1
|
Miao Y, Ge J, Zheng L, Liu G. Bioinspired Membrane-Based Cancer Vaccines for Immunotherapy: Progress and Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2412679. [PMID: 40255117 DOI: 10.1002/smll.202412679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/25/2025] [Indexed: 04/22/2025]
Abstract
Cancer vaccines hold promise for tumor immunotherapy, with their success hinging on effective systems to boost anti-tumor immunity. Biological membranes are not only a delivery vehicle but also a source of antigens and adjuvants, garnering growing interest in vaccine research. This review starts with an introduction to the composition and mechanisms of cancer vaccines and describes the sources, advantages/disadvantages, engineering strategies, and applications of these membrane-based platforms for cancer vaccine development. This review also offers a critical analysis and discusses the further direction of the vaccine platform in view of clinical translation for tumor immunotherapy.
Collapse
Affiliation(s)
- Yanyu Miao
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jianlin Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Longyi Zheng
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
2
|
Zhu N, Yang J. The potential of erythrocyte-mimic nanoparticles in the treatment of cardiovascular diseases. Nanomedicine (Lond) 2025:1-3. [PMID: 40166811 DOI: 10.1080/17435889.2025.2485674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Affiliation(s)
- Ni Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jing Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
3
|
Yang Y, Wang J, Zhong Y, Tian M, Zhang H. Advances in Radionuclide-Labeled Biological Carriers for Tumor Imaging and Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4316-4336. [PMID: 39792777 DOI: 10.1021/acsami.4c19059] [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: 01/12/2025]
Abstract
Biological carriers have emerged as significant tools to deliver radionuclides in nuclear medicine, providing a meaningful perspective for tumor imaging and treatment. Various radionuclide-labeled biological carriers have been developed to meet the needs of biomedical applications. This review introduces the principles of radionuclide-mediated imaging and therapy and the selected criteria of them, as well as a comprehensive description of the characteristics and functions of representative biological carriers including bacteria, cells, viruses, and their biological derivatives, emphasizing the labeled strategies of biological carriers combined with radionuclides. Subsequently, we in-depth introduce the application of radionuclide-labeled biological carriers in tumor imaging and treatment, including the imaging of the behaviors of biological carriers in vivo and tumor metastasis and the tumor treatment by radionuclide therapy, plus other strategies and radiation-induced photodynamic therapy. Finally, the challenges and prospects of radionuclide-labeled biological carriers are discussed to improve the shortcomings of this innovative platform and promote clinical transformation in the field of medical imaging.
Collapse
Affiliation(s)
- Yaozhang Yang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Jing Wang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Yan Zhong
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang 310014, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
4
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
5
|
He A, Huang Y, Cao C, Li X. Advances in drug delivery systems utilizing blood cells and their membrane-derived microvesicles. Drug Deliv 2024; 31:2425156. [PMID: 39520082 PMCID: PMC11552282 DOI: 10.1080/10717544.2024.2425156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The advancement of drug delivery systems (DDSs) in recent decades has demonstrated significant potential in enhancing the efficacy of pharmacological agents. Despite the approval of certain DDSs for clinical use, challenges such as rapid clearance from circulation, toxic accumulation in the body, and ineffective targeted delivery persist as obstacles to successful clinical application. Blood cell-based DDSs have emerged as a popular strategy for drug administration, offering enhanced biocompatibility, stability, and prolonged circulation. These DDSs are well-suited for systemic drug delivery and have played a crucial role in formulating optimal drug combinations for treating a variety of diseases in both preclinical studies and clinical trials. This review focuses on recent advancements and applications of DDSs utilizing blood cells and their membrane-derived microvesicles. It addresses the current therapeutic applications of blood cell-based DDSs at the organ and tissue levels, highlighting their successful deployment at the cellular level. Furthermore, it explores the mechanisms of cellular uptake of drug delivery vectors at the subcellular level. Additionally, the review discusses the opportunities and challenges associated with these DDSs.
Collapse
Affiliation(s)
- Andong He
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Yuye Huang
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Wang Y, Liu Y, Su X, Niu L, Li N, Xu C, Sun Z, Guo H, Shen S, Yu M. Non-pathogenic Trojan horse Nissle1917 triggers mitophagy through PINK1/Parkin pathway to discourage colon cancer. Mater Today Bio 2024; 29:101273. [PMID: 39415764 PMCID: PMC11480251 DOI: 10.1016/j.mtbio.2024.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Bacteria-mediated antitumor therapy has gained widespread attention for its innate tumor-targeting capability and excellent immune activation properties. Nevertheless, the clinical approval of bacterial therapies remains elusive primarily due to the formidable challenge of balancing safety with enhancing in vivo efficacy. In this study, leveraging the probiotic Escherichia coli Nissle1917 (EcN) emerges as a promising approach for colon cancer therapy, offering a high level of safety attributed to its lack of virulence factors and its tumor-targeting potential owing to its obligate anaerobic nature. Specifically, we delineate the erythrocyte (RBC) membrane-camouflaged EcN, termed as Trojan horse EcN@RBC, which triggers apoptosis in tumor cells by mitigating mitochondrial membrane potential (MMP) and subsequently activating the PINK1/Parkin pathway associated with mitophagy. Concurrently, the decline in MMP induced by mitophagy disrupts the mitochondrial permeability transition pore (MPTP), leading to the release of Cytochrome C and subsequent apoptosis induction. Moreover, synergistic effects were observed through the combination of the autophagy activator rapamycin, bolstering the antitumor efficacy in vivo. These findings offer novel insights into probiotic-mediated antitumor mechanisms and underscore the therapeutic potential of EcN@RBC for colon cancer patients.
Collapse
Affiliation(s)
- Yang Wang
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Yao Liu
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaomin Su
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Lili Niu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Nannan Li
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Ce Xu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Zanya Sun
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, Shanghai, 201399, China
| |
Collapse
|
7
|
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
|
8
|
Li S, Wang Y, Liu Q, Tang F, Zhang X, Yang S, Wang Q, Yang Q, Li S, Liu J, Han L, Liao Y, Yin X, Fan J, Feng H. RBC-hitchhiking PLGA nanoparticles loading β-glucan as a delivery system to enhance in vitro and in vivo immune responses in mice. Front Vet Sci 2024; 11:1462518. [PMID: 39351151 PMCID: PMC11439874 DOI: 10.3389/fvets.2024.1462518] [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: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 10/04/2024] Open
Abstract
Red blood cells (RBCs) naturally trap some bacterial pathogens in the circulation and kill them by oxidative stress. Following neutralization, the bacteria are presented to antigen-presenting cells in the spleen by the RBCs. This ability of RBCs has been harnessed to develop a system where they play a crucial role in enhancing the immune response, offering a novel approach to enhance the body's immunity. In this work, a conjugate, G-OVA, was formed by connecting β-glucan and OVA through a disulfide bond. Poly (lactic-co-glycolic acid) (PLGA) was then employed to encapsulate G-OVA, yielding G-OVA-PLGA. Finally, the nanoparticles were adsorbed onto RBCs to develop G-OVA-PLGA@RBC. The results demonstrated that the delivery of nanoparticles by RBCs enhanced the antibody response to antigens both in vitro and in vivo. The objective of this study was to investigate the increased immune activity of G-OVA-PLGA nanoparticles facilitated by RBCs transportation and to elucidate some of its underlying mechanisms. These findings are anticipated to contribute valuable insights for the development of efficient and safe immune enhancers.
Collapse
Affiliation(s)
- Sheng Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yao Wang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Qianqian Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Feng Tang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinnan Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shuyao Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Qiran Wang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Qian Yang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shanshan Li
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jie Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lu Han
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yi Liao
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xuemei Yin
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jing Fan
- College of Pharmacy, Chengdu University, Chengdu, China
| | - Haibo Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Zhou J, Jiang Z, Sun R, Pan D, Du Q, Zhou X, Chen Y, Chen Y, Peng J. Comparison of cell delivery and cell membrane camouflaged PLGA nanoparticles in the delivery of shikonin for colorectal cancer treatment. Colloids Surf B Biointerfaces 2024; 241:114017. [PMID: 38865869 DOI: 10.1016/j.colsurfb.2024.114017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Inspired by the "natural camouflage" strategy, cell-based biomimetic drug delivery systems (BDDS) have shown great potential in cancer therapy. Red blood cell (RBC) delivery vehicles and red blood cell membrane (RBCm)-camouflaged vehicles were commonly used strategies for drug delivery. We prepared shikonin-encapsulated PLGA nanoparticles (PLGA/SK) with different surface charges to obtain both RBC delivery and RBCm-camouflaged PLGA NPs. The physicochemical properties, in vivo circulation and antitumor effects of these biomimetic preparations were studied. Since the positive PLGA NPs may affect the morphology and function of RBCs, the biomimetic preparations prepared by the negative PLGA NPs showed better in vitro stability. However, positive PLGA NP-based biomimetic preparations exhibited longer circulation time and higher tumor region accumulation, leading to stronger anti-tumor effects. Meanwhile, the RBC delivery PLGA(+) NPs possessed better in vitro cytotoxicity, longer circulation time and higher tumor accumulation than RBCm-camouflaged PLGA(+) NPs. Collectively, RBC delivery vehicles possessed more potential than RBCm-camouflaged vehicles on drug delivery for tumor treatment, especially with positive NPs-loaded.
Collapse
Affiliation(s)
- Jia Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Zhaohui Jiang
- The First People's Hospital of Guiyang, Guizhou 550002, China
| | - Runbin Sun
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Jiangsu 210008, China
| | - Di Pan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Jiangsu 210006, China
| | - Xiang Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Jianqing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| |
Collapse
|
10
|
Qu Y, Chu B, Li J, Deng H, Niu T, Qian Z. Macrophage-Biomimetic Nanoplatform-Based Therapy for Inflammation-Associated Diseases. SMALL METHODS 2024; 8:e2301178. [PMID: 38037521 DOI: 10.1002/smtd.202301178] [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: 09/01/2023] [Revised: 10/23/2023] [Indexed: 12/02/2023]
Abstract
Inflammation-associated diseases are very common clinically with a high incidence; however, there is still a lack of effective treatments. Cell-biomimetic nanoplatforms have led to many breakthroughs in the field of biomedicine, significantly improving the efficiency of drug delivery and its therapeutic implications especially for inflammation-associated diseases. Macrophages are an important component of immune cells and play a critical role in the occurrence and progression of inflammation-associated diseases while simultaneously maintaining homeostasis and modulating immune responses. Therefore, macrophage-biomimetic nanoplatforms not only inherit the functions of macrophages including the inflammation tropism effect for targeted delivery of drugs and the neutralization effect of pro-inflammatory cytokines and toxins via membrane surface receptors or proteins, but also maintain the functions of the inner nanoparticles. Macrophage-biomimetic nanoplatforms are shown to have remarkable therapeutic efficacy and excellent application potential in inflammation-associated diseases. In this review, inflammation-associated diseases, the physiological functions of macrophages, and the classification and construction of macrophage-biomimetic nanoplatforms are first introduced. Next, the latest applications of different macrophage-biomimetic nanoplatforms for the treatment of inflammation-associated diseases are summarized. Finally, challenges and opportunities for future biomedical applications are discussed. It is hoped that the review will provide new ideas for the further development of macrophage-biomimetic nanoplatforms.
Collapse
Affiliation(s)
- Ying Qu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bingyang Chu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianan Li
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hanzhi Deng
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
11
|
Chen W, Tang C, Chen G, Li J, Li N, Zhang H, Di L, Wang R. Boosting Checkpoint Immunotherapy with Biomimetic Nanodrug Delivery Systems. Adv Healthc Mater 2024; 13:e2304284. [PMID: 38319961 DOI: 10.1002/adhm.202304284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/26/2024] [Indexed: 02/08/2024]
Abstract
Immune checkpoint blockade (ICB) has achieved unprecedented progress in tumor immunotherapy by blocking specific immune checkpoint molecules. However, the high biodistribution of the drug prevents it from specifically targeting tumor tissues, leading to immune-related adverse events. Biomimetic nanodrug delivery systems (BNDSs) readily applicable to ICB therapy have been widely developed at the preclinical stage to avoid immune-related adverse events. By exploiting or mimicking complex biological structures, the constructed BNDS as a novel drug delivery system has good biocompatibility and certain tumor-targeting properties. Herein, the latest findings regarding the aforementioned therapies associated with ICB therapy are highlighted. Simultaneously, prospective bioinspired engineering strategies can be designed to overcome the four-level barriers to drug entry into lesion sites. In future clinical translation, BNDS-based ICB combination therapy represents a promising avenue for cancer treatment.
Collapse
Affiliation(s)
- Wenjing Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Chenlu Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Guijin Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Hanwen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| | - Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing, 210023, China
| |
Collapse
|
12
|
Li Q, Jin M, Ding Z, Luo D, Wang S, Bao X, Liu Z, Wei W. Renal Clearable Nanodots-Engineered Erythrocytes with Enhanced Circulation and Tumor Accumulation for Photothermal Therapy of Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309278. [PMID: 38195972 DOI: 10.1002/smll.202309278] [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: 10/13/2023] [Revised: 12/06/2023] [Indexed: 01/11/2024]
Abstract
Living cell-mediated nanodelivery system is considered a promising candidate for targeted antitumor therapy; however, their use is restricted by the adverse interactions between carrier cells and nanocargos. Herein, a novel erythrocyte-based nanodelivery system is developed by assembling renal-clearable copper sulfide (CuS) nanodots on the outer membranes of erythrocytes via a lipid fusion approach, and demonstrate that it is an efficient photothermal platform against hepatocellular carcinoma. After intravenous injection of the nanodelivery system, CuS nanodots assembled on erythrocytes can be released from the system, accumulate in tumors in response to the high shear stress of bloodstream, and show excellent photothermal antitumor effect under the near infrared laser irradiation. Therefore, the erythrocyte-mediated nanodelivery system holds many advantages including prolonged blood circulation duration and enhanced tumor accumulation. Significantly, the elimination half-life of the nanodelivery system is 74.75 ± 8.77 h, which is much longer than that of nanodots (33.56 ± 2.36 h). Moreover, the other two kinds of nanodots can be well assembled onto erythrocytes to produce other erythrocyte-based hitchhiking platforms. Together, the findings promote not only the development of novel erythrocyte-based nanodelivery systems as potential platforms for tumor treatment but also their further clinical translation toward personalized healthcare.
Collapse
Affiliation(s)
- Quanxiao Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
- The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Meng Jin
- The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhen Ding
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Danfeng Luo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xingfu Bao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Zhen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
13
|
Ding Y, Xu Q, Chai Z, Wu S, Xu W, Wang J, Zhou J, Luo Z, Liu Y, Xie C, Lu L, Lu W. All-stage targeted red blood cell membrane-coated docetaxel nanocrystals for glioma treatment. J Control Release 2024; 369:325-334. [PMID: 38565395 DOI: 10.1016/j.jconrel.2024.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/08/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Challenges for glioma treatment with nanomedicines include physio-anatomical barriers (the blood-brain barrier and blood-brain tumor barrier), low drug loading capacity, and limited circulation time. Here, a red blood cell membrane-coated docetaxel drug nanocrystal (pV-RBCm-NC(DTX)), modified with pHA-VAP (pV) for all-stage targeting of glioma, was designed. The NC(DTX) core exhibited a high drug loading capacity but low in vivo stability, and the RBCm coating significantly enhanced the stability and prolonged in vivo circulation. Moreover, the Y-shaped targeting ligand pV was modified by a mild avidin-biotin interaction, which endowed RBCm-NC(DTX) with superior barrier-crossing ability and therapeutic efficacy. The integration of nanocrystal technology, cell membrane coating, and the avidin-biotin insertion method into this active targeting biomimetic formulation represents a promising drug delivery strategy for glioma.
Collapse
Affiliation(s)
- Yuan Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zhilan Chai
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd., Shanghai, 201314, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China.
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd., Shanghai, 201314, China; Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
14
|
Li Z, Xue L, Yang J, Wuttke S, He P, Lei C, Yang H, Zhou L, Cao J, Sinelshchikova A, Zheng G, Guo J, Lin J, Lei Q, Brinker CJ, Liu K, Zhu W. Synthetic Biohybrids of Red Blood Cells and Cascaded-Enzymes@ Metal-Organic Frameworks for Hyperuricemia Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305126. [PMID: 38054350 PMCID: PMC10837374 DOI: 10.1002/advs.202305126] [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: 07/26/2023] [Revised: 10/19/2023] [Indexed: 12/07/2023]
Abstract
Hyperuricemia, caused by an imbalance between the rates of production and excretion of uric acid (UA), may greatly increase the mortality rates in patients with cardiovascular and cerebrovascular diseases. Herein, for fast-acting and long-lasting hyperuricemia treatment, armored red blood cell (RBC) biohybrids, integrated RBCs with proximal, cascaded-enzymes of urate oxidase (UOX) and catalase (CAT) encapsulated within ZIF-8 framework-based nanoparticles, have been fabricated based on a super-assembly approach. Each component is crucial for hyperuricemia treatment: 1) RBCs significantly increase the circulation time of nanoparticles; 2) ZIF-8 nanoparticles-based superstructure greatly enhances RBCs resistance against external stressors while preserving native RBC properties (such as oxygen carrying capability); 3) the ZIF-8 scaffold protects the encapsulated enzymes from enzymatic degradation; 4) no physical barrier exists for urate diffusion, and thus allow fast degradation of UA in blood and neutralizes the toxic by-product H2 O2 . In vivo results demonstrate that the biohybrids can effectively normalize the UA level of an acute hyperuricemia mouse model within 2 h and possess a longer elimination half-life (49.7 ± 4.9 h). They anticipate that their simple and general method that combines functional nanomaterials with living cell carriers will be a starting point for the development of innovative drug delivery systems.
Collapse
Affiliation(s)
- Zeyu Li
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | - Liecong Xue
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | - Junxian Yang
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000P. R. China
| | - Stefan Wuttke
- BCMaterialsBasque Center for MaterialsUPV/EHU Science ParkLeioa48940Spain
- IKERBASQUEBasque Foundation for ScienceBilbao48009Spain
| | - Peiying He
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | - Chuanyi Lei
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | - Haowei Yang
- China National Tobacco CorporationNo.55 South Yuetan Boulevard Xicheng DistrictBeijing100045P. R. China
| | - Liang Zhou
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | - Jiangfan Cao
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | | | - Guansheng Zheng
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| | - Jimin Guo
- College of Materials Sciences and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Jiangguo Lin
- Medical Research InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510000P. R. China
| | - Qi Lei
- The Second Affiliated HospitalState Key Laboratory of Respiratory DiseaseGuangdong Provincial Key Laboratory of Allergy and Clinical ImmunologyGuangzhou Medical UniversityGuangzhou510260P.R. China
| | - C. Jeffrey Brinker
- Center for Micro‐Engineered Materials and the Department of Chemical and Biological EngineeringThe University of New MexicoAlbuquerqueNM87131USA
| | - Kaisheng Liu
- Guangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology)Shenzhen518020P. R. China
| | - Wei Zhu
- MOE International Joint Research Laboratory on Synthetic Biology and MedicinesSchool of Biology and Biological EngineeringSouth China University of TechnologyGuangzhou510006P. R. China
| |
Collapse
|
15
|
Ding J, Ding X, Liao W, Lu Z. Red blood cell-derived materials for cancer therapy: Construction, distribution, and applications. Mater Today Bio 2024; 24:100913. [PMID: 38188647 PMCID: PMC10767221 DOI: 10.1016/j.mtbio.2023.100913] [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: 09/30/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Cancer has become an increasingly important public health issue owing to its high morbidity and mortality rates. Although traditional treatment methods are relatively effective, they have limitations such as highly toxic side effects, easy drug resistance, and high individual variability. Meanwhile, emerging therapies remain limited, and their actual anti-tumor effects need to be improved. Nanotechnology has received considerable attention for its development and application. In particular, artificial nanocarriers have emerged as a crucial approach for tumor therapy. However, certain deficiencies persist, including immunogenicity, permeability, targeting, and biocompatibility. The application of erythrocyte-derived materials will help overcome the above problems and enhance therapeutic effects. Erythrocyte-derived materials can be acquired via the application of physical and chemical techniques from natural erythrocyte membranes, or through the integration of these membranes with synthetic inner core materials using cell membrane biomimetic technology. Their natural properties such as biocompatibility and long circulation time make them an ideal choice for drug delivery or nanoparticle biocoating. Thus, red blood cell-derived materials are widely used in the field of biomedicine. However, further studies are required to evaluate their efficacy, in vivo metabolism, preparation, design, and clinical translation. Based on the latest research reports, this review summarizes the biology, synthesis, characteristics, and distribution of red blood cell-derived materials. Furthermore, we provide a reference for further research and clinical transformation by comprehensively discussing the applications and technical challenges faced by red blood cell-derived materials in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Jianghua Ding
- Department of Hematology & Oncology, Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332005, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332005, China
| | - Xinjing Ding
- Oncology of Department, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 332000, China
| | - Weifang Liao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332005, China
- Department of Medical Laboratory, Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Zhihui Lu
- Oncology of Department, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 332000, China
| |
Collapse
|
16
|
Berikkhanova K, Taigulov E, Bokebaev Z, Kusainov A, Tanysheva G, Yedrissov A, Seredin G, Baltabayeva T, Zhumadilov Z. Drug-loaded erythrocytes: Modern approaches for advanced drug delivery for clinical use. Heliyon 2024; 10:e23451. [PMID: 38192824 PMCID: PMC10772586 DOI: 10.1016/j.heliyon.2023.e23451] [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: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Scientific organizations worldwide are striving to create drug delivery systems that provide a high local concentration of a drug in pathological tissue without side effects on healthy organs in the body. Important physiological properties of red blood cells (RBCs), such as frequent renewal ability, good oxygen carrying ability, unique shape and membrane flexibility, allow them to be used as natural carriers of drugs in the body. Erythrocyte carriers derived from autologous blood are even more promising drug delivery systems due to their immunogenic compatibility, safety, natural uniqueness, simple preparation, biodegradability and convenience of use in clinical practice. This review is focused on the achievements in the clinical application of targeted drug delivery systems based on osmotic methods of loading RBCs, with an emphasis on advancements in their industrial production. This article describes the basic methods used for encapsulating drugs into erythrocytes, key strategic approaches to the clinical use of drug-loaded erythrocytes obtained by hypotonic hemolysis. Moreover, clinical trials of erythrocyte carriers for the targeted delivery are discussed. This article explores the recent advancements and engineering approaches employed in the encapsulation of erythrocytes through hypotonic hemolysis methods, as well as the most promising inventions in this field. There is currently a shortage of reviews focused on the automation of drug loading into RBCs; therefore, our work fills this gap. Finally, further prospects for the development of engineering and technological solutions for the automatic production of drug-loaded RBCs were studied. Automated devices have the potential to provide the widespread production of RBC-encapsulated therapeutic drugs and optimize the process of targeted drug delivery in the body. Furthermore, they can expedite the widespread introduction of this innovative treatment method into clinical practice, thereby significantly expanding the effectiveness of treatment in both surgery and all areas of medicine.
Collapse
Affiliation(s)
- Kulzhan Berikkhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - Erlan Taigulov
- University Medical Center, Nazarbayev University, Astana, 010000, Kazakhstan
- Astana Medical University, Astana, 010000, Kazakhstan
| | - Zhanybek Bokebaev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
- Astana Medical University, Astana, 010000, Kazakhstan
| | - Aidar Kusainov
- Semey State Medical University, Semey, 071400, Kazakhstan
| | | | - Azamat Yedrissov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - German Seredin
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - Tolkyn Baltabayeva
- Scientific-Production Center of Transfusiology, Astana, 010000, Kazakhstan
| | - Zhaxybay Zhumadilov
- Departament of Surgery, School of Medicine, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| |
Collapse
|
17
|
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: 18] [Impact Index Per Article: 18.0] [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
|
18
|
An X, Zeng Y, Liu C, Liu G. Cellular-Membrane-Derived Vesicles for Cancer Immunotherapy. Pharmaceutics 2023; 16:22. [PMID: 38258033 PMCID: PMC10820497 DOI: 10.3390/pharmaceutics16010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
The medical community is constantly searching for new and innovative ways to treat cancer, and cellular-membrane-derived artificial vesicles are emerging as a promising avenue for cancer immunotherapy. These vesicles, which are derived from mammal and bacteria cell membranes, offer a range of benefits, including compatibility with living organisms, minimal immune response, and prolonged circulation. By modifying their surface, manipulating their genes, combining them with other substances, stimulating them externally, and even enclosing drugs within them, cellular vesicles have the potential to be a powerful tool in fighting cancer. The ability to merge drugs with diverse compositions and functionalities in a localized area is particularly exciting, as it offers a way to combine different immunotherapy treatments for maximum impact. This review contains information on the various sources of these vesicles and discusses some recent developments in cancer immunotherapy using this promising technology. While there are still obstacles to overcome, the possibilities for cellular vesicles in cancer treatment are truly exciting.
Collapse
Affiliation(s)
- Xiaoyu An
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China;
- State Key Laboratory of Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yun Zeng
- Department of Pharmacy, Xiamen Medical College, Xiamen 361023, China;
| | - Chao Liu
- State Key Laboratory of Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China;
- School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
19
|
Raza F, Zafar H, Jiang L, Su J, Yuan W, Qiu M, Paiva-Santos AC. Progress of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. Biomater Sci 2023; 12:57-91. [PMID: 37902579 DOI: 10.1039/d3bm01170d] [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: 10/31/2023]
Abstract
In recent years, considerable attention has been given to phototherapy, including photothermal and photodynamic therapy to kill tumor cells by producing heat or reactive oxygen species (ROS). It has the high merits of noninvasiveness and limited drug resistance. To fully utilize this therapy, an extraordinary nanovehicle is required to target phototherapeutic agents in the tumor cells. Nanovesicles embody an ideal strategy for drug delivery applications. Cell membrane-derived biomimetic nanovesicles represent a developing type of nanocarrier. Combining this technique with cancer phototherapy could enable a novel strategy. Herein, efforts are made to describe a comprehensive overview of cell membrane-derived biomimetic nanovesicles for cancer phototherapy. The description in this review is mainly based on representative examples of exosome-derived biomimetic nanomedicine research, ranging from their comparison with traditional nanocarriers to extensive applications in cancer phototherapy. Additionally, the challenges and future prospectives for translating these for clinical application are discussed.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Liangdi Jiang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
20
|
Rashid S, Raza M, Sharif M, Azam F, Kadry S, Kim J. White blood cell image analysis for infection detection based on virtual hexagonal trellis (VHT) by using deep learning. Sci Rep 2023; 13:17827. [PMID: 37857667 PMCID: PMC10587153 DOI: 10.1038/s41598-023-44352-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
White blood cells (WBCs) are an indispensable constituent of the immune system. Efficient and accurate categorization of WBC is a critical task for disease diagnosis by medical experts. This categorization helps in the correct identification of medical problems. In this research work, WBC classes are categorized with the help of a transform learning model in combination with our proposed virtual hexagonal trellis (VHT) structure feature extraction method. The VHT feature extractor is a kernel-based filter model designed over a square lattice. In the first step, Graft Net CNN model is used to extract features of augmented data set images. Later, the VHT base feature extractor extracts useful features. The CNN-extracted features are passed to ant colony optimization (ACO) module for optimal features acquisition. Extracted features from the VHT base filter and ACO are serially merged to create a single feature vector. The merged features are passed to the support vector machine (SVM) variants for optimal classification. Our strategy yields 99.9% accuracy, which outperforms other existing methods.
Collapse
Affiliation(s)
- Shahid Rashid
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Islamabad, 47040, Pakistan
| | - Mudassar Raza
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Islamabad, 47040, Pakistan.
| | - Muhammad Sharif
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Islamabad, 47040, Pakistan
| | - Faisal Azam
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Islamabad, 47040, Pakistan
| | - Seifedine Kadry
- Department of Applied Data Science, Noroff University College, Kristiansand, Norway
- Department of Electrical and Computer Engineering, Lebanese American University, Byblos, Lebanon
- Artificial Intelligence Research Center (AIRC), College of Engineering and Information Technology, Ajman University, Ajman, United Arab Emirates
| | - Jungeun Kim
- Department of Software, Kongju National University, Cheonan, 31080, Korea.
| |
Collapse
|
21
|
Gutierrez-Millan C, Barez Diaz C, Alvarez Vizan L, Colino CI. Evaluation of Two Osmosis-Based Methods for the Preparation of Drug Delivery Systems Based on Red Blood Cells. Pharmaceutics 2023; 15:2281. [PMID: 37765250 PMCID: PMC10536362 DOI: 10.3390/pharmaceutics15092281] [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: 07/31/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Erythrocytes have been thoroughly investigated as drug delivery systems for a wide range of therapeutic molecules and using different kinds of loading methods, outstanding the osmosis-based methods as the most used ones. Most of them involve too much handling of blood components and the immediate obtention of fresh blood. Based on our group's considerable experience in dialysis-based carrier erythrocyte preparation, this study details a simple method based on hypotonic dilution and subsequent resealing that has been developed for stavudine using packed erythrocytes from a local blood bank. Properties of the obtained carrier erythrocytes were studied in comparison to those prepared by dialysis. Erythrocytes' morphology, osmotic fragility, hematological parameters, and in vitro release profiles were evaluated. Loaded erythrocytes obtained with the proposed method did not show impaired properties in comparison with those obtained with our reference method, provided that the buffer composition remained the same. In the present work, we have optimized a simplified method for erythrocytes' drug loading, which can use blood transfusion products and could be easily automatized and scalable.
Collapse
Affiliation(s)
- Carmen Gutierrez-Millan
- Area of Pharmacy and Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Salamanca, 37007 Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Celia Barez Diaz
- Area of Pharmacy and Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Lydia Alvarez Vizan
- Area of Pharmacy and Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Clara I Colino
- Area of Pharmacy and Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Salamanca, 37007 Salamanca, Spain
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
22
|
Bianchi M, Rossi L, Pierigè F, Biagiotti S, Bregalda A, Tasini F, Magnani M. Preclinical and clinical developments in enzyme-loaded red blood cells: an update. Expert Opin Drug Deliv 2023; 20:921-935. [PMID: 37249524 DOI: 10.1080/17425247.2023.2219890] [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: 05/26/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION We have previously described the preclinical developments in enzyme-loaded red blood cells to be used in the treatment of several rare diseases, as well as in chronic conditions. AREA COVERED Since our previous publication we have seen further progress in the previously discussed approaches and, interestingly enough, in additional new studies that further strengthen the idea that red blood cell-based therapeutics may have unique advantages over conventional enzyme replacement therapies in terms of efficacy and safety. Here we highlight these investigations and compare, when possible, the reported results versus the current therapeutic approaches. EXPERT OPINION The continuous increase in the number of new potential applications and the progress from the encapsulation of a single enzyme to the engineering of an entire metabolic pathway open the field to unexpected developments and confirm the role of red blood cells as cellular bioreactors that can be conveniently manipulated to acquire useful therapeutic metabolic abilities. Positioning of these new approaches versus newly approved drugs is essential for the successful transition of this technology from the preclinical to the clinical stage and hopefully to final approval.
Collapse
Affiliation(s)
- Marzia Bianchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Luigia Rossi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- EryDel SpA, Bresso, MI, Italy
| | - Francesca Pierigè
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sara Biagiotti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessandro Bregalda
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Filippo Tasini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- EryDel SpA, Bresso, MI, Italy
| |
Collapse
|
23
|
Liao X, Gong G, Dai M, Xiang Z, Pan J, He X, Shang J, Blocki AM, Zhao Z, Shields CW, Guo J. Systemic Tumor Suppression via Macrophage-Driven Automated Homing of Metal-Phenolic-Gated Nanosponges for Metastatic Melanoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207488. [PMID: 37072673 PMCID: PMC10288275 DOI: 10.1002/advs.202207488] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/07/2023] [Indexed: 05/03/2023]
Abstract
Cell-based therapies comprising the administration of living cells to patients for direct therapeutic activities have experienced remarkable success in the clinic, of which macrophages hold great potential for targeted drug delivery due to their inherent chemotactic mobility and homing ability to tumors with high efficiency. However, such targeted delivery of drugs through cellular systems remains a significant challenge due to the complexity of balancing high drug-loading with high accumulations in solid tumors. Herein, a tumor-targeting cellular drug delivery system (MAGN) by surface engineering of tumor-homing macrophages (Mφs) with biologically responsive nanosponges is reported. The pores of the nanosponges are blocked with iron-tannic acid complexes that serve as gatekeepers by holding encapsulated drugs until reaching the acidic tumor microenvironment. Molecular dynamics simulations and interfacial force studies are performed to provide mechanistic insights into the "ON-OFF" gating effect of the polyphenol-based supramolecular gatekeepers on the nanosponge channels. The cellular chemotaxis of the Mφ carriers enabled efficient tumor-targeted delivery of drugs and systemic suppression of tumor burden and lung metastases in vivo. The findings suggest that the MAGN platform offers a versatile strategy to efficiently load therapeutic drugs to treat advanced metastatic cancers with a high loading capacity of various therapeutic drugs.
Collapse
Affiliation(s)
- Xue Liao
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Guidong Gong
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
| | - Mengyuan Dai
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Zhenyu Xiang
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Jiezhou Pan
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Xianglian He
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
| | - Jiaojiao Shang
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
| | - Anna Maria Blocki
- School of Biomedical SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong SAR999077China
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of Illinois at ChicagoChicagoIL60612USA
| | - C. Wyatt Shields
- Department of Chemical and Biological EngineeringUniversity of ColoradoBoulderCO80303USA
| | - Junling Guo
- BMI Center for Biomass Materials and NanointerfacesCollege of Biomass Science and EngineeringSichuan UniversityChengduSichuan610065China
- National Engineering Laboratory for Clean Technology of Leather ManufactureSichuan UniversityChengduSichuan610065China
- Bioproducts InstituteDepartment of Chemical and Biological EngineeringUniversity of British ColumbiaVancouverBCV6T 1Z4Canada
- State Key Laboratory of Polymer Materials EngineeringSichuan UniversityChengduSichuan610065China
| |
Collapse
|
24
|
Ren Y, Yan C, Yang H. Erythrocytes: Member of the Immune System that Should Not Be Ignored. Crit Rev Oncol Hematol 2023; 187:104039. [PMID: 37236411 DOI: 10.1016/j.critrevonc.2023.104039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/27/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023] Open
Abstract
Erythrocytes are the most abundant type of cells in the blood and have a relatively simple structure when mature; they have a long life-span in the circulatory system. The primary function of erythrocytes is as oxygen carriers; however, they also play an important role in the immune system. Erythrocytes recognize and adhere to antigens and promote phagocytosis. The abnormal morphology and function of erythrocytes are also involved in the pathological processes of some diseases. Owing to the large number and immune properties of erythrocytes, their immune functions should not be ignored. Currently, research on immunity is focused on immune cells other than erythrocytes. However, research on the immune function of erythrocytes and the development of erythrocyte-mediated applications is of great significance. Therefore, we aimed to review the relevant literature and summarize the immune functions of erythrocytes.
Collapse
Affiliation(s)
- Yijun Ren
- Department of Neurology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China, 410000.
| | - Chengkai Yan
- Department of Neurology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China, 410000.
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, China, 410000.
| |
Collapse
|
25
|
Chao CJ, Zhang E, Zhao Z. Engineering cells for precision drug delivery: New advances, clinical translation, and emerging strategies. Adv Drug Deliv Rev 2023; 197:114840. [PMID: 37088403 DOI: 10.1016/j.addr.2023.114840] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Cells have emerged as a promising new form of drug delivery carriers owing to their distinguished advantages such as naturally bypassing immune recognition, intrinsic capability to navigate biological barriers, and access to hard-to-reach tissues via onboarding sensing and active motility. Over the past two decades, a large body of work has focused on understanding the ability of cell carriers to breach biological barriers and to modulate drug pharmacokinetics and pharmacodynamics. These efforts have led to the engineering of various cells for tissue-specific drug delivery. Despite exciting advances, clinical translation of cell-based drug carriers demands a thorough understanding of the pressing challenges and potential strategies to overcome them. Here, we summarize recent advances and new concepts in cell-based drug carriers and their clinical translation. We also discuss key considerations and emerging strategies to engineering the next-generation cell-based delivery technologies for more precise, targeted drug delivery.
Collapse
Affiliation(s)
- Chih-Jia Chao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Endong Zhang
- 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.
| |
Collapse
|
26
|
Albarqi HA, Garg A, Ahmad MZ, Alqahtani AA, Walbi IA, Ahmad J. Recent Progress in Chitosan-Based Nanomedicine for Its Ocular Application in Glaucoma. Pharmaceutics 2023; 15:pharmaceutics15020681. [PMID: 36840002 PMCID: PMC9963436 DOI: 10.3390/pharmaceutics15020681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Glaucoma is a degenerative, chronic ocular disease that causes irreversible vision loss. The major symptom of glaucoma is high intraocular pressure, which happens when the flow of aqueous humor between the front and back of the eye is blocked. Glaucoma therapy is challenging because of the low bioavailability of drugs from conventional ocular drug delivery systems such as eye drops, ointments, and gels. The low bioavailability of antiglaucoma agents could be due to the precorneal and corneal barriers as well as the low biopharmaceutical attributes of the drugs. These limitations can be overcome by employing nanoparticulate drug delivery systems. Over the last decade, there has been a lot of interest in chitosan-based nanoparticulate systems to overcome the limitations (such as poor residence time, low corneal permeability, etc.) associated with conventional ocular pharmaceutical products. Therefore, the main aim of the present manuscript is to review the recent research work involving the chitosan-based nanoparticulate system to treat glaucoma. It discusses the significance of the chitosan-based nanoparticulate system, which provides mucoadhesion to improve the residence time of drugs and their ocular bioavailability. Furthermore, different types of chitosan-based nanoparticulate systems are also discussed, namely nanoparticles of chitosan core only, nanoparticles coated with chitosan, and hybrid nanoparticles of chitosan. The manuscript also provides a critical analysis of contemporary research related to the impact of this chitosan-based nanomedicine on the corneal permeability, ocular bioavailability, and therapeutic performance of loaded antiglaucoma agents.
Collapse
Affiliation(s)
- Hassan A. Albarqi
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Anuj Garg
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Abdulsalam A. Alqahtani
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Ismail A. Walbi
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
- Correspondence: or
| |
Collapse
|
27
|
Lai WF, Zhang D, Wong WT. Design of erythrocyte-derived carriers for bioimaging applications. Trends Biotechnol 2023; 41:228-241. [PMID: 36031485 DOI: 10.1016/j.tibtech.2022.07.010] [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: 04/24/2022] [Revised: 07/01/2022] [Accepted: 07/25/2022] [Indexed: 01/24/2023]
Abstract
Erythrocytes are physiological entities that have been exploited in both preclinical and clinical trials for the delivery of exogenous agents. Over the years, diverse erythrocyte-derived carriers (ECs) have been developed with related patents granted for industrial and commercial purposes. However, most ECs have only been exploited for drug delivery. Serious discussions regarding their applications in imaging are scarce. This article reviews the role of ECs in enhancing imaging efficiency and subsequently delineates strategies for engineering and optimising their preclinical and clinical performance. With a snapshot of the latest developments and use of ECs in imaging, directions to streamline the clinical translation of related technologies can be attained for future research.
Collapse
Affiliation(s)
- Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China; Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang 310012, China.
| | - Dahong Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Zhejiang 310012, China
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
28
|
Choi A, Javius-Jones K, Hong S, Park H. Cell-Based Drug Delivery Systems with Innate Homing Capability as a Novel Nanocarrier Platform. Int J Nanomedicine 2023; 18:509-525. [PMID: 36742991 PMCID: PMC9893846 DOI: 10.2147/ijn.s394389] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023] Open
Abstract
Nanoparticle-based drug delivery systems have been designed to treat various diseases. However, many problems remain, such as inadequate tumor targeting and poor therapeutic outcomes. To overcome these obstacles, cell-based drug delivery systems have been developed. Candidates for cell-mediated drug delivery include blood cells, immune cells, and stem cells with innate tumor tropism and low immunogenicity; they act as a disguise to deliver the therapeutic payload. In drug delivery systems, therapeutic agents are encapsulated intracellularly or attached to the surface of the plasma membrane and transported to the desired site. Here, we review the pros and cons of cell-based therapies and discuss their homing mechanisms in the tumor microenvironment. In addition, different strategies to load therapeutic agents inside or on the surface of circulating cells and the current applications for a wide range of disease treatments are summarized.
Collapse
Affiliation(s)
- Anseo Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea,Correspondence: Hansoo Park; Seungpyo Hong, School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea, Tel +82-2 820 5804, Fax +82-2 813 8159, Email ;
| |
Collapse
|
29
|
Gong Y, Liu H, Ke S, Zhuo L, Wang H. Latest advances in biomimetic nanomaterials for diagnosis and treatment of cardiovascular disease. Front Cardiovasc Med 2023; 9:1037741. [PMID: 36684578 PMCID: PMC9846151 DOI: 10.3389/fcvm.2022.1037741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
Cardiovascular disease remains one of the leading causes of death in China, with increasingly serious negative effects on people and society. Despite significant advances in preventing and treating cardiovascular diseases, such as atrial fibrillation/flutter and heart failure over the last few years, much more remains to be done. Therefore, developing innovative methods for identifying and managing cardiovascular disorders is critical. Nanomaterials provide multiple benefits in biomedicine, primarily better catalytic activity, drug loading, targeting, and imaging. Biomimetic materials and nanoparticles are specially combined to synthesize biomimetic nanoparticles that successfully reduce the nanoparticles' toxicity and immunogenicity while enhancing histocompatibility. Additionally, the biological targeting capability of nanoparticles facilitates the diagnosis and therapy of cardiovascular disease. Nowadays, nanomedicine still faces numerous challenges, which necessitates creating nanoparticles that are highly selective, toxic-free, and better clinically applicable. This study reviews the scientific accomplishments in this field over the past few years covering the classification, applications, and prospects of noble metal biomimetic nanozymes and biomimetic nanocarriers.
Collapse
Affiliation(s)
- Yuxuan Gong
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China
| | - Huaying Liu
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China
| | - Shen Ke
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China
| | - Li Zhuo
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China,Li Zhuo,
| | - Haibin Wang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China,*Correspondence: Haibin Wang,
| |
Collapse
|
30
|
Wei W, Zhang Y, Lin Z, Wu X, Fan W, Chen J. Advances, challenge and prospects in cell-mediated nanodrug delivery for cancer therapy: a review. J Drug Target 2023; 31:1-13. [PMID: 35857432 DOI: 10.1080/1061186x.2022.2104299] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nanomedicine offers considerable opportunities to improve drugability and reduce toxicity for tumour therapy. However, the application of nanomedicine has achieved little success in clinical trials due to multiple physiological barriers to drug delivery. Circulating cells are expected to improve the physical distribution of drugs and enhance the therapeutic effect by overcoming various biological barriers in collaboration with nano-drug delivery systems owing to excellent biocompatibility, low immunogenicity and a long-circulation time and strong binding specificity. Nonetheless, we have noticed some limitations in implementing tthe strategy. In this article, we intend to introduce the latest progress in research and application of circulating cell-mediated nano-drug delivery systems, describe the main cell-related drug delivery modes, sum up the relevant points of the transport systems in the process of loading, transport and release, and lastly discuss the advantages, challenges and future development trends in cell-mediated nano-drug delivery.
Collapse
Affiliation(s)
- Wuhao Wei
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine Fuzhou, Fujian, China
| | | | | | - Xin Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine Fuzhou, Fujian, China.,Shanghai Wei Er Lab, Shanghai, China
| | - Wei Fan
- Seventh People's Hospital of Shanghai University of Traditional Chinese, Shanghai, China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine Fuzhou, Fujian, China
| |
Collapse
|
31
|
Pratiwi FW, Shanthi KB, Makieieva O, Chen ZA, Zhyvolozhnyi A, Miinalainen I, Bart G, Samoylenko A, Wu SH. Biogenesis of Mesoporous Silica Nanoparticles Enclosed in Extracellular Vesicles by Mouse Renal Adenocarcinoma Cells. Methods Mol Biol 2023; 2668:241-256. [PMID: 37140801 DOI: 10.1007/978-1-0716-3203-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Integrating the versatility of synthetic nanoparticles to natural biomaterials, such as cells or cell membranes, has gained considerable attention as promising alternative cargo delivery platforms in recent years. Extracellular vesicles (EVs), natural nanomaterials composed of a protein-rich lipid bilayer secreted by cells, have also shown advantages and great potential as a nano delivery platform in combination with synthetic particles due to their specific natural properties in overcoming several biology hurdles possessed in the recipient cell. Therefore, the preservation of EV's origin properties is critical for their application as nanocarriers. This chapter will describe the encapsulation procedure of MSN encapsulated in EV membrane derived from mouse renal adenocarcinoma (Renca) cells through biogenesis. The FMSN-enclosed EVs produced through this approach still contain preserved EV's natural membrane properties.
Collapse
Affiliation(s)
- Feby Wijaya Pratiwi
- Laboratory of Developmental Biology, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu and Kvantum Institute, Oulu, Finland.
| | - Keerthanaa Balasubramanian Shanthi
- Laboratory of Developmental Biology, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu and Kvantum Institute, Oulu, Finland
| | - Olha Makieieva
- Laboratory of Developmental Biology, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu and Kvantum Institute, Oulu, Finland
| | - Zih An Chen
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Artem Zhyvolozhnyi
- Laboratory of Developmental Biology, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu and Kvantum Institute, Oulu, Finland
| | - Ilkka Miinalainen
- Biocenter Oulu, Department of Pathology, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Genevieve Bart
- Laboratory of Developmental Biology, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu and Kvantum Institute, Oulu, Finland
| | - Anatoliy Samoylenko
- Laboratory of Developmental Biology, Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu and Kvantum Institute, Oulu, Finland
| | - Si-Han Wu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
32
|
Bai YT, Zhang XQ, Chen XJ, Zhou G. Nanomedicines in oral cancer: inspiration comes from extracellular vesicles and biomimetic nanoparticles. Nanomedicine (Lond) 2022; 17:1761-1778. [PMID: 36647844 DOI: 10.2217/nnm-2022-0142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Oral cancer is a common life-threatening malignancy having high mortality and morbidity rates. During the treatment process, individuals unavoidably experience severe side effects. It is essential to develop safer and more effective strategies. Currently, extracellular vesicles (EVs) and biomimetic nanoparticles are nanomedicines with long-term blood circulation and lower off-target toxicity that orchestrate immune responses and accumulate specifically in tumor sites. EVs create a synergetic effect by encapsulating drugs and collaborating with naturally loaded elements in the EVs. Biomimetic nanoparticles retain the characteristic features of the synthetic nanocarriers and inherit the intrinsic cell membrane functionalities. This review outlines the properties, applications, challenges, pros and cons of EVs and biomimetic nanoparticles, providing novel perspectives on oral cancer.
Collapse
Affiliation(s)
- Yu-Ting Bai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xue-Qiong Zhang
- School of Chemistry, Chemical Engineering & Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Xiao-Jie Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral Medicine, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Gang Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral Medicine, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
33
|
Li D. Role of Spectrin in Endocytosis. Cells 2022; 11:cells11152459. [PMID: 35954302 PMCID: PMC9368487 DOI: 10.3390/cells11152459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Cytoskeletal spectrin is found in (non)erythroid cells. Eukaryotic endocytosis takes place for internalizing cargos from extracellular milieu. The role of spectrin in endocytosis still remains poorly understood. Here, I summarize current knowledge of spectrin function, spectrin-based cytoskeleton and endocytosis of erythrocytes, and highlight how spectrin contributes to endocytosis and working models in different types of cells. From an evolutionary viewpoint, I discuss spectrin and endocytosis in a range of organisms, particularly in plants and yeast where spectrin is absent. Together, the role of spectrin in endocytosis is related to its post-translational modification, movement/rearrangement, elimination (by proteases) and meshwork fencing.
Collapse
Affiliation(s)
- Donghai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
34
|
Ren Z, Liao T, Li C, Kuang Y. Drug Delivery Systems with a "Tumor-Triggered" Targeting or Intracellular Drug Release Property Based on DePEGylation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5290. [PMID: 35955225 PMCID: PMC9369796 DOI: 10.3390/ma15155290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022]
Abstract
Coating nanosized anticancer drug delivery systems (DDSs) with poly(ethylene glycol) (PEG), the so-called PEGylation, has been proven an effective method to enhance hydrophilicity, aqueous dispersivity, and stability of DDSs. What is more, as PEG has the lowest level of protein absorption of any known polymer, PEGylation can reduce the clearance of DDSs by the mononuclear phagocyte system (MPS) and prolong their blood circulation time in vivo. However, the "stealthy" characteristic of PEG also diminishes the uptake of DDSs by cancer cells, which may reduce drug utilization. Therefore, dynamic protection strategies have been widely researched in the past years. Coating DDSs with PEG through dynamic covalent or noncovalent bonds that are stable in blood and normal tissues, but can be broken in the tumor microenvironment (TME), can achieve a DePEGylation-based "tumor-triggered" targeting or intracellular drug release, which can effectively improve the utilization of drugs and reduce their side effects. In this review, the stimuli and methods of "tumor-triggered" targeting or intracellular drug release, based on DePEGylation, are summarized. Additionally, the targeting and intracellular controlled release behaviors of the DDSs are briefly introduced.
Collapse
Affiliation(s)
- Zhe Ren
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; (Z.R.); (T.L.)
| | - Tao Liao
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; (Z.R.); (T.L.)
| | - Cao Li
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; (Z.R.); (T.L.)
| | - Ying Kuang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
35
|
Rong R, Raza F, Liu Y, Yuan WE, Su J, Qiu M. Blood cell-based drug delivery systems: a biomimetic platform for antibacterial therapy. Eur J Pharm Biopharm 2022; 177:273-288. [PMID: 35868489 DOI: 10.1016/j.ejpb.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
With the rapid increase in multidrug-resistance against antibiotics, higher doses of antibiotics or more effective antibiotics are needed to treat diseases, which ultimately leads to a decrease in the body's immunity and seriously threatens human health worldwide. The efficiency of antibiotics has been a large challenge for years. To overcome this problem, many carriers are utilized for anti-bacteria, attempting to optimize the delivery of such drugs and transport them safely and directly to the site of disease. Blood cell-based drug delivery systems present several advantages as compared to polymeric delivery system. These blood cells including red blood cells (RBCs), leukocytes, platelets. The blood cells and their membranes can both be used as drug carriers to deliver antibacterial drugs. In addition, blood cells can overcome many physiological/pathological obstacles faced by nanoparticles in vivo and effectively deliver drugs to the site of the disease. In this paper, we review studies on blood cell-based delivery systems used in antibacterial therapy, and analyze different roles in antibacterial therapy, which provide basis for further study in this field.
Collapse
Affiliation(s)
- Ruonan Rong
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Yuhao Liu
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Wei-En Yuan
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China.
| |
Collapse
|
36
|
Hou S, Hasnat M, Chen Z, Liu Y, Faran Ashraf Baig MM, Liu F, Chen Z. Application Perspectives of Nanomedicine in Cancer Treatment. Front Pharmacol 2022; 13:909526. [PMID: 35860027 PMCID: PMC9291274 DOI: 10.3389/fphar.2022.909526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is a disease that seriously threatens human health. Based on the improvement of traditional treatment methods and the development of new treatment modes, the pattern of cancer treatment is constantly being optimized. Nanomedicine plays an important role in these evolving tumor treatment modalities. In this article, we outline the applications of nanomedicine in three important tumor-related fields: chemotherapy, gene therapy, and immunotherapy. According to the current common problems, such as poor targeting of first-line chemotherapy drugs, easy destruction of nucleic acid drugs, and common immune-related adverse events in immunotherapy, we discuss how nanomedicine can be combined with these treatment modalities, provide typical examples, and summarize the advantages brought by the application of nanomedicine.
Collapse
Affiliation(s)
- Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Muhammad Hasnat
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Ziwei Chen
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Yinong Liu
- Hospital Laboratory of Nangjing Lishui People’s Hospital, Nangjing, China
| | - Mirza Muhammad Faran Ashraf Baig
- Laboratory of Biomedical Engineering for Novel Bio-functional, and Pharmaceutical Nanomaterials, Prince Philip Dental Hospital, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fuhe Liu
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
- *Correspondence: Zelong Chen, ; Fuhe Liu,
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Province Engineering Research Center of Artificial Intelligence and Internet of Things Wise Medical, Zhengzhou, China
- *Correspondence: Zelong Chen, ; Fuhe Liu,
| |
Collapse
|
37
|
Yan H, Zhai B, Yang F, Chen Z, Zhou Q, Paiva-Santos AC, Yuan Z, Zhou Y. Nanotechnology-Based Diagnostic and Therapeutic Strategies for Neuroblastoma. Front Pharmacol 2022; 13:908713. [PMID: 35721107 PMCID: PMC9201105 DOI: 10.3389/fphar.2022.908713] [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: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Neuroblastoma (NB), as the most common extracranial solid tumor in childhood, is one of the critical culprits affecting children's health. Given the heterogeneity and invisibility of NB tumors, the existing diagnostic and therapeutic approaches are inadequate and ineffective in early screening and prognostic improvement. With the rapid innovation and development of nanotechnology, nanomedicines have attracted widespread attention in the field of oncology research for their excellent physiological and chemical properties. In this review, we first explored the current common obstacles in the diagnosis and treatment of NB. Then we comprehensively summarized the advancements in nanotechnology-based multimodal synergistic diagnosis and treatment of NB and elucidate the underlying mechanisms. In addition, a discussion of the pending challenges in biocompatibility and toxicity of nanomedicine was conducted. Finally, we described the development and application status of nanomaterials against some of the recognized targets in the field of NB research, and pointed out prospects for nanomedicine-based precision diagnosis and therapy of NB.
Collapse
Affiliation(s)
- Hui Yan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China.,Department of Cardiothoracic Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Bo Zhai
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China.,Department of Cardiothoracic Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Fang Yang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China.,Department of Cardiothoracic Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Zhenliang Chen
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China.,Department of Cardiothoracic Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Qiang Zhou
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China.,Department of Pathology, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Ana Cláudia Paiva-Santos
- Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ziqiao Yuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yang Zhou
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou, China.,Department of Cardiothoracic Surgery, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| |
Collapse
|
38
|
Xu X, Li T, Jin K. Bioinspired and Biomimetic Nanomedicines for Targeted Cancer Therapy. Pharmaceutics 2022; 14:1109. [PMID: 35631695 PMCID: PMC9147382 DOI: 10.3390/pharmaceutics14051109] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Undesirable side effects and multidrug resistance are the major obstacles in conventional chemotherapy towards cancers. Nanomedicines provide alternative strategies for tumor-targeted therapy due to their inherent properties, such as nanoscale size and tunable surface features. However, the applications of nanomedicines are hampered in vivo due to intrinsic disadvantages, such as poor abilities to cross biological barriers and unexpected off-target effects. Fortunately, biomimetic nanomedicines are emerging as promising therapeutics to maximize anti-tumor efficacy with minimal adverse effects due to their good biocompatibility and high accumulation abilities. These bioengineered agents incorporate both the physicochemical properties of diverse functional materials and the advantages of biological materials to achieve desired purposes, such as prolonged circulation time, specific targeting of tumor cells, and immune modulation. Among biological materials, mammalian cells (such as red blood cells, macrophages, monocytes, and neutrophils) and pathogens (such as viruses, bacteria, and fungi) are the functional components most often used to confer synthetic nanoparticles with the complex functionalities necessary for effective nano-biointeractions. In this review, we focus on recent advances in the development of bioinspired and biomimetic nanomedicines (such as mammalian cell-based drug delivery systems and pathogen-based nanoparticles) for targeted cancer therapy. We also discuss the biological influences and limitations of synthetic materials on the therapeutic effects and targeted efficacies of various nanomedicines.
Collapse
Affiliation(s)
- Xiaoqiu Xu
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|