1
|
Cao H, Jiang J, Chen L, Gao L. Mimicomes: Mimicking Multienzyme System by Artificial Design. Adv Healthc Mater 2025; 14:e2402372. [PMID: 39380346 DOI: 10.1002/adhm.202402372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Enzymes are widely distributed in organelles of cells, which are capable of carrying out specific catalytic reactions. In general, several enzymes collaborate to facilitate complex reactions and engage in vital biochemical processes within cells, which are also called cascade systems. The cascade systems are highly efficient, and their dysfunction is associated with a multitude of endogenous diseases. The advent of nanotechnology makes it possible to mimic these cascade systems in nature and realize partial functions of natural biological processes both in vitro and in vivo. To emphasize the significance of artificial cascade systems, mimicomes is first proposed, a new concept that refers to the artificial cascade catalytic systems. Typically, mimicomes are able to mimic specific natural biochemical catalytic processes or facilitate the overall catalytic efficiency of cascade systems. Subsequently, the evolution and development of different types of mimicomes in recent decades are elucidated exhaustedly, from the natural enzyme-based mimicomes (immobilized enzyme and vesicle mimicomes) to the nanozyme-based mimicomes and enzyme-nanozyme hybrid mimicomes. In conclusion, the remaining challenges in the design of multifunctional mimicomes and their potential applications are summarized, offering insights into their future prospects.
Collapse
Affiliation(s)
- Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
2
|
Zhang Y, Lu Z, Guo J, Wang Q, Zhang X, Yang H, Li X. Advanced Carriers for Precise Delivery and Therapeutic Mechanisms of Traditional Chinese Medicines: Integrating Spatial Multi-Omics and Delivery Visualization. Adv Healthc Mater 2025; 14:e2403698. [PMID: 39828637 DOI: 10.1002/adhm.202403698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/01/2024] [Indexed: 01/22/2025]
Abstract
The complex composition of traditional Chinese medicines (TCMs) has posed challenges for in-depth study and global application, despite their abundance of bioactive compounds that make them valuable resources for disease treatment. To overcome these obstacles, it is essential to modernize TCMs by focusing on precise disease treatment. This involves elucidating the structure-activity relationships within their complex compositions, ensuring accurate in vivo delivery, and monitoring the delivery process. This review discusses the research progress of TCMs in precision disease treatment from three perspectives: spatial multi-omics technology for precision therapeutic activity, carrier systems for precise in vivo delivery, and medical imaging technology for visualizing the delivery process. The aim is to establish a novel research paradigm that advances the precision therapy of TCMs.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Qing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, P. R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process, Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, 100029, P. R. China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, P. R. China
| |
Collapse
|
3
|
Marinho A, Reis S, Nunes C. On the design of cell membrane-coated nanoparticles to treat inflammatory conditions. NANOSCALE HORIZONS 2024; 10:38-55. [PMID: 39499543 DOI: 10.1039/d4nh00457d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Biomimetic-based drug delivery systems (DDS) attempt to recreate the complex interactions that occur naturally between cells. Cell membrane-coated nanoparticles (CMCNPs) have been one of the main strategies in this area to prevent opsonization and clearance. Moreover, coating nanoparticles with cell membranes allows them to acquire functions and properties inherent to the mother cells. In particular, cells from bloodstream show to have specific advantages depending on the cell type to be used for that application, specifically in cases of chronic inflammation. Thus, this review focuses on the biomimetic strategies that use membranes from blood cells to target and treat inflammatory conditions.
Collapse
Affiliation(s)
- Andreia Marinho
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
- LAQV, REQUIMTE, Faculdade de Ciências, Universidade do Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
| | - Cláudia Nunes
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal
| |
Collapse
|
4
|
Shen H, Ouyang Y, Zhang L, Li J, Wang S. Blood Cell Membrane-Coated Nanomaterials as a Versatile Biomimetic Nanoplatform for Antitumor Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1757. [PMID: 39513837 PMCID: PMC11548044 DOI: 10.3390/nano14211757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The application of nanomaterials in tumor therapy is increasingly widespread, offering more possibilities for enhanced tumor therapy. However, the unclear biological distribution and metabolism of nanomaterials may lead to immune rejection or inflammatory reactions, posing numerous challenges to their clinical translation. The rich diversity and multifaceted functions of blood cells offer promising biological avenues for enhancing the application of nanoparticles in cancer therapy. Blood cell membranes, being made of naturally found components in the body, exhibit significant biocompatibility, which can reduce the body's immune rejection response, extend the drug's residence time in the bloodstream, and enhance its bioavailability. Integrating blood cell membranes with nanomaterials enhances tumor therapy by improving targeted delivery, prolonging circulation time, and evading immune responses. This review summarizes recent advancements in the application of blood cell membrane-coated nanomaterials for antitumor therapy, with a particular focus on their use in photodynamic and photothermal treatments. Additionally, it explores their potential for synergistic effects when combined with other therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | - Jing Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; (H.S.); (L.Z.)
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; (H.S.); (L.Z.)
| |
Collapse
|
5
|
Li J, Xing H, Meng F, Liu T, Hong X, Han X, Dong Y, Li M, Wang Z, Zhang S, Cui C, Zheng A. Virus-Mimetic Extracellular-Vesicle Vaccine Boosts Systemic and Mucosal Immunity via Immune Recruitment. ACS NANO 2024. [PMID: 39013102 DOI: 10.1021/acsnano.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Mucosal vaccines can prevent viruses from infecting the respiratory mucosa, rather than only curtailing infection and protecting against the development of disease symptoms. The SARS-CoV-2 spike receptor-binding domain (RBD) is a compelling vaccine target but is undermined by suboptimal mucosal immunogenicity. Here, we report a SARS-CoV-2-mimetic extracellular-vesicle vaccine developed using genetic engineering and dendritic cell membrane budding. After mucosal immunization, the vaccine recruits antigen-presenting cells rapidly initiating a strong innate immune response. Notably, it obviates the need for adjuvants and can induce germinal center formation through both intramuscular and intratracheal vaccination. It not only elicits high levels of RBD-specific antibodies but also stimulates extensive cellular immunity in the respiratory mucosa. A sequential immunization strategy, starting with an intramuscular injection followed by an intratracheal booster, significantly bolsters mucosal immunity with high levels of IgA and tissue-resident memory T cell responses, thereby establishing a formidable defense against pseudovirus infection.
Collapse
Affiliation(s)
- Jingru Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Haonan Xing
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fan Meng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ting Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Xiaoxuan Hong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Xiaolu Han
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yuhan Dong
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meng Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zengming Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shuang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Aiping Zheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
6
|
Huang X, Zhang W. Macrophage membrane-camouflaged biomimetic nanovesicles for targeted treatment of arthritis. Ageing Res Rev 2024; 95:102241. [PMID: 38387516 DOI: 10.1016/j.arr.2024.102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Arthritis has become the most common joint disease globally. Current attention has shifted towards preventing the disease and exploring pharmaceutical and surgical treatments for early-stage arthritis. M2 macrophages are known for their anti-inflammatory properties and their ability to support cartilage repair, offering relief from arthritis. Whereas, it remains a great challenge to promote the beneficial secretion of M2 macrophages to prevent the progression of arthritis. Therefore, it is warranted to investigate new strategies that could use the functions of M2 macrophages and enhance its therapeutic effects. This review aims to explore the macrophage cell membrane-coated biomimetic nanovesicles for targeted treatment of arthritis such as osteoarthritis (OA), rheumatoid arthritis (RA), and gouty arthritis (GA). Cell membrane-camouflaged biomimetic nanovesicle has attracted increasing attention, which successfully combine the advantages and properties of both cell membrane and delivered drug. We discuss the roles of macrophages in the pathophysiology and therapeutic targets of arthritis. Then, the common preparation strategies of macrophage membrane-coated nanovesicles are concluded. Moreover, we investigate the applications of macrophage cell membrane-camouflaged nanovesicles for arthritis, such as OA, RA, and GA. Taken together, macrophage cell membrane-camouflaged nanovesicles hold the tremendous prospect for biomedical applications in the targeted treatment of arthritis.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
7
|
Mao W, Zhang L, Wang Y, Sun S, Wu J, Sun J, Zou X, Chen M, Zhang G. Cisplatin induces acute kidney injury by downregulating miR-30e-5p that targets Galnt3 to activate the AMPK signaling pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:1567-1580. [PMID: 38010663 DOI: 10.1002/tox.24054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
Cisplatin nephrotoxicity is an etiological factor for acute kidney injury (AKI). MicroRNA (miRNA) expression is dysregulated in cisplatin-induced AKI (cAKI) although the underlying mechanisms are unclear. A cAKI model was established by intraperitoneally injecting cisplatin, and key miRNAs were screened using high-throughput miRNA sequencing. The functions of key miRNAs were determined using the cell viability, live/dead, reactive oxygen species (ROS), and 5-ethynyl-2'-deoxyuridine (EdU) proliferation assays. Additionally, the macrophage membrane was wrapped around a metal-organic framework (MOF) loaded with miRNA agomir to develop a novel composite material, macrophage/MOF/miRNA agomir nanoparticles (MMA NPs). High-throughput miRNA sequencing revealed that miR-30e-5p is a key miRNA that is downregulated in cAKI. The results of in vitro experiments demonstrated that miR-30e-5p overexpression partially suppressed the cisplatin-induced or lipopolysaccharide (LPS)-induced downregulation of cell viability, proliferation, upregulation of ROS production, and cell death. Meanwhile, the results of in vivo and in vitro experiments demonstrated that MMA NPs alleviated cAKI by exerting anti-inflammatory effects. Mechanistically, cisplatin downregulates the expression of miR-30e-5p, and the downregulated miR-30e-5p can target Galnt3 to activate the adenosine 5'-monophosphate activated protein kinase (AMPK) signaling pathway, which promotes the progression of AKI. Our study found that miR-30e-5p is a key downregulated miRNA in cAKI. The downregulated miR-30e-5p promotes AKI progression by targeting Galnt3 to activate the AMPK signaling pathway. The newly developed MMA NPs were found to have protective effects on cAKI, suggesting a potential novel strategy for preventing cAKI.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
- Department of Urology, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Lei Zhang
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
| | - Yiduo Wang
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
| | - Si Sun
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
| | - Jianping Wu
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
- Department of Urology, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyu Zou
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Ming Chen
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
- Department of Urology, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Guangyuan Zhang
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
| |
Collapse
|
8
|
Ijaz M, Aslam B, Hasan I, Ullah Z, Roy S, Guo B. Cell membrane-coated biomimetic nanomedicines: productive cancer theranostic tools. Biomater Sci 2024; 12:863-895. [PMID: 38230669 DOI: 10.1039/d3bm01552a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
As the second-leading cause of human death, cancer has drawn attention in the area of biomedical research and therapy from all around the world. Certainly, the development of nanotechnology has made it possible for nanoparticles (NPs) to be used as a carrier for delivery systems in the treatment of tumors. This is a biomimetic approach established to craft remedial strategies comprising NPs cloaked with membrane obtained from various natural cells like blood cells, bacterial cells, cancer cells, etc. Here we conduct an in-depth exploration of cell membrane-coated NPs (CMNPs) and their extensive array of applications including drug delivery, vaccination, phototherapy, immunotherapy, MRI imaging, PET imaging, multimodal imaging, gene therapy and a combination of photothermal and chemotherapy. This review article provides a thorough summary of the most recent developments in the use of CMNPs for the diagnosis and treatment of cancer. It critically assesses the state of research while recognizing significant accomplishments and innovations. Additionally, it indicates ongoing problems in clinical translation and associated queries that warrant deeper research. By doing so, this study encourages creative thinking for future projects in the field of tumor therapy using CMNPs while also educating academics on the present status of CMNP research.
Collapse
Affiliation(s)
- Muhammad Ijaz
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
- Institute of Microbiology, Government College University Faisalabad Pakistan, Pakistan
| | - Bilal Aslam
- Institute of Microbiology, Government College University Faisalabad Pakistan, Pakistan
| | - Ikram Hasan
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Zia Ullah
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen-518055, China.
| |
Collapse
|
9
|
Zhang S, Wang Y, Zhang S, Huang C, Ding Q, Xia J, Wu D, Gao W. Emerging Anesthetic Nanomedicines: Current State and Challenges. Int J Nanomedicine 2023; 18:3913-3935. [PMID: 37489141 PMCID: PMC10363368 DOI: 10.2147/ijn.s417855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
Anesthetics, which include both local and general varieties, are a unique class of drugs widely utilized in clinical surgery to alleviate pain and promote relaxation in patients. Although numerous anesthetics and their traditional formulations are available in the market, only a select few exhibit excellent anesthetic properties that meet clinical requirements. The main challenges are the potential toxic and adverse effects of anesthetics, as well as the presence of the blood-brain barrier (BBB), which makes it difficult for most general anesthetics to effectively penetrate to the brain. Loading anesthetics onto nanocarriers as anesthetic nanomedicines might address these challenges and improve anesthesia effectiveness, reduce toxic and adverse effects, while significantly enhance the efficiency of general anesthetics passing through the BBB. Consequently, anesthetic nanomedicines play a crucial role in the field of anesthesia. Despite their significance, research on anesthetic nanomedicines is still in its infancy, especially when compared to other types of nanomedicines in terms of depth and breadth. Although local anesthetic nanomedicines have received considerable attention and essentially meet clinical needs, there are few reported instances of nanomedicines for general anesthetics. Given the extensive usage of anesthetics and the many of them need for improved performance, emerging anesthetic nanomedicines face both unparalleled opportunities and considerable challenges in terms of theory and technology. Thus, a comprehensive summary with systematic analyses of anesthetic nanomedicines is urgently required. This review provides a comprehensive summary of the classification, properties, and research status of anesthetic nanomedicines, along with an exploration of their opportunities and challenges. In addition, future research directions and development prospects are discussed. It is hoped that researchers from diverse disciplines will collaborate to study anesthetic nanomedicines and develop them as a valuable anesthetic dosage form for clinical surgery.
Collapse
Affiliation(s)
- Shuo Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Yishu Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Shuai Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Chengqi Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Qiyang Ding
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Ji Xia
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| |
Collapse
|
10
|
Chang W, Shen J, Liu Z, Chen Q. Application of organic nanocarriers for intraocular drug delivery. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:259-266. [PMID: 37476937 PMCID: PMC10409895 DOI: 10.3724/zdxbyxb-2023-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/31/2023] [Indexed: 07/22/2023]
Abstract
The application of intraocular drug delivery is usually limited due to special anatomical and physiological barriers, and the elimination mechanisms in the eye. Organic nano-drug delivery carriers exhibit excellent adhesion, permeability, targeted modification and controlled release abilities to overcome the obstacles and improve the efficiency of drug delivery and bioavailability. Solid lipid nanoparticles can entrap the active components in the lipid structure to improve the stability of drugs and reduce the production cost. Liposomes can transport hydrophobic or hydrophilic molecules, including small molecules, proteins and nucleic acids. Compared with linear macromolecules, dendrimers have a regular structure and well-defined molecular mass and size, which can precisely control the molecular shape and functional groups. Degradable polymer materials endow nano-delivery systems a variety of size, potential, morphology and other characteristics, which enable controlled release of drugs and are easy to modify with a variety of ligands and functional molecules. Organic biomimetic nanocarriers are highly optimized through evolution of natural particles, showing better biocompatibility and lower toxicity. In this article, we summarize the advantages of organic nanocarriers in overcoming multiple barriers and improving the bioavailability of drugs, and highlight the latest research progresses on the application of organic nanocarriers for treatment of ocular diseases.
Collapse
Affiliation(s)
- Wanwan Chang
- Institute of Functional Nano & Soft Materials, Soochow University, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Suzhou 215123, Jiangsu Province, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Macau 999078, China
| | - Jingjing Shen
- Institute of Functional Nano & Soft Materials, Soochow University, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Suzhou 215123, Jiangsu Province, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials, Soochow University, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Suzhou 215123, Jiangsu Province, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Macau 999078, China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials, Soochow University, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Suzhou 215123, Jiangsu Province, China.
| |
Collapse
|
11
|
Xiao M, Tang Q, Zeng S, Yang Q, Yang X, Tong X, Zhu G, Lei L, Li S. Emerging biomaterials for tumor immunotherapy. Biomater Res 2023; 27:47. [PMID: 37194085 PMCID: PMC10189985 DOI: 10.1186/s40824-023-00369-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The immune system interacts with cancer cells in various intricate ways that can protect the individual from overproliferation of cancer cells; however, these interactions can also lead to malignancy. There has been a dramatic increase in the application of cancer immunotherapy in the last decade. However, low immunogenicity, poor specificity, weak presentation efficiency, and off-target side effects still limit its widespread application. Fortunately, advanced biomaterials effectively contribute immunotherapy and play an important role in cancer treatment, making it a research hotspot in the biomedical field. MAIN BODY This review discusses immunotherapies and the development of related biomaterials for application in the field. The review first summarizes the various types of tumor immunotherapy applicable in clinical practice as well as their underlying mechanisms. Further, it focuses on the types of biomaterials applied in immunotherapy and related research on metal nanomaterials, silicon nanoparticles, carbon nanotubes, polymer nanoparticles, and cell membrane nanocarriers. Moreover, we introduce the preparation and processing technologies of these biomaterials (liposomes, microspheres, microneedles, and hydrogels) and summarize their mechanisms when applied to tumor immunotherapy. Finally, we discuss future advancements and shortcomings related to the application of biomaterials in tumor immunotherapy. CONCLUSION Research on biomaterial-based tumor immunotherapy is booming; however, several challenges remain to be overcome to transition from experimental research to clinical application. Biomaterials have been optimized continuously and nanotechnology has achieved continuous progression, ensuring the development of more efficient biomaterials, thereby providing a platform and opportunity for breakthroughs in tumor immunotherapy.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
12
|
Krishnan N, Peng FX, Mohapatra A, Fang RH, Zhang L. Genetically engineered cellular nanoparticles for biomedical applications. Biomaterials 2023; 296:122065. [PMID: 36841215 PMCID: PMC10542936 DOI: 10.1016/j.biomaterials.2023.122065] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
In recent years, nanoparticles derived from cellular membranes have been increasingly explored for the prevention and treatment of human disease. With their flexible design and ability to interface effectively with the surrounding environment, these biomimetic nanoparticles can outperform their traditional synthetic counterparts. As their popularity has increased, researchers have developed novel ways to modify the nanoparticle surface to introduce new or enhanced capabilities. Moving beyond naturally occurring materials derived from wild-type cells, genetic manipulation has proven to be a robust and flexible method by which nanoformulations with augmented functionalities can be generated. In this review, an overview of genetic engineering approaches to express novel surface proteins is provided, followed by a discussion on the various biomedical applications of genetically modified cellular nanoparticles.
Collapse
Affiliation(s)
- Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Fei-Xing Peng
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Animesh Mohapatra
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Zhang E, Phan P, Zhao Z. Cellular nanovesicles for therapeutic immunomodulation: A perspective on engineering strategies and new advances. Acta Pharm Sin B 2023; 13:1789-1827. [PMID: 37250173 PMCID: PMC10213819 DOI: 10.1016/j.apsb.2022.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023] Open
Abstract
Cellular nanovesicles which are referred to as cell-derived, nanosized lipid bilayer structures, have emerged as a promising platform for regulating immune responses. Owing to their outstanding advantages such as high biocompatibility, prominent structural stability, and high loading capacity, cellular nanovesicles are suitable for delivering various immunomodulatory molecules, such as small molecules, nucleic acids, peptides, and proteins. Immunomodulation induced by cellular nanovesicles has been exploited to modulate immune cell behaviors, which is considered as a novel cell-free immunotherapeutic strategy for the prevention and treatment of diverse diseases. Here we review emerging concepts and new advances in leveraging cellular nanovesicles to activate or suppress immune responses, with the aim to explicate their applications for immunomodulation. We overview the general considerations and principles for the design of engineered cellular nanovesicles with tailored immunomodulatory activities. We also discuss new advances in engineering cellular nanovesicles as immunotherapies for treating major diseases.
Collapse
Affiliation(s)
- Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Philana Phan
- 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
|
14
|
Huang X, Guo H, Wang L, Zhang Z, Zhang W. Biomimetic cell membrane-coated nanocarriers for targeted siRNA delivery in cancer therapy. Drug Discov Today 2023; 28:103514. [PMID: 36736580 DOI: 10.1016/j.drudis.2023.103514] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Small interfering RNA (siRNA) therapeutics for cancer are a focus of increasing research interest. However, the major obstacle to their clinical application is the targeted delivery of siRNA to cancer cells at desirable levels. Cell membrane-coated nanocarriers have the advantage of combining the properties of both cell membranes and nanoparticles (NPs). In this review, we highlight the most common RNAi therapeutics and the extracellular and intracellular barriers to siRNA delivery. Moreover, we discuss clinical applications of different cell membrane-coated nanocarriers for targeted siRNA delivery, including cancer cell membranes (CCMs), platelet membranes, erythrocyte membranes, stem cell membranes, exosome membranes, and hybrid membranes. Taken together, biomimetic cell membrane-coated nanotechnology is a promising strategy for targeted siRNA delivery for cancer treatment.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Haoyu Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lutong Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhicai Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
15
|
Yu H, Wu M, Chen S, Song M, Yue Y. Biomimetic nanoparticles for tumor immunotherapy. Front Bioeng Biotechnol 2022; 10:989881. [PMID: 36440446 PMCID: PMC9682960 DOI: 10.3389/fbioe.2022.989881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/11/2023] Open
Abstract
Currently, tumor treatment research still focuses on the cancer cells themselves, but the fact that the immune system plays an important role in inhibiting tumor development cannot be ignored. The activation of the immune system depends on the difference between self and non-self. Unfortunately, cancer is characterized by genetic changes in the host cells that lead to uncontrolled cell proliferation and evade immune surveillance. Cancer immunotherapy aims to coordinate a patient's immune system to target, fight, and destroy cancer cells without destroying the normal cells. Nevertheless, antitumor immunity driven by the autoimmune system alone may be inadequate for treatment. The development of drug delivery systems (DDS) based on nanoparticles can not only promote immunotherapy but also improve the immunosuppressive tumor microenvironment (ITM), which provides promising strategies for cancer treatment. However, conventional nano drug delivery systems (NDDS) are subject to several limitations in clinical transformation, such as immunogenicity and the potential toxicity risks of the carrier materials, premature drug leakage at off-target sites during circulation and drug load content. In order to address these limitations, this paper reviews the trends and progress of biomimetic NDDS and discusses the applications of each biomimetic system in tumor immunotherapy. Furthermore, we review the various combination immunotherapies based on biomimetic NDDS and key considerations for clinical transformation.
Collapse
Affiliation(s)
- Hanqing Yu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Wu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yulin Yue
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Ma J, Jiang L, Liu G. Cell membrane-coated nanoparticles for the treatment of bacterial infection. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1825. [PMID: 35725897 DOI: 10.1002/wnan.1825] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 06/15/2023]
Abstract
Despite the enormous success of antibiotics in antimicrobial therapy, the rapid emergence of antibiotic resistance and the complexity of the bacterial infection microenvironment make traditional antibiotic therapy face critical challenges against resistant bacteria, antitoxin, and intracellular infections. Consequently, there is a critical need to design antimicrobial agents that target infection microenvironment and alleviate antibiotic resistance. Cell membrane-coated nanoparticles (CMCNPs) are biomimetic materials that can be obtained by wrapping the cell membrane vesicles directly onto the surface of the nanoparticles (NPs) through physical means. Incorporating the biological functions of cell membrane vesicles and the superior physicochemical properties of NPs, CMCNPs have shown great promise in recent years for targeting infections, neutralizing bacterial toxins, and designing bacterial infection vaccines. This review highlights topics where CMCNPs present great value in advancing the treatment of bacterial infections, including drug delivery, detoxification, and vaccination. Lastly, we discuss the future hurdles and prospects of translating this technique into clinical practice, providing a comprehensive review of the technological developments of CMCNPs in the treatment of bacterial infections. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Jiaxin Ma
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Lai Jiang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Gang Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
17
|
Han S, Bi S, Guo T, Sun D, Zou Y, Wang L, Song L, Chu D, Liao A, Song X, Yu Z, Guo J. Nano co-delivery of Plumbagin and Dihydrotanshinone I reverses immunosuppressive TME of liver cancer. J Control Release 2022; 348:250-263. [PMID: 35660631 DOI: 10.1016/j.jconrel.2022.05.057] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is resistant to current immunotherapy. This poor outcome mainly results from the immunosuppressive characteristics of tumor microenvironment (TME). Accumulating evidence indicates that some chemotherapy agents trigger immunogenic cell death (ICD), providing a promising strategy to remodel the immunosuppressive TME. The role of Plumbagin (PLB, a naphthoquinone compound from Plumbago zeylanica L.) as the ICD inducer for HCC cells was confirmed in this study. Dihydrotanshinone I (DIH, a phenanthraquinone compound of Salvia miltiorrhiza) functioned as the ICD enhancer by generating the reactive oxygen species (ROS). A poly(D,L-lactic-co-glycolic acid) (PLGA)-based nanoparticle (NP) was used to co-encapsulate PLB, DIH and NH4HCO3 (a pH sensitive adjuvant). This NP was further coated with the mannose-inserted erythrocyte membrane to produce a nanoformulation. This nanoformulation significantly increased the half-life and tumor targeting of two drugs in orthotopic HCC mice, generating chemo-immunotherapeutic effects for reversal of immunosuppressive TME. Consequently, the biomimetic nanoformulation loaded with low doses of PLB and DIH achieved significantly longer survival of HCC mice, without causing toxic signs. Our study demonstrates a promising strategy for remodeling the immunosuppressive TME of liver cancer.
Collapse
Affiliation(s)
- Shulan Han
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Shengnan Bi
- Department of Pharmacy, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Tingting Guo
- Department of Pharmacy, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Dandan Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Lingzhi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Liu Song
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Di Chu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Anqi Liao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xiaohuan Song
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; Third-Grade Laboratory of Chinese Medicine Chemistry, National Administration of Traditional Chinese Medicine, Jilin University, Changchun 130021, China; Jilin Provincial Key Experiment Education Center for Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
18
|
Dhas N, García MC, Kudarha R, Pandey A, Nikam AN, Gopalan D, Fernandes G, Soman S, Kulkarni S, Seetharam RN, Tiwari R, Wairkar S, Pardeshi C, Mutalik S. Advancements in cell membrane camouflaged nanoparticles: A bioinspired platform for cancer therapy. J Control Release 2022; 346:71-97. [PMID: 35439581 DOI: 10.1016/j.jconrel.2022.04.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022]
Abstract
The idea of employing natural cell membranes as a coating medium for nanoparticles (NPs) endows man-made vectors with natural capabilities and benefits. In addition to retaining the physicochemical characteristics of the NPs, the biomimetic NPs also have the functionality of source cell membranes. It has emerged as a promising approach to enhancing the properties of NPs for drug delivery, immune evasion, imaging, cancer-targeting, and phototherapy sensitivity. Several studies have been reported with a multitude of approaches to reengineering the surface of NPs using biological membranes. Owing to their low immunogenicity and intriguing biomimetic properties, cell-membrane-based biohybrid delivery systems have recently gained a lot of interest as therapeutic delivery systems. This review summarises different kinds of biomimetic NPs reported so far, their fabrication aspects, and their application in the biomedical field. Finally, it briefs on the latest advances available in this biohybrid concept.
Collapse
Affiliation(s)
- Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Mónica C García
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Ciudad Universitaria, X5000HUA Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Ciudad Universitaria, X5000HUA Córdoba, Argentina
| | - Ritu Kudarha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Gasper Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Raviraja N Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Ruchi Tiwari
- Pranveer Singh Institute of Technology, Kanpur, Uttar Pradesh 209305, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, 400056, India
| | - Chandrakantsing Pardeshi
- R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India.
| |
Collapse
|
19
|
Nehru S, Misra R, Bhaswant M. Multifaceted Engineered Biomimetic Nanorobots Toward Cancer Management. ACS Biomater Sci Eng 2022; 8:444-459. [PMID: 35118865 DOI: 10.1021/acsbiomaterials.1c01352] [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] [Indexed: 12/25/2022]
Abstract
The noteworthy beneficiary to date in nanotechnology is cancer management. Nanorobots are developed as the result of advancements in the nanostructure, robotics, healthcare, and computer systems. These devices at the nanoscale level are beneficial in the prevention, diagnosis, and treatment of various health conditions notably cancer. Though these structures have distinct potentialities, the usage of inorganic substances in their construction can affect their performance and can cause health issues in the body. To overcome this, naturally inspired substances are incorporated in the fabrication process of nanorobots termed biomimetic nanorobots that can overcome the immunological responses and reduce the side effects with effective functionalization. These biomimetic nanorobots can widen the opportunities in cancer imaging and therapy. Herein, an up-to-date review of biomimetic nanorobots along with their applications in cancer management is provided. Furthermore, the safety issues and future directions of biomimetic nanorobots to achieve clinical translation are also stated.
Collapse
Affiliation(s)
- Sushmitha Nehru
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai-600119, India
| | - Ranjita Misra
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai-600119, India
| | - Maharshi Bhaswant
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai-600119, India
| |
Collapse
|
20
|
Xu WJ, Cai JX, Li YJ, Wu JY, Xiang D. Recent progress of macrophage vesicle-based drug delivery systems. Drug Deliv Transl Res 2022; 12:2287-2302. [PMID: 34984664 DOI: 10.1007/s13346-021-01110-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 12/13/2022]
Abstract
Nanoparticle drug delivery systems (NDDSs) are promising platforms for efficient delivery of drugs. In the past decades, many nanomedicines have received clinical approval and completed translation. With the rapid advance of nanobiotechnology, natural vectors are emerging as novel strategies to carry and delivery nanoparticles and drugs for biomedical applications. Among diverse types of cells, macrophage is of great interest for their essential roles in inflammatory and immune responses. Macrophage-derived vesicles (MVs), including exosomes, microvesicles, and those from reconstructed membranes, may inherit the chemotactic migration ability and high biocompatibility. The unique properties of MVs make them competing candidates as novel drug delivery systems for precision nanomedicine. In this review, the advantages and disadvantages of existing NDDSs and MV-based drug delivery systems (MVDDSs) were compared. Then, we summarized the potential applications of MVDDSs and discuss future perspectives. The development of MVDDS may provide avenues for the treatment of diseases involving an inflammatory process.
Collapse
Affiliation(s)
- Wen-Jie Xu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Jia-Xin Cai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Jun-Yong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China.,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China. .,Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Hunan Province, Changsha, China.
| |
Collapse
|
21
|
Zhang M, Bai Y, Xu C, Lin J, Jin J, Xu A, Lou JN, Qian C, Yu W, Wu Y, Qi Y, Tao H. Novel optimized drug delivery systems for enhancing spinal cord injury repair in rats. Drug Deliv 2021; 28:2548-2561. [PMID: 34854786 PMCID: PMC8648032 DOI: 10.1080/10717544.2021.2009937] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Effective and accurate delivery of drugs to tissue with spinal cord injury (SCI) is the key to rehabilitating neurological deficits. Sustained-release microspheres (MS) have excellent degradability and can aid in the long-term release of drugs. However, the burst release phenomenon can cause unexpected side effects. Herein, we developed and optimized an injectable poly(lactic-co-glycolic acid) (PLGA) MS loaded with melatonin(Mel), which were mixed further with Laponite hydrogels (Lap/MS@Mel, a micro-gel compound) in order to reduce the burst release of MS. Thus, these MS were able to achieve stable and prolonged Mel release, as well as synergistic Lap hydrogel in order to repair neural function in SCI by in situ injection. In clinical practice, patients with SCI have complicated conditions and significant inter-individual differences, which means that a single route of administration does not meet actual clinical needs. Thus, the nanospheres are synthesized and subsequently coated with platelet membrane (PM) in order to form PM/MS@Mel (nano-PM compound) for sustained and precision-targeted delivery of Mel intravenously in the SCI. Notably, optimized microsphere delivery systems have improved Mel regulation polarization of spinal microglial/macrophages, which can reduce loss of biomaterials due to macrophage-induced immune response during implantation of spinal cord tissue. These two new delivery systems that are based on MS provide references for the clinical treatment of SCI, according to different requirements.
Collapse
Affiliation(s)
- Man Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Yang Bai
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China
| | - Chang Xu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China
| | - Jinti Lin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - JiaKang Jin
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Ankai Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Jia Nan Lou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Chao Qian
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Wei Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Yulian Wu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China
| | - Yiying Qi
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| | - Huimin Tao
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, PR China.,Orthopedics Research Institute of Zhejiang University, Hangzhou City, PR China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, PR China
| |
Collapse
|
22
|
Pan H, Zheng M, Ma A, Liu L, Cai L. Cell/Bacteria-Based Bioactive Materials for Cancer Immune Modulation and Precision Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2100241. [PMID: 34121236 DOI: 10.1002/adma.202100241] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/24/2021] [Indexed: 06/12/2023]
Abstract
Numerous clinical trials for cancer precision medicine research are limited due to the drug resistance, side effects, and low efficacy. Unsatisfactory outcomes are often caused by complex physiologic barriers and abnormal immune events in tumors, such as tumor target alterations and immunosuppression. Cell/bacteria-derived materials with unique bioactive properties have emerged as attractive tools for personalized therapy in cancer. Naturally derived bioactive materials, such as cell and bacterial therapeutic agents with native tropism or good biocompatibility, can precisely target tumors and effectively modulate immune microenvironments to inhibit tumors. Here, the recent advances in the development of cell/bacteria-based bioactive materials for immune modulation and precision therapy in cancer are summarized. Cell/bacterial constituents, including cell membranes, bacterial vesicles, and other active substances have inherited their unique targeting properties and antitumor capabilities. Strategies for engineering living cell/bacteria to overcome complex biological barriers and immunosuppression to promote antitumor efficacy are also summarized. Moreover, past and ongoing trials involving personalized bioactive materials and promising agents such as cell/bacteria-based micro/nano-biorobotics are further discussed, which may become another powerful tool for treatment in the near future.
Collapse
Affiliation(s)
- Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mingbin Zheng
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518112, P. R. China
| | - Aiqing Ma
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
23
|
Çelik P, Derkuş B, Erdoğan K, Barut D, Manga EB, Yıldırım Y, Pecha S, Çabuk A. Bacterial membrane vesicle functions, laboratory methods, and applications. Biotechnol Adv 2021; 54:107869. [PMID: 34793882 DOI: 10.1016/j.biotechadv.2021.107869] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022]
Abstract
Bacterial membrane vesicles are cupped-shaped structures formed by bacteria in response to environmental stress, genetic alteration, antibiotic exposure, and others. Due to the structural similarities shared with the producer organism, they can retain certain characteristics like stimulating immune responses. They are also able to carry molecules for long distances, without changes in the concentration and integrity of the molecule. Bacteria originally secrete membrane vesicles for gene transfer, excretion, cell to cell interaction, pathogenesis, and protection against phages. These functions are unique and have several innovative applications in the pharmaceutical industry that have attracted both scientific and commercial interest.This led to the development of efficient methods to artificially stimulate vesicle production, purification, and manipulation in the lab at nanoscales. Also, for specific applications, engineering methods to impart pathogen antigens against specific diseases or customization as cargo vehicles to deliver payloads to specific cells have been reported. Many applications of bacteria membrane vesicles are in cancer drugs, vaccines, and adjuvant development with several candidates in clinical trials showing promising results. Despite this, applications in therapy and commercialization stay timid probably due to some challenges one of which is the poor understanding of biogenesis mechanisms. Nevertheless, so far, bacterial membrane vesicles seem to be a reliable and cost-efficient technology with several therapeutic applications. Research toward characterizing more membrane vesicles, genetic engineering, and nanotechnology will enable the scope of applications to widen. This might include solutions to other currently faced medical and healthcare-related challenges.
Collapse
Affiliation(s)
- PınarAytar Çelik
- Environmental Protection and Control Program, Eskişehir Osmangazi University, Eskişehir 26110, Turkey; Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Burak Derkuş
- Department of Chemistry, Faculty of Science, Ankara University, 06560 Ankara, Turkey
| | - Kübra Erdoğan
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Dilan Barut
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Enuh Blaise Manga
- Department of Biotechnology and Biosafety, Graduate School of Natural and Applied Science, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Yalın Yıldırım
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Simon Pecha
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Ahmet Çabuk
- Department of Biology, Faculty of Science and Letter, Eskişehir Osmangazi University, Eskişehir 26040, Turkey
| |
Collapse
|
24
|
Luo ZW, Xia K, Liu YW, Liu JH, Rao SS, Hu XK, Chen CY, Xu R, Wang ZX, Xie H. Extracellular Vesicles from Akkermansia muciniphila Elicit Antitumor Immunity Against Prostate Cancer via Modulation of CD8 + T Cells and Macrophages. Int J Nanomedicine 2021; 16:2949-2963. [PMID: 33907401 PMCID: PMC8068512 DOI: 10.2147/ijn.s304515] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Prostate cancer (PCa) is one of the most common malignancies in males. Despite the success of immunotherapy in many malignant cancers, strategies are still needed to improve therapeutic efficacy in PCa. This study aimed to investigate the effects of Akkermansia muciniphila-derived extracellular vesicles (Akk-EVs) on PCa and elucidate the underlying immune-related mechanism. METHODS Akk-EVs were isolated by ultracentrifugation and intravenously injected to treat syngeneic PCa-bearing immune-competent mice. Immunophenotypic changes in immune cells, such as cytotoxic T lymphocytes and macrophages, were measured via flow cytometry analysis. Histological examination was used to detect morphological changes in major organs after Akk-EVs treatments. In vitro, flow cytometry was performed to confirm the effects of Akk-EVs on the activation of CD8+ T cells. Quantitative PCR and immunofluorescence staining were carried out to test the impact of Akk-EVs on macrophage polarization. Cell counting kit-8 (CCK-8) analysis, colony formation assays, and scratch wound healing assays were conducted to assess the effects of Akk-EVs-treated macrophages on the proliferation and invasion of PCa cells. CCK-8 assays also confirmed the impact of Akk-EVs on the viability of normal cells. RESULTS Intravenous injection of Akk-EVs in immune-competent mice reduced the tumor burden of PCa without inducing obvious toxicity in normal tissues. This treatment elevated the proportion of granzyme B-positive (GZMB+) and interferon γ-positive (IFN-γ+) lymphocytes in CD8+ T cells and caused macrophage recruitment, with increased tumor-killing M1 macrophages and decreased immunosuppressive M2 macrophages. In vitro, Akk-EVs increased the number of GZMB+CD8+ and IFN-γ+CD8+ T cells and M1-like macrophages. In addition, conditioned medium from Akk-EVs-treated macrophages suppressed the proliferation and invasion of prostate cells. Furthermore, the effective dose of Akk-EVs was well-tolerated in normal cells. CONCLUSION Our study revealed the promising prospects of Akk-EVs as an efficient and biocompatible immunotherapeutic agent for PCa treatment.
Collapse
Affiliation(s)
- Zhong-Wei Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Kun Xia
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yi-Wei Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Jiang-Hua Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Shan-Shan Rao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Xiangya Nursing School, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xiong-Ke Hu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Chun-Yuan Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhen-Xing Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Hui Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Organ Injury, Aging and Regenerative Medicine, Changsha, Hunan, People’s Republic of China
- Hunan Key Laboratory of Bone Joint Degeneration and Injury, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
25
|
Liu W, Yan Q, Xia C, Wang X, Kumar A, Wang Y, Liu Y, Pan Y, Liu J. Recent advances in cell membrane coated metal-organic frameworks (MOFs) for tumor therapy. J Mater Chem B 2021; 9:4459-4474. [PMID: 33978055 DOI: 10.1039/d1tb00453k] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In improving the tumor-targeting ability of metal-organic frameworks (MOFs) for tumor therapy and avoiding the clearance as well as capture by the immune system, there are still several challenges, which limit the development and bio-applications of MOFs. To overcome these challenges, various targeted modification strategies have been proposed. Amongst all the strategies, a promising cell membrane coating method has been explored and utilized for the syntheses of new cell membrane biomimetic MOFs (CMMs). Through such coating, various source cell membranes (e.g., red blood cell, immune cell, cancer cell, platelet, and fusion cell membranes) can be endowed with excellent properties such as long blood circulation, immune escape, and targeting ability. In the presented perspective, the synthetic method, characterization, and research progress in tumor therapy based on CMMs have been summarized. This is because, like many other technologies, the cell membrane coating technology also has several challenges to overcome. Hence, addressing and overcoming such challenges will promote and extend the bio-applications of MOFs which in the future may become a prospective carrier for cancer nano-medicine. Finally, the prospects and challenges of utilizing CMMs for tumor therapy have been discussed.
Collapse
Affiliation(s)
- Weicong Liu
- Department of Pharmacy, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China.
| | - Qianwen Yan
- Department of Pathology, The First People's Hospital of Foshan, Foshan 528000, China
| | - Chen Xia
- Department of Pharmacy, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China.
| | - Xiaoxiong Wang
- School of Civil and Environmental Engineering, Shenzhen Polytechnic, Shenzhen 518055, China.
| | - Abhinav Kumar
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226 007, India
| | - Yan Wang
- Department of Pharmacy, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China.
| | - Yiwei Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Ying Pan
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Jianqiang Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
26
|
Rayamajhi S, Marasini R, Nguyen TDT, Plattner BL, Biller D, Aryal S. Strategic reconstruction of macrophage-derived extracellular vesicles as a magnetic resonance imaging contrast agent. Biomater Sci 2020; 8:2887-2904. [DOI: 10.1039/d0bm00128g] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Reconstruction of extracellular vesicles with imaging agents allows precise downstream analysis using clinical imaging modalities, for example, MRI. This will further improve the biocompatibility of agents thereby enhancing clinical investigations.
Collapse
Affiliation(s)
- Sagar Rayamajhi
- Department of Chemistry
- Nanotechnology Innovation Center of Kansas State (NICKS)
- Kansas State University
- Manhattan
- USA
| | - Ramesh Marasini
- Department of Chemistry
- Nanotechnology Innovation Center of Kansas State (NICKS)
- Kansas State University
- Manhattan
- USA
| | - Tuyen Duong Thanh Nguyen
- Department of Chemistry
- Nanotechnology Innovation Center of Kansas State (NICKS)
- Kansas State University
- Manhattan
- USA
| | - Brandon L. Plattner
- Department of Diagnostic Medicine and Pathobiology
- College of Veterinary Medicine
- Kansas State University
- Manhattan
- USA
| | - David Biller
- Department of Clinical Sciences
- College of Veterinary Medicine
- Kansas State University
- Manhattan
- USA
| | - Santosh Aryal
- Department of Chemistry
- Nanotechnology Innovation Center of Kansas State (NICKS)
- Kansas State University
- Manhattan
- USA
| |
Collapse
|