, Chen Y, Yang M, Lin Y, Ye J, Li N, Qin H, Yan X, Shi J, Yuan F, Song S, Xu Z, Song Y, Fu J, Xu B, Nie G, Yu JK. Chondrocyte membrane-coated nanoparticles promote drug retention and halt cartilage damage in rat and canine osteoarthritis. Sci Transl Med 2024; 16:eadh9751. [PMID: 38381849 DOI: 10.1126/scitranslmed.adh9751] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by progressive degeneration of articular cartilage. A challenge in the development of disease-modifying drugs is effective delivery to chondrocytes. The unique structure of the joint promotes rapid clearance of drugs through synovial fluid, and the dense and avascular cartilage extracellular matrix (ECM) limits drug penetration. Here, we show that poly(lactide-co-glycolic acid) nanoparticles coated in chondrocyte membranes (CM-NPs) were preferentially taken up by rat chondrocytes ex vivo compared with uncoated nanoparticles. Internalization of the CM-NPs was mediated primarily by E-cadherin, clathrin-mediated endocytosis, and micropinocytosis. These CM-NPs adhered to the cartilage ECM in rat knee joints in vivo and penetrated deeply into the cartilage matrix with a residence time of more than 34 days. Simulated synovial fluid clearance studies showed that CM-NPs loaded with a Wnt pathway inhibitor, adavivint (CM-NPs-Ada), delayed the catabolic metabolism of rat and human chondrocytes and cartilage explants under inflammatory conditions. In a surgical model of rat OA, drug-loaded CM-NPs effectively restored gait, attenuated periarticular bone remodeling, and provided chondroprotection against cartilage degeneration. OA progression was also mitigated by CM-NPs-Ada in a canine model of anterior cruciate ligament transection. These results demonstrate the feasibility of using chondrocyte membrane-coated nanoparticles to improve the pharmacokinetics and efficacy of anti-OA drugs.
Collapse
Affiliation(s)
- Ronghui Deng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Zining Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Meng Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Ye
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xin Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
| | - Fuzhen Yuan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Shitang Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Zijie Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Yifan Song
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Jiangnan Fu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Bingbing Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jia-Kuo Yu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, P. R. China
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, P. R. China
- Orthopedic Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Affiliated Hospital of Tsinghua University, Beijing 102218, P. R. China
| |
Collapse
|
47
|
Singh R, Kumawat M, Gogoi H, Madhyastha H, Lichtfouse E, Daima HK. Engineered Nanomaterials for Immunomodulation: A Review. ACS APPLIED BIO MATERIALS 2024; 7:727-751. [PMID: 38166376 DOI: 10.1021/acsabm.3c00940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
The immune system usually provides a defense against invading pathogenic microorganisms and any other particulate contaminants. Nonetheless, it has been recently reported that nanomaterials can evade the immune system and modulate immunological responses due to their unique physicochemical characteristics. Consequently, nanomaterial-based activation of immune components, i.e., neutrophils, macrophages, and other effector cells, may induce inflammation and alter the immune response. Here, it is essential to distinguish the acute and chronic modulations triggered by nanomaterials to determine the possible risks to human health. Nanomaterials size, shape, composition, surface charge, and deformability are factors controlling their uptake by immune cells and the resulting immune responses. The exterior corona of molecules adsorbed over nanomaterials surfaces also influences their immunological effects. Here, we review current nanoengineering trends for targeted immunomodulation with an emphasis on the design, safety, and potential toxicity of nanomaterials. First, we describe the characteristics of engineered nanomaterials that trigger immune responses. Then, the biocompatibility and immunotoxicity of nanoengineered particles are debated, because these factors influence applications. Finally, future nanomaterial developments in terms of surface modifications, synergistic approaches, and biomimetics are discussed.
Collapse
Affiliation(s)
| | - Mamta Kumawat
- Department of Biotechnology, School of Sciences, JECRC University, Sitapura Extension, Jaipur 303905, Rajasthan, India
| | - Himanshu Gogoi
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, University of Miyazaki, Miyazaki 8891692, Japan
| | - Eric Lichtfouse
- State Key Laboratory of Multiphase Flow in Power Engineering, Xi'an Jiaotong University Xi'an, Shaanxi 710049, China
| | - Hemant Kumar Daima
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindari 305817, Ajmer, India
| |
Collapse
|
48
|
Fernández-Borbolla A, García-Hevia L, Fanarraga ML. Cell Membrane-Coated Nanoparticles for Precision Medicine: A Comprehensive Review of Coating Techniques for Tissue-Specific Therapeutics. Int J Mol Sci 2024; 25:2071. [PMID: 38396747 PMCID: PMC10889273 DOI: 10.3390/ijms25042071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Nanoencapsulation has become a recent advancement in drug delivery, enhancing stability, bioavailability, and enabling controlled, targeted substance delivery to specific cells or tissues. However, traditional nanoparticle delivery faces challenges such as a short circulation time and immune recognition. To tackle these issues, cell membrane-coated nanoparticles have been suggested as a practical alternative. The production process involves three main stages: cell lysis and membrane fragmentation, membrane isolation, and nanoparticle coating. Cell membranes are typically fragmented using hypotonic lysis with homogenization or sonication. Subsequent membrane fragments are isolated through multiple centrifugation steps. Coating nanoparticles can be achieved through extrusion, sonication, or a combination of both methods. Notably, this analysis reveals the absence of a universally applicable method for nanoparticle coating, as the three stages differ significantly in their procedures. This review explores current developments and approaches to cell membrane-coated nanoparticles, highlighting their potential as an effective alternative for targeted drug delivery and various therapeutic applications.
Collapse
Affiliation(s)
- Andrés Fernández-Borbolla
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Lorena García-Hevia
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Mónica L. Fanarraga
- The Nanomedicine Group, Institute Valdecilla-IDIVAL, 39011 Santander, Spain; (A.F.-B.); (L.G.-H.)
- Molecular Biology Department, Faculty of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| |
Collapse
|
49
|
Wang X, Jia B, Lee K, Davis B, Wen C, Wang Y, Zheng H, Wang Y. Biomimetic Bacterial Capsule for Enhanced Aptamer Display and Cell Recognition. J Am Chem Soc 2024; 146:868-877. [PMID: 38153404 DOI: 10.1021/jacs.3c11208] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Great effort has been made to encapsulate or coat living mammalian cells for a variety of applications ranging from diabetes treatment to three-dimensional printing. However, no study has reported the synthesis of a biomimetic bacterial capsule to display high-affinity aptamers on the cell surface for enhanced cell recognition. Therefore, we synthesized an ultrathin alginate-polylysine coating to display aptamers on the surface of living cells with natural killer (NK) cells as a model. The results show that this coating-mediated aptamer display is more stable than direct cholesterol insertion into the lipid bilayer. The half-life of the aptamer on the cell surface can be increased from less than 1.5 to over 20 h. NK cells coated with the biomimetic bacterial capsule exhibit a high efficiency in recognizing and killing target cells. Therefore, this work has demonstrated a promising cell coating method for the display of aptamers for enhanced cell recognition.
Collapse
Affiliation(s)
- Xuelin Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Bei Jia
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Kyungsene Lee
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Brandon Davis
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Yixun Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Hong Zheng
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
50
|
Gao C, Wang Q, Ding Y, Kwong CHT, Liu J, Xie B, Wei J, Lee SMY, Mok GSP, Wang R. Targeted therapies of inflammatory diseases with intracellularly gelated macrophages in mice and rats. Nat Commun 2024; 15:328. [PMID: 38184609 PMCID: PMC10771422 DOI: 10.1038/s41467-023-44662-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/28/2023] [Indexed: 01/08/2024] Open
Abstract
Membrane-camouflaged nanomedicines often suffer from reduced efficacy caused by membrane protein disintegration and spatial disorder caused by separation and reassembly of membrane fragments during the coating process. Here we show that intracellularly gelated macrophages (GMs) preserve cell membrane structures, including protein content, integration and fluidity, as well as the membrane lipid order. Consequently, in our testing GMs act as cellular sponges to efficiently neutralize various inflammatory cytokines via receptor-ligand interactions, and serve as immune cell-like carriers to selectively bind inflammatory cells in culture medium, even under a flow condition. In a rat model of collagen-induced arthritis, GMs alleviate the joint injury, and suppress the overall arthritis severity. Upon intravenous injection, GMs efficiently accumulate in the inflammatory lungs of acute pneumonia mice for anti-inflammatory therapy. Conveniently, GMs are amenable to lyophilization and can be stored at ambient temperatures for at least 1 month without loss of integrity and bio-activity. This intracellular gelation technology provides a universal platform for targeted inflammation neutralization treatment.
Collapse
Affiliation(s)
- Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, 999078, China
| | - Qingfu Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
| | - Yuanfu Ding
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
- Biomedical Imaging Laboratory (BIG), Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao, 999078, China
| | - Cheryl H T Kwong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
| | - Jinwei Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
| | - Beibei Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
| | - Jianwen Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
| | - Simon M Y Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, 999078, China
| | - Greta S P Mok
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, 999078, China
- Biomedical Imaging Laboratory (BIG), Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao, 999078, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, 999078, China.
| |
Collapse
|