1
|
Markova E, Wolowczyk C, Mohamed A, Sofias AM, Martin-Armas M, Sundset R, Berndtsson J, Hak S, Škalko-Basnet N. Liposomal Nω-hydroxy-l-norarginine, a proof-of-concept: Arginase inhibitors can be incorporated in liposomes while retaining their therapeutic activity ex vivo. Eur J Pharm Sci 2024; 204:106959. [PMID: 39521192 DOI: 10.1016/j.ejps.2024.106959] [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: 06/07/2024] [Revised: 10/02/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Cancer immunotherapy has evolved significantly over the last decade, with therapeutics targeting the adaptive immune system showing exciting effects in clinics. Yet, the modulation of the innate immune system, particularly the tumor-associated innate immune cells which are an integral part of immune responses in cancer, remains less understood. The arginase 1 (Arg1) pathway is a pivotal metabolic pathway that tumor-associated innate immune cells exploit to create an immunosuppressive tumor microenvironment, leading to the evasion of immune surveillance. The inhibition of Arg1 presents a therapeutic opportunity to reverse this immunosuppression, and Nω‑hydroxy-l-norarginine (nor-NOHA) has emerged as a potent arginase inhibitor with promising in vivo efficacy. However, the rapid systemic clearance of nor-NOHA poses a significant challenge for its therapeutic application. This study pioneers the encapsulation of nor-NOHA in liposomes, aiming to enhance its bioavailability and prolong its inhibitory activity against Arg1. Historically, the extensive interaction between innate immune cells and nanoparticles has been one of the biggest drawbacks in nanomedicine. Here we seek to utilize this effect and deliver liposomal nor-NOHA to the arginase 1 expressing innate immune cells. We systematically investigated the effect of lipid composition, acyl chain length, manufacturing and loading methodology on the encapsulation efficiency (EE%) and release profile of nor-NOHA. Our results indicate that while the manufacturing method and lipid acyl chain length do not significantly impact EE%, they crucially influence the release kinetics of nor-NOHA, with longer acyl chains demonstrating a more sustained release of nor-NOHA from liposomes enabling continuous inhibition of Arg1. Our findings suggest that liposomal nor-NOHA retains its functional inhibitory activity and could offer improved pharmacokinetic properties, making it a compelling base for iterations for further innovative cancer immunotherapeutic strategies in preclinical and clinical evaluations.
Collapse
Affiliation(s)
- Elena Markova
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway.
| | - Camilla Wolowczyk
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences & Centre of Molecular Inflammation Research (CEMIR), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Aly Mohamed
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Montserrat Martin-Armas
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Rune Sundset
- Nuclear Medicine and Radiation Biology Research Group, Department of Clinical Medicine, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway; PET Imaging Center Tromsø, University Hospital of North Norway (UNN), Tromsø, Norway
| | - Jens Berndtsson
- Centre for Cellular Imaging, Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sjoerd Hak
- Department of Biotechnology and Nanomedicine, SINTEF Industry & Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
2
|
Ji P, Xu Q, Li J, Wang Z, Mao W, Yan P. Advances in nanoparticle-based therapeutics for ischemic stroke: Enhancing drug delivery and efficacy. Biomed Pharmacother 2024; 180:117564. [PMID: 39405899 DOI: 10.1016/j.biopha.2024.117564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
Ischemic stroke, characterized by vascular occlusion, has recently emerged as one of the primary causes of mortality and disability worldwide. Conventional treatment modalities, such as thrombolytic and neuroprotective therapies, face numerous challenges, including limited bioavailability, significant neurotoxicity, suboptimal targeting, short half-life, and poor blood-brain barrier (BBB) penetration. Nanoparticle-based drug delivery systems present distinct advantages, such as small size, enhanced lipophilicity, and modifiability, which can potentially address these limitations. Utilizing nanoparticles for drug delivery in ischemic stroke therapy offers improved drug bioavailability, reduced neurotoxicity, enhanced targeted delivery, prolonged drug half-life, and better dissolution kinetics. This review aims to provide a comprehensive overview of current strategies in preclinical studies for managing or preventing ischemic stroke from a nanomaterial perspective, highlighting the advantages and limitations of each approach.
Collapse
Affiliation(s)
- Peng Ji
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou 225300, China; Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Qingqing Xu
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou 225300, China
| | - Jiahui Li
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou 225300, China
| | - Zihan Wang
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou 225300, China
| | - Wanyi Mao
- College of Pharmacy and Chemistry & Chemical Engineering, Taizhou University, Taizhou 225300, China
| | - Peng Yan
- Taizhou Second People's Hospital Affiliated to Yangzhou University, Taizhou 225300, China.
| |
Collapse
|
3
|
Pednekar K, Minnee J, de Vries IJM, Prakash J. Targeted nanomedicine for reprogramming the tumor innate immune system: From bench to bedside. Eur J Pharm Biopharm 2024; 204:114510. [PMID: 39307440 DOI: 10.1016/j.ejpb.2024.114510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Tumor-associated innate immune cells such as tumor-associated macrophages, neutrophils, dendritic cells play a crucial role in tumor progression, angiogenesis and metastasis. These cells also control the efficacy of chemotherapy and immunotherapy by inducing drug resistance and immunosuppression, leading to therapeutic failures. Therefore, targeting the tumor-associated innate immune cells has gained high attention for the development of effective cancer therapy. Nanomedicine based strategies to target these cells are highly relevant and can be used to reprogram these cells. In this review, we discuss the fundamental roles of the tumor-associated innate immune cells in the tumor microenvironment and different strategies to modulate them. Then, nanomedicine-based strategies to target different tumor innate immune cells are explained in detail. While the clinical development of the targeted nanomedicine remains a great challenge in practice, we have provided our perspectives on various factors such as pharmaceutical aspects, preclinical testing and biological aspects which are crucial to consider before translating these targeting strategies to clinics.
Collapse
Affiliation(s)
- Kunal Pednekar
- Engineered Therapeutics, Department of Advanced Organ bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Julia Minnee
- Department of Medical BioSciences (MBS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Medical BioSciences (MBS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jai Prakash
- Engineered Therapeutics, Department of Advanced Organ bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
4
|
Yang Y, Du J, Gan J, Song X, Shu J, An C, Lu L, Wei H, Che J, Zhao X. Neutrophil-Mediated Nanozyme Delivery System for Acute Kidney Injury Therapy. Adv Healthc Mater 2024; 13:e2401198. [PMID: 38899383 DOI: 10.1002/adhm.202401198] [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: 03/31/2024] [Revised: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Reactive oxygen species (ROS) scavenging of nanozymes toward acute kidney injury (AKI) is a current promising strategy, however, the glomerular filtration barrier (GFB) limits their application for treating kidney related diseases. Here, a neutrophil-mediated delivery system able to hijack neutrophil to transport nanozyme-loaded cRGD-liposomes to inflamed kidney for AKI treatment by cRGD targeting integrin αvβ1 is reported. The neutrophil-mediated nanozyme delivery system demonstrated great antioxidant and anti-apoptosis ability in HK-2 and NRK-52E cell lines. Moreover, in ischemia-reperfusion (I/R) induced AKI mice, a single dose of LM@cRGD-LPs 12 h post-ischemia significantly reduces renal function indicators, alleviates renal pathological changes, and inhibits apoptosis of renal tubular cells and the expression of renal tubular injured marker, thus remarkably reducing the damage of AKI. Mechanistically, the treatment of LM@cRGD-LPs markedly inhibits the process of Nrf2 to the nucleus and reduces the expression of the downstream HO-1, achieves a 99.51% increase in renal tissue Nrf2 levels, and an 86.31% decrease in HO-1 levels after LM@cRGD-LPs treatment. In short, the strategy of neutrophil-mediated nanozyme delivery system hold great promise as a potential therapy for AKI or other inflammatory diseases.
Collapse
Affiliation(s)
- Yu Yang
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Jiang Du
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jingjing Gan
- Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Xiang Song
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Jiaxin Shu
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
| | - Chaoli An
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Li Lu
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Junyi Che
- Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Xiaozhi Zhao
- Department of Andrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210008, China
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| |
Collapse
|
5
|
Bezze A, Mattioda C, Ciardelli G, Mattu C. Harnessing cells to improve transport of nanomedicines. Eur J Pharm Biopharm 2024; 203:114446. [PMID: 39122052 DOI: 10.1016/j.ejpb.2024.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Efficient tumour treatment is hampered by the poor selectivity of anticancer drugs, resulting in scarce tumour accumulation and undesired off-target effects. Nano-sized drug-delivery systems in the form of nanoparticles (NPs) have been proposed to improve drug distribution to solid tumours, by virtue of their ability of passive and active tumour targeting. Despite these advantages, literature studies indicated that less than 1% of the administered NPs can successfully reach the tumour mass, highlighting the necessity for more efficient drug transporters in cancer treatment. Living cells, such as blood cells, circulating immune cells, platelets, and stem cells, are often found as an infiltrating component in most solid tumours, because of their ability to naturally circumvent immune recognition, bypass biological barriers, and reach inaccessible tissues through innate tropism and active motility. Therefore, the tumour-homing ability of these cells can be harnessed to design living cell carriers able to improve the transport of drugs and NPs to tumours. Albeit promising, this approach is still in its beginnings and suffers from difficult scalability, high cost, and poor reproducibility. In this review, we present an overview of the most common cell transporters of drugs and NPs, and we discuss how different cell types interact with biological barriers to deliver cargoes of various natures to tumours. Finally, we analyse the different techniques used to load drugs or NPs in living cells and discuss their advantages and disadvantages.
Collapse
Affiliation(s)
- Andrea Bezze
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Carlotta Mattioda
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Gianluca Ciardelli
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Clara Mattu
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
6
|
Liu W, Cheng G, Cui H, Tian Z, Li B, Han Y, Wu JX, Sun J, Zhao Y, Chen T, Yu G. Theoretical basis, state and challenges of living cell-based drug delivery systems. Theranostics 2024; 14:5152-5183. [PMID: 39267776 PMCID: PMC11388066 DOI: 10.7150/thno.99257] [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: 06/04/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
The therapeutic efficacy of drugs is determined, to a certain extent, by the efficiency of drug delivery. The low efficiency of drug delivery systems (DDSs) is frequently associated with serious toxic side effects and can even prove fatal in certain cases. With the rapid development of technology, drug delivery has evolved from using traditional frameworks to using nano DDSs (NDDSs), endogenous biomaterials DDSs (EBDDSs), and living cell DDSs (LCDDSs). LCDDSs are receiving widespread attention from researchers at present owing to the unique advantages of living cells in targeted drug delivery, including their excellent biocompatibility properties, low immunogenicity, unique biological properties and functions, and role in the treatment of diseases. However, the theoretical basis and techniques involved in the application of LCDDSs have not been extensively summarized to date. Therefore, this review comprehensively summarizes the properties and applications of living cells, elaborates the various drug loading approaches and controlled drug release, and discusses the results of clinical trials. The review also discusses the current shortcomings and prospects for the future development of LCDDSs, which will serve as highly valuable insights for the development and clinical transformation of LCDDSs in the future.
Collapse
Affiliation(s)
- Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Guowang Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Bowen Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yanhua Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jia-Xin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jie Sun
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Yuyue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Guangtao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
7
|
Tong S, Niu J, Wang Z, Jiao Y, Fu Y, Li D, Pan X, Sheng N, Yan L, Min P, Chen D, Cui S, Liu Y, Lin S. The Evolution of Microfluidic-Based Drug-Loading Techniques for Cells and Their Derivatives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2403422. [PMID: 39152940 DOI: 10.1002/smll.202403422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/03/2024] [Indexed: 08/19/2024]
Abstract
Conventional drug delivery techniques face challenges related to targeting and adverse reactions. Recent years have witnessed significant advancements in nanoparticle-based drug carriers. Nevertheless, concerns persist regarding their safety and insufficient metabolism. Employing cells and their derivatives, such as cell membranes and extracellular vesicles (EVs), as drug carriers effectively addresses the challenges associated with nanoparticle carriers. However, an essential hurdle remains in efficiently loading drugs into these carriers. With the advancement of microfluidic technology and its advantages in precise manipulation at the micro- and nanoscales, as well as minimal sample loss, it has found extensive application in the loading of drugs using cells and their derivatives, thereby fostering the development of drug-loading techniques. This paper outlines the characteristics and benefits of utilizing cells and their derivatives as drug carriers and provides an overview of current drug-loading techniques, particularly those rooted in microfluidic technology. The significant potential for microfluidic technology in targeted disease therapy through drug delivery systems employing cells and their derivatives, is foreseen.
Collapse
Affiliation(s)
- Siyu Tong
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiaqi Niu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhitao Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yingao Jiao
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yanfei Fu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Dongxia Li
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinni Pan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Nengquan Sheng
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Li Yan
- Department of Geriatric Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Peiru Min
- Shanghai 9th People's Hospital, Shanghai Jiao Tong University College of Medicine, Shanghai, 200240, China
| | - Di Chen
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shengsheng Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Yanlei Liu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shujing Lin
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
8
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024:10.1038/s41551-024-01230-6. [PMID: 38951139 DOI: 10.1038/s41551-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/01/2024] [Indexed: 07/03/2024]
Abstract
By combining living cells with therapeutics, cell-drug conjugates can potentiate the functions of both components, particularly for applications in drug delivery and therapy. The conjugates can be designed to persist in the bloodstream, undergo chemotaxis, evade surveillance by the immune system, proliferate, or maintain or transform their cellular phenotypes. In this Review, we discuss strategies for the design of cell-drug conjugates with specific functions, the techniques for their preparation, and their applications in the treatment of cancers, autoimmune diseases and other pathologies. We also discuss the translational challenges and opportunities of this class of drug-delivery systems and therapeutics.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiaqi Shi
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Minhang Xin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Cai J, Lin Y, Zhou B, Xiao F, Xu G, Lu J. SHARPIN contributes to sevoflurane-induced neonatal neurotoxicity through up-regulating HMGB1 to repress M2 like-macrophage polarization. Metab Brain Dis 2024; 39:841-853. [PMID: 38805141 DOI: 10.1007/s11011-024-01355-2] [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: 08/14/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Sevoflurane exposure can result in neurotoxicity especially among children, which remains an important complication after surgery. However, its related mechanisms remain unclear. Here, we investigated the biological roles of SHARPIN in sevoflurane-induced neurotoxicity. As detected by qPCR, Western blotting and immunohistochemical staining, SHARPIN and HMGB1 expression was elevated in sevoflurane-stimulated mice as compared with the control mice. SHARPIN depletion attenuated hippocampus injury, repressed the expression of HMGB1 and M1-like macrophage markers (iNOS, TNF-α, IL-1β, IL-6), but enhanced the expression of M2-like macrophage markers (ARG-1, IL-10). GST pull-down and Co-IP assays demonstrated that SHARPIN directly interacted with HMGB1 to enhance HMGB1 expression in SH-SY5Y cells. The inhibitory effects of SHARPIN silencing on inflammatory reaction and M1-like macrophages were counteracted by HMGB1 overexpression. Finally, SHARPIN-HMGB1 pathway affected neuroinflammation triggered by sevoflurane via modulating macrophage polarization. Collectively, our data suggested that SHARPIN stimulated sevoflurane-induced neurotoxicity via converting M2-like macrophages to M1-like macrophages by enhancing HMGB1 expression. SHARPIN intervention may be a promising therapeutic method to relieve sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Junying Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jun Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
10
|
Asimakidou E, Tan JKS, Zeng J, Lo CH. Blood-Brain Barrier-Targeting Nanoparticles: Biomaterial Properties and Biomedical Applications in Translational Neuroscience. Pharmaceuticals (Basel) 2024; 17:612. [PMID: 38794182 PMCID: PMC11123901 DOI: 10.3390/ph17050612] [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: 03/26/2024] [Revised: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Overcoming the blood-brain barrier (BBB) remains a significant hurdle in effective drug delivery to the brain. While the BBB serves as a crucial protective barrier, it poses challenges in delivering therapeutic agents to their intended targets within the brain parenchyma. To enhance drug delivery for the treatment of neurological diseases, several delivery technologies to circumvent the BBB have been developed in the last few years. Among them, nanoparticles (NPs) are one of the most versatile and promising tools. Here, we summarize the characteristics of NPs that facilitate BBB penetration, including their size, shape, chemical composition, surface charge, and importantly, their conjugation with various biological or synthetic molecules such as glucose, transferrin, insulin, polyethylene glycol, peptides, and aptamers. Additionally, we discuss the coating of NPs with surfactants. A comprehensive overview of the common in vitro and in vivo models of the BBB for NP penetration studies is also provided. The discussion extends to discussing BBB impairment under pathological conditions and leveraging BBB alterations under pathological conditions to enhance drug delivery. Emphasizing the need for future studies to uncover the inherent therapeutic properties of NPs, the review advocates for their role beyond delivery systems and calls for efforts translating NPs to the clinic as therapeutics. Overall, NPs stand out as a highly promising therapeutic strategy for precise BBB targeting and drug delivery in neurological disorders.
Collapse
Affiliation(s)
- Evridiki Asimakidou
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK;
| | - Justin Kok Soon Tan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore;
- The N.1 Institute for Health, National University of Singapore, Singapore 117456, Singapore
| | - Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
11
|
Yuan S, Hu Q. Convergence of nanomedicine and neutrophils for drug delivery. Bioact Mater 2024; 35:150-166. [PMID: 38318228 PMCID: PMC10839777 DOI: 10.1016/j.bioactmat.2024.01.022] [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: 10/31/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
Neutrophils have recently emerged as promising carriers for drug delivery due to their unique properties including rapid response toward inflammation, chemotaxis, and transmigration. When integrated with nanotechnology that has enormous advantages in improving treatment efficacy and reducing side effects, neutrophil-based nano-drug delivery systems have expanded the repertoire of nanoparticles employed in precise therapeutic interventions by either coating nanoparticles with their membranes, loading nanoparticles inside living cells, or engineering chimeric antigen receptor (CAR)-neutrophils. These neutrophil-inspired therapies have shown superior biocompatibility, targeting ability, and therapeutic robustness. In this review, we summarized the benefits of combining neutrophils and nanotechnologies, the design principles and underlying mechanisms, and various applications in disease treatments. The challenges and prospects for neutrophil-based drug delivery systems were also discussed.
Collapse
Affiliation(s)
- Sichen Yuan
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| |
Collapse
|
12
|
Liu X, Ou X, Zhang T, Li X, Qiao Q, Jia L, Xu Z, Zhang F, Tian T, Lan H, Yang C, Kong L, Zhang Z. In situ neutrophil apoptosis and macrophage efferocytosis mediated by Glycyrrhiza protein nanoparticles for acute inflammation therapy. J Control Release 2024; 369:215-230. [PMID: 38508529 DOI: 10.1016/j.jconrel.2024.03.029] [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: 12/25/2023] [Revised: 02/28/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
In the progression of acute inflammation, the activation and recruitment of macrophages and neutrophils are mutually reinforcing, leading to amplified inflammatory response and severe tissue damage. Therefore, to regulate the axis of neutrophils and macrophages is essential to avoid tissue damage induced from acute inflammatory. Apoptotic neutrophils can regulate the anti-inflammatory activity of macrophages through the efferocytosis. The strategy of in situ targeting and inducing neutrophil apoptosis has the potential to modulate macrophage activity and transfer anti-inflammatory drugs. Herein, a natural glycyrrhiza protein nanoparticle loaded with dexamethasone (Dex@GNPs) was constructed, which could simultaneously regulate neutrophil and macrophage function during acute inflammation treatment by combining in situ neutrophil apoptosis and macrophage efferocytosis. Dex@GNPs can be rapidly and selectively internalized by neutrophils and subsequently induce neutrophils apoptosis through a ROS-dependent mechanism. The efferocytosis of apoptotic neutrophils not only promoted the polarization of macrophages into anti-inflammatory state, but also facilitated the transfer of Dex@GNPs to macrophages. This enabled dexamethasone to further modulate macrophage function. In mouse models of acute respiratory distress syndrome and sepsis, Dex@GNPs significantly ameliorated the disordered immune microenvironment and alleviated tissue injury. This study presents a novel strategy for drug delivery and inflammation regulation to effectively treat acute inflammatory diseases.
Collapse
Affiliation(s)
- Xiong Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangjun Ou
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tiantian Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaonan Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Qiao
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liyuan Jia
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhangxi Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fangming Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianyi Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbing Lan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
13
|
Cao Y, Yu Y, Pan L, Han W, Zeng F, Wang J, Mei Q, Liu C. Sulfated Polysaccharide-Based Nanocarrier Drives Microenvironment-Mediated Cerebral Neurovascular Remodeling for Ischemic Stroke Treatment. NANO LETTERS 2024; 24:5214-5223. [PMID: 38649327 DOI: 10.1021/acs.nanolett.4c00650] [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: 04/25/2024]
Abstract
Stroke is a leading cause of global mortality and severe disability. However, current strategies used for treating ischemic stroke lack specific targeting capabilities, exhibit poor immune escape ability, and have limited drug release control. Herein, we developed an ROS-responsive nanocarrier for targeted delivery of the neuroprotective agent rapamycin (RAPA) to mitigate ischemic brain damage. The nanocarrier consisted of a sulfated chitosan (SCS) polymer core modified with a ROS-responsive boronic ester enveloped by a red blood cell membrane shell incorporating a stroke homing peptide. When encountering high levels of intracellular ROS in ischemic brain tissues, the release of SCS combined with RAPA from nanoparticle disintegration facilitates effective microglia polarization and, in turn, maintains blood-brain barrier integrity, reduces cerebral infarction, and promotes cerebral neurovascular remodeling in a mouse stroke model involving transient middle cerebral artery occlusion (tMCAO). This work offers a promising strategy to treat ischemic stroke therapy.
Collapse
Affiliation(s)
- Yinli Cao
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Lina Pan
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Weili Han
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Feng Zeng
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, People's Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Qiyong Mei
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, People's Republic of China
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| |
Collapse
|
14
|
Zhou A, Kong D, Zhou X, Liu Y, Zhang Y, Li J, Xu Y, Ning X. Bioengineered Neutrophils for Smart Response in Brain Infection Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311661. [PMID: 38252744 DOI: 10.1002/adma.202311661] [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: 11/04/2023] [Revised: 01/09/2024] [Indexed: 01/24/2024]
Abstract
Brain infections, frequently accompanied by significant inflammation, necessitate comprehensive therapeutic approaches targeting both infections and associated inflammation. A major impediment to such combined treatment is the blood-brain barrier (BBB), which significantly restricts therapeutic agents from achieving effective concentrations within the central nervous system. Here, a neutrophil-centric dual-responsive delivery system, coined "CellUs," is pioneered. This system is characterized by live neutrophils enveloping liposomes of dexamethasone, ceftriaxone, and oxygen-saturated perfluorocarbon (Lipo@D/C/P). CellUs is meticulously engineered to co-deliver antibiotics, anti-inflammatory agents, and oxygen, embodying a comprehensive strategy against brain infections. CellUs leverages the intrinsic abilities of neutrophils to navigate through BBB, accurately target infection sites, and synchronize the release of Lipo@D/C/P with local inflammatory signals. Notably, the incorporation of ultrasound-responsive perfluorocarbon within Lipo@D/C/P ensures the on-demand release of therapeutic agents at the afflicted regions. CellUs shows considerable promise in treating Staphylococcus aureus infections in mice with meningitis, particularly when combined with ultrasound treatments. It effectively penetrates BBB, significantly eliminates bacteria, reduces inflammation, and delivers oxygen to the affected brain tissue, resulting in a substantial improvement in survival rates. Consequently, CellUs harnesses the natural chemotactic properties of neutrophils and offers an innovative pathway to improve treatment effectiveness while minimizing adverse effects.
Collapse
Affiliation(s)
- Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing, 210093, China
| | - Delian Kong
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211000, China
| | - Xinyuan Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Yao Liu
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211000, China
| | - Yiping Zhang
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Junrong Li
- Department of Neurology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211000, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
15
|
Zhang Q, Huang S, Liu X, Wang W, Zhu Z, Chen L. Innovations in Breaking Barriers: Liposomes as Near-Perfect Drug Carriers in Ischemic Stroke Therapy. Int J Nanomedicine 2024; 19:3715-3735. [PMID: 38681090 PMCID: PMC11046314 DOI: 10.2147/ijn.s462194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024] Open
Abstract
Liposomes, noted for their tunable particle size, surface customization, and varied drug delivery capacities, are increasingly acknowledged in therapeutic applications. These vesicles exhibit surface flexibility, enabling the incorporation of targeting moieties or peptides to achieve specific targeting and avoid lysosomal entrapment. Internally, their adaptable architecture permits the inclusion of a broad spectrum of drugs, contingent on their solubility characteristics. This study thoroughly reviews liposome fabrication, surface modifications, and drug release mechanisms post-systemic administration, with a particular emphasis on drugs crossing the blood-brain barrier (BBB) to address lesions. Additionally, the review delves into recent developments in the use of liposomes in ischemic stroke models, offering a comparative evaluation with other nanocarriers like exosomes and nano-micelles, thereby facilitating their clinical advancement.
Collapse
Affiliation(s)
- Qiankun Zhang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Songze Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiaowen Liu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhihan Zhu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
16
|
Zhang SS, Li RQ, Chen Z, Wang XY, Dumont AS, Fan X. Immune cells: potential carriers or agents for drug delivery to the central nervous system. Mil Med Res 2024; 11:19. [PMID: 38549161 PMCID: PMC10979586 DOI: 10.1186/s40779-024-00521-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
Drug delivery systems (DDS) have recently emerged as a promising approach for the unique advantages of drug protection and targeted delivery. However, the access of nanoparticles/drugs to the central nervous system (CNS) remains a challenge mainly due to the obstruction from brain barriers. Immune cells infiltrating the CNS in the pathological state have inspired the development of strategies for CNS foundation drug delivery. Herein, we outline the three major brain barriers in the CNS and the mechanisms by which immune cells migrate across the blood-brain barrier. We subsequently review biomimetic strategies utilizing immune cell-based nanoparticles for the delivery of nanoparticles/drugs to the CNS, as well as recent progress in rationally engineering immune cell-based DDS for CNS diseases. Finally, we discuss the challenges and opportunities of immune cell-based DDS in CNS diseases to promote their clinical development.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Ruo-Qi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Zhong Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiao-Ying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA
| | - Aaron S Dumont
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, 70122, USA.
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
17
|
Yu Y, Wang LY, Liu YC, Cui H, Yuan C, Wang CX. Acetylcholine Analog-Modified Albumin Nanoparticles for the Enhanced and Synchronous Brain Delivery of Saponin Components of Panax Notoginseng. Pharm Res 2024; 41:513-529. [PMID: 38383935 DOI: 10.1007/s11095-024-03670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/28/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Panax notoginseng saponins (PNS) are commonly used first-line drugs for treating cerebral thrombosis and stroke in China. However, the synchronized and targeted delivery of active ingredients in traditional Chinese medicine (TCM) poses a significant challenge for modern TCM formulations. METHODS Bovine serum albumin (BSA) was modified using 2-methacryloyloxyethyl phosphorylcholine (MPC), an analog of acetylcholine, and subsequently adsorbed the major PNS onto the modified albumin to produce MPC-BSA@PNS nanoparticles (NPs). This novel delivery system facilitated efficient and synchronized transport of PNS across the blood-brain barrier (BBB) through active transport mediated by nicotinic acetylcholine receptors. RESULTS In vitro experiments demonstrated that the transport rates of R1, Rg1, Rb1, and Rd across the BBB were relatively synchronous in MPC-BSA@PNS NPs compared to those in the PNS solution. Additionally, animal experiments revealed that the brain-targeting efficiencies of R1 + Rg1 + Rb1 in MPC-BSA@PNS NPs were 2.02 and 7.73 times higher than those in BSA@PNS NPs and the free PNS group, respectively. CONCLUSIONS This study presents a simple and feasible approach for achieving the targeted delivery of complex active ingredient clusters in TCM.
Collapse
Affiliation(s)
- Ying Yu
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming, 650500, China
| | - Li Yun Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming, 650500, China
| | - Yan Chi Liu
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming, 650500, China
| | - Hao Cui
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming, 650500, China
| | - Cheng Yuan
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Cheng Xiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, China.
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming, 650500, China.
| |
Collapse
|
18
|
Wu J, Ma T, Zhu M, Mu J, Huang T, Xu D, Lin N, Gao J. A Pluripotential Neutrophil-Mimic Nanovehicle Modulates Immune Microenvironment with Targeted Drug Delivery for Augmented Antitumor Chemotherapy. ACS NANO 2024. [PMID: 38335121 DOI: 10.1021/acsnano.3c12694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
The limited therapeutic outcomes and severe systemic toxicity of chemotherapy remain major challenges to the current clinical antitumor therapeutic regimen. Tumor-targeted drug delivery that diminishes the undifferentiated systemic distribution is a practical solution to ameliorating systemic toxicity. However, the tumor adaptive immune microenvironment still poses a great threat that compromises the therapeutic efficacy of chemotherapy by promoting the tolerance of the tumor cells. Herein, a pluripotential neutrophil-mimic nanovehicle (Neutrosome(L)) composed of an activated neutrophil membrane-incorporated liposome is proposed to modulate the immune microenvironment and synergize antitumor chemotherapy. The prominent tumor targeting capability inherited from activated neutrophils and the improved tumor penetration ability of Neutrosome(L) enable considerable drug accumulation in tumor tissues (more than sixfold that of free drug). Importantly, Neutrosome(L) can modulate the immune microenvironment by restricting neutrophil infiltration in tumor tissue, which may be attributed to the neutralization of inflammatory cytokines, thus potentiating antitumor chemotherapy. As a consequence, the treatment of cisplatin-loaded Neutrosome(L) performs prominent tumor suppression effects, reduces systemic drug toxicity, and prolongs the survival period of tumor-bearing mice. The pluripotential neutrophil-mimic nanovehicle proposed in this study can not only enhance the tumor accumulation of chemotherapeutics but also modulate the immune microenvironment, providing a compendious strategy for augmented antitumor chemotherapy.
Collapse
Affiliation(s)
- Jiahe Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Teng Ma
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Manning Zhu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiafu Mu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Tianchen Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Donghang Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
- Cancer Center of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Cancer Center of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
19
|
Lin L, Geng D, She D, Kuai X, Du C, Fu P, Zhu Y, Wang J, Pang Z, Zhang J. Targeted nanotheranostics for the treatment of epilepsy through in vivo hijacking of locally activated macrophages. Acta Biomater 2024; 174:314-330. [PMID: 38036284 DOI: 10.1016/j.actbio.2023.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023]
Abstract
Epilepsy refers to a disabling neurological disorder featured by the long-term and unpredictable occurrence of seizures owing to abnormal excessive neuronal electrical activity and is closely linked to unresolved inflammation, oxidative stress, and hypoxia. The difficulty of accurate localization and targeted drug delivery to the lesion hinders the effective treatment of this disease. The locally activated inflammatory cells in the epileptogenic region offer a new opportunity for drug delivery to the lesion. In this work, CD163-positive macrophages in the epileptogenic region were first harnessed as Trojan horses after being hijacked by targeted albumin manganese dioxide nanoparticles, which effectively penetrated the brain endothelial barrier and delivered multifunctional nanomedicines to the epileptic foci. Hence, accumulative nanoparticles empowered the visualization of the epileptogenic lesion through microenvironment-responsive MR T1-weight imaging of manganese dioxide. Besides, these manganese-based nanomaterials played a pivotal role in shielding neurons from cell apoptosis mediated by oxidative stress and hypoxia. Taken together, the present study provides an up-to-date approach for integrated diagnosis and treatment of epilepsy and other hypoxia-associated inflammatory diseases. STATEMENT OF SIGNIFICANCE: The therapeutic effects of antiepileptic drugs (AEDs) are hindered by insufficient drug accumulation in the epileptic site. Herein, we report an efficient strategy to use locally activated macrophages as carriers to deliver multifunctional nanoparticles to the brain lesion. As MR-responsive T1 contrast agents, multifunctional BMC nanoparticles can be harnessed to accurately localize the epileptogenic region with high sensitivity and specificity. Meanwhile, catalytic nanoparticles BMC can synergistically scavenge ROS, generate O2 and regulate neuroinflammation for the protection of neurons in the brain.
Collapse
Affiliation(s)
- Lin Lin
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China; National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Dejun She
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Xinping Kuai
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Chengjuan Du
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Pengfei Fu
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Yuefei Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery Ministry of Education, Shanghai 201203, China
| | - Jianhong Wang
- National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China; Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery Ministry of Education, Shanghai 201203, China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; National Center for Neurological Disorders, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| |
Collapse
|
20
|
Liu Y, Xiang R, Lu W, Qin X. Symptom-oriented network pharmacology revealed the mechanism of HuangQi-DanShen herb pair against cerebral ischemia coupled with comprehensive chemical characterization. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116845. [PMID: 37437791 DOI: 10.1016/j.jep.2023.116845] [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: 05/28/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In the clinical practice of traditional Chinese medicine, HuangQi-DanShen (HD) is an important drug pair for the treatment of cerebral ischemia (CI). AIM OF THE STUDY Elucidate the mechanism of HD against CI based on symptom-oriented network pharmacology coupled with comprehensive chemical characterization. MATERIALS AND METHODS UHPLC-Q-Exactive Orbitrap-MS technology was firstly used to obtain the chemical profile of HD constituents. A comprehensive strategy combining in-house library, diagnostic ions, Compound Discover software and network databases was then established to identify its chemical constitutes. Symptomatic treatment is a treatment aimed at relieving or eliminating symptoms which is often characterized as a stop-gap measure due to its inability to cure the disease fundamentally. Nevertheless, symptomatic treatment is an indispensable part of clinical practice and has an important place in medical therapeutics. Therefore, network pharmacology technique were used to elucidate molecular mechanisms from the symptoms of CI. Finally, some literatures were further mined to support our conclusions. RESULTS A total of 190 ingredients were identified in HD. Symptom-oriented network pharmacology analysis indicated that compounds of HD relieved "blood" through the regulation of ADORA2A, ADORA1, PTPN11, MMP9 and EGFR, relieved "qi" via the regulation of ADORA2A, EGFR, MMP9 and CA2. The therapeutic effect of HD on "faint" was linked to PTPN11 and MMP9, while the regulation of "dyskinesia" was related to ADORA2A and EGFR, and ADORA1, PTPN11 and MMP9 were associated withe its effect on "speech disorder". ADORA1, ADORA2A and MMP9 were key to the HD component in treating "visual disturbance". CONCLUSION The approach of symptom-oriented network pharmacology coupled with comprehensive chemical characterization proposed a further orientation for exploring the mechanisms of HD against CI.
Collapse
Affiliation(s)
- Yuetao Liu
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Ruoxin Xiang
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Wentian Lu
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, No. 92, Wucheng Road, Taiyuan, 030006, Shanxi, PR China.
| |
Collapse
|
21
|
Udofa E, Zhao Z. In situ cellular hitchhiking of nanoparticles for drug delivery. Adv Drug Deliv Rev 2024; 204:115143. [PMID: 38008185 PMCID: PMC10841869 DOI: 10.1016/j.addr.2023.115143] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/04/2023] [Accepted: 11/16/2023] [Indexed: 11/28/2023]
Abstract
Since the inception of the concept of "magic bullet", nanoparticles have evolved to be one of the most effective carriers in drug delivery. Nanoparticles improve the therapeutic efficacy of drugs offering benefits to treating various diseases. Unlike free drugs which freely diffuse and distribute through the body, nanoparticles protect the body from the drug by reducing non-specific interactions while also improving the drug's pharmacokinetics. Despite acquiring some FDA approvals, further clinical application of nanoparticles is majorly hindered by its limited ability to overcome biological barriers resulting in uncontrolled biodistribution and high clearance. The use of cell-inspired systems has emerged as a promising approach to overcome this challenge as cells are biocompatible and have improved access to tissues and organs. One of such is the hitchhiking of nanoparticles to circulating cells such that they are recognized as 'self' components evading clearance and resulting in site-specific drug delivery. In this review, we discuss the concept of nanoparticle cellular hitchhiking, highlighting its advantages, the principles governing the process and the challenges currently limiting its clinical translation. We also discuss in situ hitchhiking as a tool for overcoming these challenges and the considerations to be taken to guide research efforts in advancing this promising technology.
Collapse
Affiliation(s)
- Edidiong Udofa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA.
| |
Collapse
|
22
|
Li S, Zhang R, Wang A, Li Y, Zhang M, Kim J, Zhu Y, Wang Q, Zhang Y, Wei Y, Wang J. Panax notoginseng: derived exosome-like nanoparticles attenuate ischemia reperfusion injury via altering microglia polarization. J Nanobiotechnology 2023; 21:416. [PMID: 37946257 PMCID: PMC10636993 DOI: 10.1186/s12951-023-02161-1] [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: 08/09/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
Cerebral ischemia/reperfusion (CI/R) injury is a clinical conundrum during the treatment of ischemic stroke. Cell-derived exosomes (CDE) were proved to be therapeutically effective for CI/R injury. However, production of CDE is time and effort consuming. Increasing studies reported that plants can also generate exosome-like nanoparticles (ELN) which are therapeutically effective and have higher yield compared with CDE. In this study, a commonly used Chinese herb Panax notoginseng (PN), whose active ingredients were well-documented in the treatment of CI/R injury, was chosen as a source of ELNs. It was found that Panax notoginseng derived exosome like nanoparticles (PDN) could enter the brain without modification and ameliorate cerebral infarct volume, improve behavior outcome and maintained the integrity of BBB. PDNs attenuated CI/R injury by altering the phenotype of microglia from "pro-inflammation" M1 type to "anti-inflammation" M2 type. Also, we found that lipids from PDNs were the major therapeutic effective component. As a mechanism of action, PDN was proved to exert therapeutic effect via activating pI3k/Akt pathway.
Collapse
Affiliation(s)
- Shiyi Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Ru Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Anni Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Yang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Miaomiao Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Jisu Kim
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Ying Zhu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Qizheng Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Yue Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 201203, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China.
- Institutes of Integrative Medicine, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
23
|
Mozafari N, Mozafari N, Dehshahri A, Azadi A. Knowledge Gaps in Generating Cell-Based Drug Delivery Systems and a Possible Meeting with Artificial Intelligence. Mol Pharm 2023; 20:3757-3778. [PMID: 37428824 DOI: 10.1021/acs.molpharmaceut.3c00162] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Cell-based drug delivery systems are new strategies in targeted delivery in which cells or cell-membrane-derived systems are used as carriers and release their cargo in a controlled manner. Recently, great attention has been directed to cells as carrier systems for treating several diseases. There are various challenges in the development of cell-based drug delivery systems. The prediction of the properties of these platforms is a prerequisite step in their development to reduce undesirable effects. Integrating nanotechnology and artificial intelligence leads to more innovative technologies. Artificial intelligence quickly mines data and makes decisions more quickly and accurately. Machine learning as a subset of the broader artificial intelligence has been used in nanomedicine to design safer nanomaterials. Here, how challenges of developing cell-based drug delivery systems can be solved with potential predictive models of artificial intelligence and machine learning is portrayed. The most famous cell-based drug delivery systems and their challenges are described. Last but not least, artificial intelligence and most of its types used in nanomedicine are highlighted. The present Review has shown the challenges of developing cells or their derivatives as carriers and how they can be used with potential predictive models of artificial intelligence and machine learning.
Collapse
Affiliation(s)
- Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Niloofar Mozafari
- Design and System Operations Department, Regional Information Center for Science and Technology, 71946 94171 Shiraz, Iran
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Centre, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Centre, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| |
Collapse
|
24
|
Wu D, Chen Q, Chen X, Han F, Chen Z, Wang Y. The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduct Target Ther 2023; 8:217. [PMID: 37231000 PMCID: PMC10212980 DOI: 10.1038/s41392-023-01481-w] [Citation(s) in RCA: 201] [Impact Index Per Article: 201.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Blood-brain barrier (BBB) is a natural protective membrane that prevents central nervous system (CNS) from toxins and pathogens in blood. However, the presence of BBB complicates the pharmacotherapy for CNS disorders as the most chemical drugs and biopharmaceuticals have been impeded to enter the brain. Insufficient drug delivery into the brain leads to low therapeutic efficacy as well as aggravated side effects due to the accumulation in other organs and tissues. Recent breakthrough in materials science and nanotechnology provides a library of advanced materials with customized structure and property serving as a powerful toolkit for targeted drug delivery. In-depth research in the field of anatomical and pathological study on brain and BBB further facilitates the development of brain-targeted strategies for enhanced BBB crossing. In this review, the physiological structure and different cells contributing to this barrier are summarized. Various emerging strategies for permeability regulation and BBB crossing including passive transcytosis, intranasal administration, ligands conjugation, membrane coating, stimuli-triggered BBB disruption, and other strategies to overcome BBB obstacle are highlighted. Versatile drug delivery systems ranging from organic, inorganic, and biologics-derived materials with their synthesis procedures and unique physio-chemical properties are summarized and analyzed. This review aims to provide an up-to-date and comprehensive guideline for researchers in diverse fields, offering perspectives on further development of brain-targeted drug delivery system.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| | - Qi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| |
Collapse
|
25
|
Ou A, Wang Y, Zhang J, Huang Y. Living Cells and Cell-Derived Vesicles: A Trojan Horse Technique for Brain Delivery. Pharmaceutics 2023; 15:pharmaceutics15041257. [PMID: 37111742 PMCID: PMC10145830 DOI: 10.3390/pharmaceutics15041257] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Brain diseases remain a significant global healthcare burden. Conventional pharmacological therapy for brain diseases encounters huge challenges because of the blood-brain barrier (BBB) limiting the delivery of therapeutics into the brain parenchyma. To address this issue, researchers have explored various types of drug delivery systems. Cells and cell derivatives have attracted increasing interest as "Trojan horse" delivery systems for brain diseases, owing to their superior biocompatibility, low immunogenicity, and BBB penetration properties. This review provided an overview of recent advancements in cell- and cell-derivative-based delivery systems for the diagnosis and treatment of brain diseases. Additionally, it discussed the challenges and potential solutions for clinical translation.
Collapse
Affiliation(s)
- Ante Ou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuewei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
26
|
Chen Y, Qin D, Zou J, Li X, Guo XD, Tang Y, Liu C, Chen W, Kong N, Zhang CY, Tao W. Living Leukocyte-Based Drug Delivery Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207787. [PMID: 36317596 DOI: 10.1002/adma.202207787] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/10/2022] [Indexed: 05/17/2023]
Abstract
Leukocytes play a vital role in immune responses, including defending against invasive pathogens, reconstructing impaired tissue, and maintaining immune homeostasis. When the immune system is activated in vivo, leukocytes accomplish a series of orderly and complex regulatory processes. While cancer and inflammation-related diseases like sepsis are critical medical difficulties plaguing humankind around the world, leukocytes have been shown to largely gather at the focal site, and significantly contribute to inflammation and cancer progression. Therefore, the living leukocyte-based drug delivery systems have attracted considerable attention in recent years due to the innate and specific targeting effect, low immunogenicity, improved therapeutic efficacy, and low reverse effect. In this review, the recent advances in the development of living leukocyte-based drug delivery systems including macrophages, neutrophils, and lymphocytes as promising treatment strategies for cancer and inflammation-related diseases are introduced. The advantages, current challenges, and limitations of these delivery systems are also discussed, as well as perspectives on the future development of precision and targeted therapy in the clinics are provided. Collectively, it is expected that such kind of living cell-based drug delivery system is promising to improve or even revolutionize the treatments of cancers and inflammation-related diseases in the clinics.
Collapse
Affiliation(s)
- Yaxin Chen
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Duotian Qin
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zou
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau (SAR), 519020, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xiaobin Li
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Xin Dong Guo
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yi Tang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Chuang Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Na Kong
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, 311121, China
| | - Can Yang Zhang
- Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 440300, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
27
|
Wang W, Hassan MM, Mao G. Colloidal Perspective on Targeted Drug Delivery to the Central Nervous System. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:3235-3245. [PMID: 36825490 DOI: 10.1021/acs.langmuir.2c02949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
This article describes a new approach in targeted drug delivery to the central nervous system (CNS) in a significant departure from the predominant systematic drug administration attempting to penetrate the blood-brain barrier (BBB). Nanoparticles chemically conjugated to neural tract tracer proteins are capable of path-specific axonal retrograde transport, transneuronal transport, and anatomical tract flow to bypass the BBB. To celebrate the work by Dr. Bettye Washington Greene on the physical chemistry of colloidal particles, this article focuses on the physiochemical characteristics of the nanoparticles, various colloidal forces that impact the colloidal stability of nanoparticles in biological media, and surface chemistry strategies to avoid nanoparticle aggregation-induced poor therapeutic outcomes. The biological environment for the anatomical retrograde transport of neural tract tracers is examined to directly link factors impacting the colloidal stability of the new class of CNS-targeting nanoconjugates such as nanoconjugate size, shape, surface charge, surface chemistry, ionic strength, pH, and protein adsorption on the nanoparticle. We conclude with opportunities and challenges for future research.
Collapse
Affiliation(s)
- Wenqian Wang
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Md Musfizur Hassan
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Guangzhao Mao
- School of Chemical Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| |
Collapse
|
28
|
Xiong T, Yang K, Zhao T, Zhao H, Gao X, You Z, Fan C, Kang X, Yang W, Zhuang Y, Chen Y, Dai J. Multifunctional Integrated Nanozymes Facilitate Spinal Cord Regeneration by Remodeling the Extrinsic Neural Environment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205997. [PMID: 36646515 PMCID: PMC9982579 DOI: 10.1002/advs.202205997] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/16/2022] [Indexed: 06/17/2023]
Abstract
High levels of reactive oxygen species (ROS) and inflammation create a complicated extrinsic neural environment that dominates the initial post-injury period after spinal cord injury (SCI). The compensatory pathways between ROS and inflammation limited the efficacy of modulating the above single treatment regimen after SCI. Here, novel "nanoflower" Mn3 O4 integrated with "pollen" IRF-5 SiRNA was designed as a combination antioxidant and anti-inflammatory treatment after SCI. The "nanoflower" and "pollen" structure was encapsulated with a neutrophil membrane for protective and targeted delivery. Furthermore, valence-engineered nanozyme Mn3 O4 imitated the cascade response of antioxidant enzymes with a higher substrate affinity compared to natural antioxidant enzymes. Nanozymes effectively catalyzed ROS to generate O2 , which is advantageous for reducing oxidative stress and promoting angiogenesis. The screened "pollen" IRF-5 SiRNA could reverse the inflammatory phenotype by reducing interferon regulatory factors-5 (IRF-5) expression (protein level: 73.08% and mRNA level: 63.10%). The decreased expression of pro-inflammatory factors reduced the infiltration of inflammatory cells, resulting in less neural scarring. In SCI rats, multifunctional nanozymes enhanced the proliferation of various neuronal subtypes (motor neurons, interneurons, and sensory neurons) and the recovery of locomotor function, demonstrating that the remodeling of the extrinsic neural environment is a promising strategy to facilitate nerve regeneration.
Collapse
Affiliation(s)
- Tiandi Xiong
- School of Nano Technology and Nano BionicsUniversity of Science and Technology of ChinaHefei230026China
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Keni Yang
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Tongtong Zhao
- School of Nano Technology and Nano BionicsUniversity of Science and Technology of ChinaHefei230026China
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Haitao Zhao
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Xu Gao
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Zhifeng You
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Caixia Fan
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Xinyi Kang
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Wen Yang
- School of Nano Technology and Nano BionicsUniversity of Science and Technology of ChinaHefei230026China
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Yan Zhuang
- School of Nano Technology and Nano BionicsUniversity of Science and Technology of ChinaHefei230026China
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Yanyan Chen
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Jianwu Dai
- Key Laboratory for Nano‐Bio Interface ResearchDivision of NanobiomedicineSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- State Key Laboratory of Molecular Development BiologyInstitute of Genetics and Developmental BiologyChinese Academy of SciencesBeijing100101China
| |
Collapse
|
29
|
Yousefpour P, Ni K, Irvine DJ. Targeted modulation of immune cells and tissues using engineered biomaterials. NATURE REVIEWS BIOENGINEERING 2023; 1:107-124. [PMID: 37772035 PMCID: PMC10538251 DOI: 10.1038/s44222-022-00016-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 09/30/2023]
Abstract
Therapies modulating the immune system offer the prospect of treating a wide range of conditions including infectious diseases, cancer and autoimmunity. Biomaterials can promote specific targeting of immune cell subsets in peripheral or lymphoid tissues and modulate the dosage, timing and location of stimulation, thereby improving safety and efficacy of vaccines and immunotherapies. Here we review recent advances in biomaterials-based strategies, focusing on targeting of lymphoid tissues, circulating leukocytes, tissue-resident immune cells and immune cells at disease sites. These approaches can improve the potency and efficacy of immunotherapies by promoting immunity or tolerance against different diseases.
Collapse
Affiliation(s)
- Parisa Yousefpour
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaiyuan Ni
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
30
|
Lu J, Gao X, Wang S, He Y, Ma X, Zhang T, Liu X. Advanced strategies to evade the mononuclear phagocyte system clearance of nanomaterials. EXPLORATION (BEIJING, CHINA) 2023; 3:20220045. [PMID: 37323617 PMCID: PMC10191055 DOI: 10.1002/exp.20220045] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/12/2022] [Indexed: 06/17/2023]
Abstract
Nanomaterials are promising carriers to improve the bioavailability and therapeutic efficiency of drugs by providing preferential drug accumulation at their sites of action, but their delivery efficacy is severely limited by a series of biological barriers, especially the mononuclear phagocytic system (MPS)-the first and major barrier encountered by systemically administered nanomaterials. Herein, the current strategies for evading the MPS clearance of nanomaterials are summarized. First, engineering nanomaterials methods including surface modification, cell hitchhiking, and physiological environment modulation to reduce the MPS clearance are explored. Second, MPS disabling methods including MPS blockade, suppression of macrophage phagocytosis, and macrophages depletion are examined. Last, challenges and opportunities in this field are further discussed.
Collapse
Affiliation(s)
- Junjie Lu
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'anChina
| | - Xiao Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China of the Ministry of EducationSchool of MedicineNorthwest UniversityXi'anChina
| | - Siyao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China of the Ministry of EducationSchool of MedicineNorthwest UniversityXi'anChina
| | - Yuan He
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'anChina
| | - Xiaowei Ma
- National Center for Veterinary Drug Safety EvaluationCollege of Veterinary MedicineChina Agricultural UniversityBeijingChina
| | - Tingbin Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of EducationCollege of Chemistry and Materials ScienceNorthwest UniversityXi'anChina
| | - Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China of the Ministry of EducationSchool of MedicineNorthwest UniversityXi'anChina
- Institute of Regenerative and Reconstructive MedicineMed‐X InstituteNational Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
31
|
Pawar B, Vasdev N, Gupta T, Mhatre M, More A, Anup N, Tekade RK. Current Update on Transcellular Brain Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14122719. [PMID: 36559214 PMCID: PMC9786068 DOI: 10.3390/pharmaceutics14122719] [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: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
It is well known that the presence of a blood-brain barrier (BBB) makes drug delivery to the brain more challenging. There are various mechanistic routes through which therapeutic molecules travel and deliver the drug across the BBB. Among all the routes, the transcellular route is widely explored to deliver therapeutics. Advances in nanotechnology have encouraged scientists to develop novel formulations for brain drug delivery. In this article, we have broadly discussed the BBB as a limitation for brain drug delivery and ways to solve it using novel techniques such as nanomedicine, nose-to-brain drug delivery, and peptide as a drug delivery carrier. In addition, the article will help to understand the different factors governing the permeability of the BBB, as well as various formulation-related factors and the body clearance of the drug delivered into the brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rakesh Kumar Tekade
- Correspondence: ; Tel.: +91-796674550 or +91-7966745555; Fax: +91-7966745560
| |
Collapse
|
32
|
Hu R, Yang Y, Song G, Zhao F, Chen S, Zhou Z, Zheng J, Shen W. In vivo targeting capacities of different nanoparticles to prostate tissues based on a mouse model of chronic bacterial prostatitis. Front Bioeng Biotechnol 2022; 10:1021385. [PMID: 36277385 PMCID: PMC9582453 DOI: 10.3389/fbioe.2022.1021385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic bacterial prostatitis usually occurs in men and seriously affects the quality of life of patients. The efficacy of chronic bacterial prostatitis treatment is limited by the difficulty for free drugs (e.g., antibiotics) to penetrate the prostate epithelium and target inflammatory tissues. The advent of nanotechnology offers the possibility to address this issue, such as the development of targeted nanoparticle delivery strategies that may overcome these important limitations. The physicochemical properties of nanoparticles, such as particle size, shape and surface modification ligands, determine their targeting effectiveness. In this study, nanoparticles with different physicochemical properties were prepared to explore and confirm their targeting capacities to inflammatory prostate tissues of chronic bacterial prostatitis, focusing on the effects of size and different modification ligands on the targeting performance. In vivo and ex vivo imaging results verified that folic acid-modified nanoparticles with a particle size of 180–190 nm via tail intravenous injection had the optimal targeting efficiency to prostate tissues. Our results provide an experimental basis and reference value for targeted therapy of prostate-related diseases with nanotechnology in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Zheng
- *Correspondence: Jun Zheng, ; Wenhao Shen,
| | | |
Collapse
|
33
|
Mitusova K, Peltek OO, Karpov TE, Muslimov AR, Zyuzin MV, Timin AS. Overcoming the blood-brain barrier for the therapy of malignant brain tumor: current status and prospects of drug delivery approaches. J Nanobiotechnology 2022; 20:412. [PMID: 36109754 PMCID: PMC9479308 DOI: 10.1186/s12951-022-01610-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/18/2022] [Indexed: 01/06/2023] Open
Abstract
Besides the broad development of nanotechnological approaches for cancer diagnosis and therapy, currently, there is no significant progress in the treatment of different types of brain tumors. Therapeutic molecules crossing the blood-brain barrier (BBB) and reaching an appropriate targeting ability remain the key challenges. Many invasive and non-invasive methods, and various types of nanocarriers and their hybrids have been widely explored for brain tumor treatment. However, unfortunately, no crucial clinical translations were observed to date. In particular, chemotherapy and surgery remain the main methods for the therapy of brain tumors. Exploring the mechanisms of the BBB penetration in detail and investigating advanced drug delivery platforms are the key factors that could bring us closer to understanding the development of effective therapy against brain tumors. In this review, we discuss the most relevant aspects of the BBB penetration mechanisms, observing both invasive and non-invasive methods of drug delivery. We also review the recent progress in the development of functional drug delivery platforms, from viruses to cell-based vehicles, for brain tumor therapy. The destructive potential of chemotherapeutic drugs delivered to the brain tumor is also considered. This review then summarizes the existing challenges and future prospects in the use of drug delivery platforms for the treatment of brain tumors.
Collapse
Affiliation(s)
- Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Oleksii O Peltek
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Albert R Muslimov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
- Sirius University of Science and Technology, Olympic Ave 1, Sirius, 354340, Russian Federation
| | - Mikhail V Zyuzin
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation.
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation.
| |
Collapse
|
34
|
Zhang M, Wang Z, Shao Y, Zhao Y, Liu Z. Complement-Opsonized NIR-IIb Emissive Immunotracers for Dynamically Monitoring Neutrophils in Inflammation-Related Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2203477. [PMID: 35793262 DOI: 10.1002/adma.202203477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Real-time monitoring of neutrophil dynamics is crucial for timely diagnosis and effective treatment of inflammation-related diseases, which requires a reliable tracer for in vivo tracking of neutrophils. However, immunotracers for neutrophils are extremely limited because of the difficulty in labeling the cells. Inspired by the natural biological function of the complement system, a strategy of enhancing the complement C3 opsonization of lanthanide-doped nanoparticles (LnNPs) by modulating their surface chemistry, thus developing a near infrared-IIb emissive nanotracer for neutrophils, is reported herein. Four kinds of surface-modified LnNPs are fabricated, among which phospholipids DOPG-modified LnNPs (LnNPs@PG) with weak antifouling ability and hydroxyl groups adsorb more complement C3 proteins and form covalent linkages with C3b active fragments under inflammation conditions, inducing enhanced complement C3 opsonization. Therefore, LnNPs@PG with enhanced complement C3 opsonization are capable of efficiently labeling inflammation-stimulated neutrophils in vivo through complement-receptors-mediated phagocytosis and achieve dynamic monitoring neutrophils during cutaneous wound healing and cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Meng Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Zijun Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Yunlong Shao
- Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry and Chemistry Engineering, Beijing University of Technology, Beijing, 100124, China
| | - Yaoyao Zhao
- Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry and Chemistry Engineering, Beijing University of Technology, Beijing, 100124, China
| | - Zhihong Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
35
|
Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187:114394. [PMID: 35718252 DOI: 10.1016/j.addr.2022.114394] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
Abstract
Cell-based drug delivery systems (DDSs) have received attention recently because of their unique biological properties and self-powered functions, such as excellent biocompatibility, low immunogenicity, long circulation time, tissue-homingcharacteristics, and ability to cross biological barriers. A variety of cells, including erythrocytes, stem cells, and lymphocytes, have been explored as functional vectors for the loading and delivery of various therapeutic payloads (e.g., small-molecule and nucleic acid drugs) for subsequent disease treatment. These cell-based DDSs have their own unique in vivo fates, which are attributed to various factors, including their biological properties and functions, the loaded drugs and loading process, physiological and pathological circumstances, and the body's response to these carrier cells, which result in differences in drug delivery efficiency and therapeutic effect. In this review, we summarize the main cell-based DDSs and their biological properties and functions, applications in drug delivery and disease treatment, and in vivo fate and influencing factors. We envision that the unique biological properties, combined with continuing research, will enable development of cell-based DDSs as friendly drug vectors for the safe, effective, and even personalized treatment of diseases.
Collapse
|
36
|
Habibi N, Brown TD, Adu-Berchie K, Christau S, Raymond JE, Mooney DJ, Mitragotri S, Lahann J. Nanoparticle Properties Influence Transendothelial Migration of Monocytes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:5603-5616. [PMID: 35446569 DOI: 10.1021/acs.langmuir.2c00200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanoparticle-based delivery of therapeutics to the brain has had limited clinical impact due to challenges crossing the blood-brain barrier (BBB). Certain cells, such as monocytes, possess the ability to migrate across the BBB, making them attractive candidates for cell-based brain delivery strategies. In this work, we explore nanoparticle design parameters that impact both monocyte association and monocyte-mediated BBB transport. We use electrohydrodynamic jetting to prepare nanoparticles of varying sizes, compositions, and elasticity to address their impact on uptake by THP-1 monocytes and permeation across the BBB. An in vitro human BBB model is developed using human cerebral microvascular endothelial cells (hCMEC/D3) for the assessment of migration. We compare monocyte uptake of both polymeric and synthetic protein nanoparticles (SPNPs) of various sizes, as well as their effect on cell migration. SPNPs (human serum albumin/HSA or human transferrin/TF) are shown to promote increased monocyte-mediated transport across the BBB over polymeric nanoparticles. TF SPNPs (200 nm) associate readily, with an average uptake of 138 particles/cell. Nanoparticle loading is shown to influence the migration of THP-1 monocytes. The migration of monocytes loaded with 200 nm TF and 200 nm HSA SPNPs was 2.3-fold and 2.1-fold higher than that of an untreated control. RNA-seq analysis after TF SPNP treatment suggests that the upregulation of several migration genes may be implicated in increased monocyte migration (ex. integrin subunits α M and α L). Integrin β 2 chain combines with either integrin subunit α M chain or integrin subunit α L chain to form macrophage antigen 1 and lymphocyte function-associated antigen 1 integrins. Both products play a pivotal role in the transendothelial migration cascade. Our findings highlight the potential of SPNPs as drug and/or gene delivery platforms for monocyte-mediated BBB transport, especially where conventional polymer nanoparticles are ineffective or otherwise not desirable.
Collapse
Affiliation(s)
- Nahal Habibi
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tyler D Brown
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Kwasi Adu-Berchie
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Stephanie Christau
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeffery E Raymond
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - David J Mooney
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Samir Mitragotri
- Wyss Institute of Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, Massachusetts 02115, United States
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, Massachusetts 02318, United States
| | - Joerg Lahann
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Material Science & Engineering, Department of Macromolecular Science & Engineering, and Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
37
|
Chen J, Jin J, Li K, Shi L, Wen X, Fang F. Progresses and Prospects of Neuroprotective Agents-Loaded Nanoparticles and Biomimetic Material in Ischemic Stroke. Front Cell Neurosci 2022; 16:868323. [PMID: 35480961 PMCID: PMC9035592 DOI: 10.3389/fncel.2022.868323] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/16/2022] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke remains the leading cause of death and disability, while the main mechanisms of dominant neurological damage in stroke contain excitotoxicity, oxidative stress, and inflammation. The clinical application of many neuroprotective agents is limited mainly due to their inability to cross the blood-brain barrier (BBB), short half-life and low bioavailability. These disadvantages can be better eliminated/reduced by nanoparticle as the carrier of these drugs. This review expounded the currently hot researched nanomedicines from the perspective of the mechanism of ischemic stroke. In addition, this review describes the bionic nanomedicine delivery strategies containing cells, cell membrane vesicles and exosomes that can effectively avoid the risk of clearance by the reticuloendothelial system. The potential challenges and application prospect for clinical translation of these delivery platforms were also discussed.
Collapse
Affiliation(s)
- Junfa Chen
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jing Jin
- Laboratory Medicine Center, Zhejiang Center for Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Kaiqiang Li
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lin Shi
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Xuehua Wen
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Xuehua Wen,
| | - Fuquan Fang
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Fuquan Fang,
| |
Collapse
|
38
|
Wu Y, Liu Y, Wang T, Jiang Q, Xu F, Liu Z. Living Cell for Drug Delivery. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
39
|
Advanced drug delivery system against ischemic stroke. J Control Release 2022; 344:173-201. [DOI: 10.1016/j.jconrel.2022.02.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 02/06/2023]
|
40
|
miR-155-5p in Extracellular Vesicles Derived from Choroid Plexus Epithelial Cells Promotes Autophagy and Inflammation to Aggravate Ischemic Brain Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8603427. [PMID: 35222806 PMCID: PMC8865969 DOI: 10.1155/2022/8603427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/08/2022] [Indexed: 12/12/2022]
Abstract
Ischemic stroke is a common disease of the central nervous system, and ischemic brain injury (IBI) is its main manifestation. Recently, extracellular vesicles (EVs) have been strongly related to the diagnosis and treatment of IBI. However, the underlying mechanism of their effects remains enigmatic. In the present study, we aimed to study how miR-155-5p plays a role in choroid plexus epithelial (CPE) cell-derived EVs in IBI pathology. We found that miR-155-5p expression was enriched in CPE cell-derived EVs, which were subsequently internalized by neurons, enabling the delivery of miR-155-5p into neurons. An inducible oxygen and glucose deprivation and reoxygenation (OGD/R) cell model was developed to mimic ischemic neuronal injury in vitro. miR-155-5p overexpression led to reduced neuron viability, promoted apoptosis, elevated autophagic proteins' expression, and activated NLR family pyrin domain-containing 3- (NLRP3-) related inflammasomes, thereby aggravating OGD-induced neuronal injury. A dual-luciferase reporter assay exhibited that miR-155-5p could inhibit the Ras homolog enriched in brain (Rheb) expression, a mechanism critical for miR-155-5p-mediated neuronal injury. Furthermore, a mouse IBI model was developed using the transient middle cerebral artery occlusion (tMCAO) method. Animal experiments verified that miR-155p delivery via CPE cell-derived EVs aggravated IBI by suppressing Rheb expression. In conclusion, miR-155-5p in CPE-derived EVs can aggravate IBI pathology by suppressing Rheb expression and promoting NLRP3-mediated inflammasomes, suggesting its role as a potential therapeutic target in IBI.
Collapse
|
41
|
Fukuta T, Oku N, Kogure K. Application and Utility of Liposomal Neuroprotective Agents and Biomimetic Nanoparticles for the Treatment of Ischemic Stroke. Pharmaceutics 2022; 14:pharmaceutics14020361. [PMID: 35214092 PMCID: PMC8877231 DOI: 10.3390/pharmaceutics14020361] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke is still one of the leading causes of high mortality and severe disability worldwide. Therapeutic options for ischemic stroke and subsequent cerebral ischemia/reperfusion injury remain limited due to challenges associated with drug permeability through the blood-brain barrier (BBB). Neuroprotectant delivery with nanoparticles, including liposomes, offers a promising solution to address this problem, as BBB disruption following ischemic stroke allows nanoparticles to pass through the intercellular gaps between endothelial cells. To ameliorate ischemic brain damage, a number of nanotherapeutics encapsulating neuroprotective agents, as well as surface-modified nanoparticles with specific ligands targeting the injured brain regions, have been developed. Combination therapy with nanoparticles encapsulating neuroprotectants and tissue plasminogen activator (t-PA), a globally approved thrombolytic agent, has been demonstrated to extend the narrow therapeutic time window of t-PA. In addition, the design of biomimetic drug delivery systems (DDS) employing circulating cells (e.g., leukocytes, platelets) with unique properties has recently been investigated to overcome the injured BBB, utilizing these cells’ inherent capability to penetrate the ischemic brain. Herein, we review recent findings on the application and utility of nanoparticle DDS, particularly liposomes, and various approaches to developing biomimetic DDS functionalized with cellular membranes/membrane proteins for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama 640-8156, Japan
| | - Naoto Oku
- Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima 770-8505, Japan
| |
Collapse
|
42
|
Tang L, He S, Yin Y, Li J, Xiao Q, Wang R, Gao L, Wang W. Combining nanotechnology with the multifunctional roles of neutrophils against cancer and inflammatory disease. NANOSCALE 2022; 14:1621-1645. [PMID: 35079756 DOI: 10.1039/d1nr07725b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neutrophils, the most abundant leukocytes in humans, play a crucial role in acute inflammation during infection and tumorigenesis. Neutrophils are the major types of cells recruited to the inflammation sites induced by pathogens, exhibiting great homing ability towards inflammatory disorders and tumor sites. Therefore, a neutrophil-based drug delivery system (NDDS) has become a promising platform for anti-cancer and anti-inflammatory treatment. Recent decades have witnessed the huge progress of applying nanomaterials in drug delivery. Nanomaterials are regarded as innovative components to enrich the field of neutrophil-based therapies due to their unique physiochemical characteristics. In this review, the latest advancement of combining diverse nanomaterials with an NDDS for cancer and inflammatory disease treatment will be summarized. It is discussed how nanomaterials empower the therapeutic area of an NDDS and how an NDDS circumvents the limitations of nanomaterials. Moreover, based on the finding that neutrophils are closely involved in the progression of cancer and inflammatory diseases, emerging therapeutic strategies that target neutrophils will be outlined. Finally, as neutrophils were demonstrated to play a central role in the immunopathology of COVID-19, which causes necroinflammation that is responsible for the cytokine storm and sepsis during coronavirus infections, novel therapeutic approaches that anchor neutrophils against the pathological consequences related to COVID-19 will be highlighted as well.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Jing Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Qiaqia Xiao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Ruotong Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Lijun Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| |
Collapse
|
43
|
cRGD enables rapid phagocytosis of liposomal vancomycin for intracellular bacterial clearance. J Control Release 2022; 344:202-213. [DOI: 10.1016/j.jconrel.2022.02.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/24/2022]
|
44
|
|
45
|
Li YJ, Wu JY, Liu J, Qiu X, Xu W, Tang T, Xiang DX. From blood to brain: blood cell-based biomimetic drug delivery systems. Drug Deliv 2021; 28:1214-1225. [PMID: 34142628 PMCID: PMC8259840 DOI: 10.1080/10717544.2021.1937384] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
Brain drug delivery remains a major difficulty for several challenges including the blood-brain barrier, lesion spot targeting, and stability during circulation. Blood cells including erythrocytes, platelets, and various subpopulations of leukocytes have distinct features such as long-circulation, natural targeting, and chemotaxis. The development of biomimetic drug delivery systems based on blood cells for brain drug delivery is growing fast by using living cells, membrane coating nanotechnology, or cell membrane-derived nanovesicles. Blood cell-based vehicles are superior delivery systems for their engineering feasibility and versatile delivery ability of chemicals, proteins, and all kinds of nanoparticles. Here, we focus on advances of blood cell-based biomimetic carriers for from blood to brain drug delivery and discuss their translational challenges in the future.
Collapse
Affiliation(s)
- Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jun-Yong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jihua Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiaohan Qiu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenjie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Da-Xiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
46
|
Cell-based therapeutics for the treatment of hematologic diseases inside the bone marrow. J Control Release 2021; 339:1-13. [PMID: 34536449 DOI: 10.1016/j.jconrel.2021.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022]
Abstract
Cell-based therapies could overcome the limitations of traditional drugs for the treatment of refractory diseases. Cell exchange between the bone marrow and blood is bidirectional. Several kinds of cells in the blood have the capability to enter the bone marrow by interacting with sinusoidal cells under specific physiological or pathological conditions. These cells are the potential living therapeutics or delivery vehicles to treat or prevent bone marrow-related hematologic diseases. In this review, we summarized the in vivo molecular mechanisms and kinetics of these cells in entering the bone marrow. The advances in the fabrication of living cell drugs and the strategies to design cell-based carriers into the bone marrow were discussed. The latest studies on how to use blood cells as living drugs or as drug carriers to improve therapeutic outcomes of hematologic diseases inside the bone marrow were highlighted.
Collapse
|
47
|
Sialic acid conjugate-modified liposomes enable tumor homing of epirubicin via neutrophil/monocyte infiltration for tumor therapy. Acta Biomater 2021; 134:702-715. [PMID: 34339869 DOI: 10.1016/j.actbio.2021.07.063] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/26/2022]
Abstract
Neutrophils and monocytes (N/Ms) are potential candidates for the delivery of therapeutic agents to the tumor microenvironment (TME) because of their tumor-accumulating nature. L-selectin and Siglec-1, receptors for sialic acid (SA), are highly expressed in circulating neutrophils and monocytes, respectively, in tumor-bearing mice, and N/Ms are recruited to tumors in response to inflammatory cytokines secreted by the TME, promoting tumor growth and invasion. Therefore, we constructed a drug delivery nano-platform using N/Ms as vehicles. SA-stearic acid conjugate was synthesized and utilized to modify epirubicin-loaded liposomes (EPI-SL) for enhanced endocytosis of liposomes by circulating N/Ms. Cellular uptake studies showed that SA modification improved the accumulation of EPI in N/Ms and did not alter the inherent chemotaxis of N/Ms. In tumor-bearing mice, EPI-SL significantly improved the tumor-targeting efficiency and therapeutic efficacy of EPI compared to other preparations and even eradicated tumors because of the tumor-accumulating and inhibitory effects of N/Ms containing EPI-SL. Our research showed, for the first time, that as an N/M-based drug delivery platform, EPI-SL remedied the limited tumor targeting in the conventional EPR effect-based treatment strategy, contributing to the exploitation of a new drug delivery platform for cancer treatment. STATEMENT OF SIGNIFICANCE: Tumor-associated neutrophils (TANs) and macrophages (TAMs) are closely associated with tumor growth and invasion, and therefore the development of therapeutic strategies targeting TANs and TAMs is crucial for tumor treatment. Given that most TANs and TAMs are derived from peripheral blood neutrophils and monocytes (N/Ms), respectively, we synthesized sialic acid-stearic acid conjugates that specifically bind N/Ms for the surface modification of liposomal epirubicin (EPI-SL). The N/Ms loaded with EPI-SL maintained their inherent chemotaxis toward the tumor. Additionally, EPI-SL significantly improved the survival of tumor-bearing mice and even eradicated tumors. These findings suggested that EPI-SL has substantial potential for clinical application by compensating for the previous low efficacy of ex vivo transformed cell infusion and improving the tumor-targeting efficiency.
Collapse
|
48
|
Wang F, Hou W, Xiao C, Hao Y, Su N, Deng Y, Wang J, Yu L, Xie JM, Xiong JW, Luo Y. Endothelial cell membrane-based biosurface for targeted delivery to acute injury: analysis of leukocyte-mediated nanoparticle transportation. NANOSCALE 2021; 13:14636-14643. [PMID: 34558568 DOI: 10.1039/d1nr04181a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Mimicking and leveraging biological structures and materials provide important approaches to develop functional vehicles for drug delivery. Taking advantage of the affinity and adhesion between the activated endothelial cells and innate immune cells during inflammatory responses, hybrid polyester nanoparticles coated with endothelial cell membranes (EM-P) containing adhesion molecules were fabricated and their capability as vehicles to travel to the acute injury sites through leukocyte-mediated processes was investigated. The in vivo studies and quantitative analyses performed through the lung-inflammation mouse models demonstrated that the EM-Ps preferentially interacted with the neutrophils and monocytes in the circulation and the cellular membrane-based biosurface improved the nanoparticle transportation to the inflamed lung possibly via the motility of neutrophils. Utilizing the transgenic zebrafish model, the leukocyte-mediated transportation and biodistribution of EM-Ps were further visualized in real time at the whole-organism level. Endothelial membranes provided a new biosurface for developing biomimetic vehicles to allow the immune cell-mediated transportation and may enable advanced systems for active and highly efficient drug delivery.
Collapse
Affiliation(s)
- Fang Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Wenda Hou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Chenglu Xiao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Yaoyao Hao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Ni Su
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Yu Deng
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jieting Wang
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Luying Yu
- Nanomedical Technology Research Institute, Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, China 350122
| | - Jing-Ming Xie
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
| | - Jing-Wei Xiong
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China 100871
| | - Ying Luo
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China 100871.
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America 02155
| |
Collapse
|
49
|
He W, Zhang Z, Sha X. Nanoparticles-mediated emerging approaches for effective treatment of ischemic stroke. Biomaterials 2021; 277:121111. [PMID: 34488117 DOI: 10.1016/j.biomaterials.2021.121111] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022]
Abstract
Ischemic stroke leads to high disability and mortality. The limited delivery efficiency of most therapeutic substances is a major challenge for effective treatment of ischemic stroke. Inspired by the prominent merit of nanoscale particles in brain targeting and blood-brain barrier (BBB) penetration, various functional nanoparticles have been designed as promising drug delivery platforms that are expected to improve the therapeutic effect of ischemic stroke. Based on the complex pathological mechanisms of ischemic stroke, this review outline and summarize the rationally designed nanoparticles-mediated emerging approaches for effective treatment of ischemic stroke, including recanalization therapy, neuroprotection therapy, and combination therapy. On this bases, the potentials and challenges of nanoparticles in the treatment of ischemic stroke are revealed, and new thoughts and perspectives are proposed for the design of feasible nanoparticles for effective treatment of ischemic stroke.
Collapse
Affiliation(s)
- Wenxiu He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xianyi Sha
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, China; The Institutes of Integrative Medicine of Fudan University, 120 Urumqi Middle Road, Shanghai, 200040, China.
| |
Collapse
|
50
|
Du Y, Wang S, Zhang M, Chen B, Shen Y. Cells-Based Drug Delivery for Cancer Applications. NANOSCALE RESEARCH LETTERS 2021; 16:139. [PMID: 34478000 PMCID: PMC8417195 DOI: 10.1186/s11671-021-03588-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/03/2021] [Indexed: 05/04/2023]
Abstract
The application of cells as carriers to encapsulate chemotherapy drugs is of great significance in antitumor therapy. The advantages of reducing systemic toxicity, enhancing targeting and enhancing the penetrability of drugs to tumor cells make it have great potential for clinical application in the future. Many studies and advances have been made in the encapsulation of drugs by using erythrocytes, white blood cells, platelets, immune cells and even tumor cells. The results showed that the antitumor effect of cell encapsulation chemotherapy drugs was better than that of single chemotherapy drugs. In recent years, the application of cell-based vectors in cancer has become diversified. Both chemotherapeutic drugs and photosensitizers can be encapsulated, so as to achieve multiple antitumor effects of chemotherapy, photothermal therapy and photodynamic therapy. A variety of ways of coordinated treatment can produce ideal results even in the face of multidrug-resistant and metastatic tumors. However, it is regrettable that this technology is only used in vitro for the time being. Standard answers have not yet been obtained for the preservation of drug-loaded cells and the safe way of infusion into human body. Therefore, the successful application of drug delivery technology in clinical still faces many challenges in the future. In this paper, we discuss the latest development of different cell-derived drug delivery systems and the challenges it will face in the future.
Collapse
Affiliation(s)
- Ying Du
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, School of Medicine, Southeast University, Ding JiaQiao Street 87, Nanjing, 210009, People's Republic of China
| | - Shujun Wang
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, School of Medicine, Southeast University, Ding JiaQiao Street 87, Nanjing, 210009, People's Republic of China
| | - Meilin Zhang
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, School of Medicine, Southeast University, Ding JiaQiao Street 87, Nanjing, 210009, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, School of Medicine, Southeast University, Ding JiaQiao Street 87, Nanjing, 210009, People's Republic of China.
| | - Yanfei Shen
- Department of Chemistry and Chemical Engineering, Southeast University School of Medicine, Ding JiaQiao Street 87, Nanjing, 210009, People's Republic of China.
| |
Collapse
|