51
|
Cell Membrane-Cloaked Nanotherapeutics for Targeted Drug Delivery. Int J Mol Sci 2022; 23:ijms23042223. [PMID: 35216342 PMCID: PMC8879543 DOI: 10.3390/ijms23042223] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
Cell membrane cloaking technique is bioinspired nanotechnology that takes advantage of naturally derived design cues for surface modification of nanoparticles. Unlike modification with synthetic materials, cell membranes can replicate complex physicochemical properties and biomimetic functions of the parent cell source. This technique indeed has the potential to greatly augment existing nanotherapeutic platforms. Here, we provide a comprehensive overview of engineered cell membrane-based nanotherapeutics for targeted drug delivery and biomedical applications and discuss the challenges and opportunities of cell membrane cloaking techniques for clinical translation.
Collapse
|
52
|
Aghajanzadeh M, Zamani M, Rajabi Kouchi F, Eixenberger J, Shirini D, Estrada D, Shirini F. Synergic Antitumor Effect of Photodynamic Therapy and Chemotherapy Mediated by Nano Drug Delivery Systems. Pharmaceutics 2022; 14:pharmaceutics14020322. [PMID: 35214054 PMCID: PMC8880656 DOI: 10.3390/pharmaceutics14020322] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
This review provides a summary of recent progress in the development of different nano-platforms for the efficient synergistic effect between photodynamic therapy and chemotherapy. In particular, this review focuses on various methods in which photosensitizers and chemotherapeutic agents are co-delivered to the targeted tumor site. In many cases, the photosensitizers act as drug carriers, but this review, also covers different types of appropriate nanocarriers that aid in the delivery of photosensitizers to the tumor site. These nanocarriers include transition metal, silica and graphene-based materials, liposomes, dendrimers, polymers, metal–organic frameworks, nano emulsions, and biologically derived nanocarriers. Many studies have demonstrated various benefits from using these nanocarriers including enhanced water solubility, stability, longer circulation times, and higher accumulation of therapeutic agents/photosensitizers at tumor sites. This review also describes novel approaches from different research groups that utilize various targeting strategies to increase treatment efficacy through simultaneous photodynamic therapy and chemotherapy.
Collapse
Affiliation(s)
- Mozhgan Aghajanzadeh
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
| | - Mostafa Zamani
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
| | - Fereshteh Rajabi Kouchi
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
| | - Josh Eixenberger
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
- Center for Advanced Energy Studies, Boise State University, Boise, ID 83725, USA
- Correspondence: (J.E.); or (F.S.)
| | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
| | - David Estrada
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
- Center for Advanced Energy Studies, Boise State University, Boise, ID 83725, USA
| | - Farhad Shirini
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
- Correspondence: (J.E.); or (F.S.)
| |
Collapse
|
53
|
Hou K, Zhang Y, Bao M, Xin C, Wei Z, Lin G, Wang Z. A Multifunctional Magnetic Red Blood Cell-Mimetic Micromotor for Drug Delivery and Image-Guided Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3825-3837. [PMID: 35025195 DOI: 10.1021/acsami.1c21331] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Inspired by nature, innovative devices have been made to imitate the morphology and functions of natural red blood cells (RBCs). Here, we report a red blood cell-mimetic micromotor (RBCM), which was fabricated based on a layer-by-layer assembly method and precisely controlled by an external rotating uniform magnetic field. The main framework of the RBCM was constructed by the natural protein zein and finally camouflaged with the RBC membrane. Functional cargos such as Fe3O4 nanoparticles and the chemotherapeutic agent doxorubicin were loaded within the wall part of the RBCM for tumor therapy. Due to the massive loading of Fe3O4 nanoparticles, the RBCM can be precisely navigated by an external rotating uniform magnetic field and be used as a magnetic resonance imaging contrast agent for tumor imaging. The RBCM has been proven to be biocompatible, biodegradable, magnetically manipulated, and imageable, which are key requisites to take micromotors from the chalkboard to clinics. We expect the RBC-inspired biohybrid device to achieve wide potential applications.
Collapse
Affiliation(s)
- Kexin Hou
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 150001 Harbin, China
| | - Yandong Zhang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 150001 Harbin, China
| | - Meili Bao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 150001 Harbin, China
| | - Chao Xin
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 150001 Harbin, China
| | - Zengyan Wei
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 150001 Harbin, China
| | - Guochang Lin
- School of Astronautics, Harbin Institute of Technology, 150001 Harbin, China
| | - Zhenyu Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, 150001 Harbin, China
| |
Collapse
|
54
|
Lei W, Yang C, Wu Y, Ru G, He X, Tong X, Wang S. Nanocarriers surface engineered with cell membranes for cancer targeted chemotherapy. J Nanobiotechnology 2022; 20:45. [PMID: 35062958 PMCID: PMC8781141 DOI: 10.1186/s12951-022-01251-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Inspired by nature, the biomimetic approach has been incorporated into drug nanocarriers for cancer targeted chemotherapy. The nanocarriers are cloaked in cell membranes, which enables them to incorporate the functions of natural cells.
Key scientific concepts of review
Nanocarriers surface engineered with cell membranes have emerged as a fascinating source of materials for cancer targeted chemotherapy. A distinctive characteristic of cell membrane-coated nanocarriers (CMCNs) is that they include carbohydrates, proteins, and lipids, in addition to being biocompatible. CMCNs are capable of interacting with the complicated biological milieu of the tumor because they contain the signaling networks and intrinsic functions of their parent cells. Numerous cell membranes have been investigated for the purpose of masking nanocarriers with membranes, and various tumor-targeting methods have been devised to improve cancer targeted chemotherapy. Moreover, the diverse structure of the membrane from different cell sources broadens the spectrum of CMCNs and offers an entirely new class of drug-delivery systems.
Aim of review
This review will describe the manufacturing processes for CMCNs and the therapeutic uses for different kinds of cell membrane-coated nanocarrier-based drug delivery systems, as well as addressing obstacles and future prospects.
Graphical Abstract
Collapse
|
55
|
|
56
|
Ren X, Yuan W, Ma J, Wang P, Sun S, Wang S, Zhao R, Liang X. Magnetic nanoclusters mediated photothermal effect and macrophage modulation for synergistically photothermal immunotherapy of cancer. Biomater Sci 2022; 10:3188-3200. [PMID: 35579248 DOI: 10.1039/d1bm01770e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In tumor microenvironment, macrophages predominately exhibit M2-type functionalities which promote malignant progression and cancer metastasis, thus bring big hurdle to current anticancer strategies. Different approaches had been exploited to reverse...
Collapse
Affiliation(s)
- Xiaoqing Ren
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
| | - Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Jing Ma
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Ping Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Shumin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Rongsheng Zhao
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
57
|
Safarpour F, Kharaziha M, Emadi R. Inspiring biomimetic system based on red blood cell membrane vesicles for effective curcumin loading and release. Int J Pharm 2021; 613:121419. [PMID: 34954002 DOI: 10.1016/j.ijpharm.2021.121419] [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: 10/26/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/17/2022]
Abstract
The aim of this study is to introduce an inspiring biomimetic system based on the red blood cell membrane (RBCM) vesicles for improved encapsulation efficiency and release of curcumin (Cur). Here, the role of the sonication time (0.5, 1.5, 3 and 5 min) on the properties of RBCM-CUR vesicles is investigated. It is determined that the hydrodynamic vesicle size, zeta potential, and release behavior are tunable by changing the sonication time. Noticeably, the average size of vesicles decreased from 163.0 ± 21 nm to 116.3 ± 16 nm by increasing the sonication time from 0.5 to 5 min. Moreover, the drug release value, after 24 h incubation, enhances from 57 to 99% with the expansion of sonication from 0.5 to 5 min. Additionally, the entrapment efficiency of Cur as a model drug is high in whole sonication time, owing to the amphiphilic nature of RBCM. Finally, the RBCM-CUR vesicles are not only cytocompatible, but also could support the attachment and proliferation of fibroblast cells in vitro. The RBCM based system for delivery of Cur could be a promising system for the wound healing applications.
Collapse
Affiliation(s)
- F Safarpour
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - M Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - R Emadi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
58
|
Yu X, Sha L, Liu Q, Zhao Y, Fang H, Cao Y, Zhao J. Recent advances in cell membrane camouflage-based biosensing application. Biosens Bioelectron 2021; 194:113623. [PMID: 34530371 DOI: 10.1016/j.bios.2021.113623] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 01/04/2023]
Abstract
Cell membrane, a semi-permeable membrane composed of phospholipid bilayers, is a natural barrier to prevent extracellular substances from freely entering the cell. Cell membrane with selective permeability and fluidity ensures the relative stability of the intracellular environment and enables various biochemical reactions to smoothly operate in an orderly manner. Inspired by the natural composition and transport process, various cell membranes and synthetic bionic films as the mimics of cell membranes have emerged as appealing camouflage materials for biosensing applications. The membranes are devoted to surface modification and substance delivery, and realize the detection or in situ analysis of multiple biomarkers, such as glucose, nucleic acids, virus, and circulating tumor cells. In this review, we summarize the recent advances in cell membrane camouflage-based biosensing applications, mainly focusing on the use of the membranes extracted from natural cells (e.g., blood cells and cancer cells) as well as biomimetic membranes. Materials and surfaces camouflaged with cell membranes are shown to have superior stability and biocompatibility as well as intrinsic properties of original cells, which greatly facilitate their use in biosensing. In specific, camouflage with blood cell membranes bestows low immunogenicity and prolonged blood circulation time, camouflage with cancer cell membranes provides homologous targeting ability, and camouflage with biomimetic membranes endows considerable plasticity for functionalization. Further research is expected to focus on the deeper understanding of cell-specific properties of membranes and the exploration of hybrid membranes, which might provide new development opportunities for cell membrane camouflage-based biosensing application.
Collapse
Affiliation(s)
- Xiaomeng Yu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, PR China; Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Lingjun Sha
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Qi Liu
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Yingyan Zhao
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Huan Fang
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Ya Cao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, PR China; Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Jing Zhao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, PR China; Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
59
|
Wang S, Shen H, Mao Q, Tao Q, Yuan G, Zeng L, Chen Z, Zhang Y, Cheng L, Zhang J, Dai H, Hu C, Pan Y, Li Y. Macrophage-Mediated Porous Magnetic Nanoparticles for Multimodal Imaging and Postoperative Photothermal Therapy of Gliomas. ACS APPLIED MATERIALS & INTERFACES 2021; 13:56825-56837. [PMID: 34825820 DOI: 10.1021/acsami.1c12406] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Because of the blood-brain barrier and the high infiltration of glioma cells, the diagnostic accuracy and treatment efficiency of gliomas are still facing challenges. There is an urgent need to explore the integration of diagnostic and therapeutic methods to achieve an accurate diagnosis, guide surgery, and inhibit postoperative recurrence. In this work, we developed a macrophage loaded with a photothermal nanoprobe (MFe3O4-Cy5.5), which is able to cross the blood-brain barrier and accumulate into deep gliomas to achieve multimodal imaging and guided glioma surgery purposes. With desirable probing depth and high signal-to-noise ratio, Fe3O4-Cy5.5 can perform fluorescence, photoacoustic, and magnetic resonance imaging, which can distinguish brain tumors from the surrounding normal tissues and accurately guide glioma resection. Meanwhile, Fe3O4-Cy5.5 can effectively induce local photothermal therapy and inhibit the recurrence of glioma after surgery. These results demonstrate that the macrophage-mediated Fe3O4-Cy5.5, which can achieve a multimodal diagnosis, accurate imaging-guided surgery, and effective photothermal therapy, is a promising nanoplatform for gliomas.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Hailin Shen
- Department of Radiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, Jiangsu, China
| | - Qiulian Mao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qing Tao
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Guotao Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Lingli Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ziying Chen
- Nanobio Laboratory, Institute of Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Yunjiao Zhang
- Nanobio Laboratory, Institute of Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jingzhong Zhang
- The Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Hui Dai
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yonggang Li
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
- Institute of Medical Imaging, Soochow University, Suzhou 215000, Jiangsu, China
| |
Collapse
|
60
|
Guo K, Xiao N, Liu Y, Wang Z, Tóth J, Gyenis J, Thakur VK, Oyane A, Shubhra QT. Engineering polymer nanoparticles using cell membrane coating technology and their application in cancer treatments: Opportunities and challenges. NANO MATERIALS SCIENCE 2021. [DOI: 10.1016/j.nanoms.2021.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
61
|
Chugh V, Vijaya Krishna K, Pandit A. Cell Membrane-Coated Mimics: A Methodological Approach for Fabrication, Characterization for Therapeutic Applications, and Challenges for Clinical Translation. ACS NANO 2021; 15:17080-17123. [PMID: 34699181 PMCID: PMC8613911 DOI: 10.1021/acsnano.1c03800] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/13/2021] [Indexed: 05/04/2023]
Abstract
Cell membrane-coated (CMC) mimics are micro/nanosystems that combine an isolated cell membrane and a template of choice to mimic the functions of a cell. The design exploits its physicochemical and biological properties for therapeutic applications. The mimics demonstrate excellent biological compatibility, enhanced biointerfacing capabilities, physical, chemical, and biological tunability, ability to retain cellular properties, immune escape, prolonged circulation time, and protect the encapsulated drug from degradation and active targeting. These properties and the ease of adapting them for personalized clinical medicine have generated a significant research interest over the past decade. This review presents a detailed overview of the recent advances in the development of cell membrane-coated (CMC) mimics. The primary focus is to collate and discuss components, fabrication methodologies, and the significance of physiochemical and biological characterization techniques for validating a CMC mimic. We present a critical analysis of the two main components of CMC mimics: the template and the cell membrane and mapped their use in therapeutic scenarios. In addition, we have emphasized on the challenges associated with CMC mimics in their clinical translation. Overall, this review is an up to date toolbox that researchers can benefit from while designing and characterizing CMC mimics.
Collapse
Affiliation(s)
| | | | - Abhay Pandit
- CÚRAM, SFI Research
Centre for Medical Devices, National University
of Ireland Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
62
|
Fibroblast membrane-camouflaged nanoparticles for inflammation treatment in the early stage. Int J Oral Sci 2021; 13:39. [PMID: 34785637 PMCID: PMC8595357 DOI: 10.1038/s41368-021-00144-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 02/05/2023] Open
Abstract
Unrestrained inflammation is harmful to tissue repair and regeneration. Immune cell membrane-camouflaged nanoparticles have been proven to show promise as inflammation targets and multitargeted inflammation controls in the treatment of severe inflammation. Prevention and early intervention of inflammation can reduce the risk of irreversible tissue damage and loss of function, but no cell membrane-camouflaged nanotechnology has been reported to achieve stage-specific treatment in these conditions. In this study, we investigated the prophylactic and therapeutic efficacy of fibroblast membrane-camouflaged nanoparticles for topical treatment of early inflammation (early pulpitis as the model) with the help of in-depth bioinformatics and molecular biology investigations in vitro and in vivo. Nanoparticles have been proven to act as sentinels to detect and competitively neutralize invasive Escherichia coli lipopolysaccharide (E. coli LPS) with resident fibroblasts to effectively inhibit the activation of intricate signaling pathways. Moreover, nanoparticles can alleviate the secretion of multiple inflammatory cytokines to achieve multitargeted anti-inflammatory effects, attenuating inflammatory conditions in the early stage. Our work verified the feasibility of fibroblast membrane-camouflaged nanoparticles for inflammation treatment in the early stage, which widens the potential cell types for inflammation regulation.
Collapse
|
63
|
Wang Y, Xu X, Chen X, Li J. Multifunctional Biomedical Materials Derived from Biological Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 34:e2107406. [PMID: 34739155 DOI: 10.1002/adma.202107406] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/24/2021] [Indexed: 02/06/2023]
Abstract
The delicate structure and fantastic functions of biological membranes are the successful evolutionary results of a long-term natural selection process. Their excellent biocompatibility and biofunctionality are widely utilized to construct multifunctional biomedical materials mainly by directly camouflaging materials with single or mixed biological membranes, decorating or incorporating materials with membrane-derived vesicles (e.g., exosomes), and designing multifunctional materials with the structure/functions of biological membranes. Here, the structure-function relationship of some important biological membranes and biomimetic membranes are discussed, such as various cell membranes, extracellular vesicles, and membranes from bacteria and organelles. Selected literature examples of multifunctional biomaterials derived from biological membranes for biomedical applications, such as drug- and gene-delivery systems, tissue-repair scaffolds, bioimaging, biosensors, and biological detection, are also highlighted. These designed materials show excellent properties, such as long circulation time, disease-targeted therapy, excellent biocompatibility, and selective recognition. Finally, perspectives and challenges associated with the clinical applications of biological-membrane-derived materials are discussed.
Collapse
Affiliation(s)
- Yuemin Wang
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| | - Xinyuan Xu
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| | - Xingyu Chen
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
- College of Medicine Southwest Jiaotong University Chengdu 610003 China
| | - Jianshu Li
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology Med‐X Center for Materials Sichuan University Chengdu 610041 China
| |
Collapse
|
64
|
Chen H, Zhang H, Xu T, Yu J. An Overview of Micronanoswarms for Biomedical Applications. ACS NANO 2021; 15:15625-15644. [PMID: 34647455 DOI: 10.1021/acsnano.1c07363] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Micronanoswarms have attracted extensive attention worldwide due to their great promise in biomedical applications. The collective behaviors among thousands, or even millions, of tiny active agents indicate immense potential for benefiting the progress of clinical therapeutic and diagnostic methods. In recent years, with the development of smart materials, remote actuation modalities, and automatic control strategies, the motion dexterity, environmental adaptability, and functionality versatility of micronanoswarms are improved. Swarms can thus be designed as dexterous platforms inside living bodies to perform a multitude of tasks related to healthcare. Existing surveys summarize the design, functionalization, and biomedical applications of micronanorobots and the actuation and motion control strategies of micronanoswarms. This review presents the recent progress of micronanoswarms, aiming for biomedical applications. The recent advances on structural design of artificial, living, and hybrid micronanoswarms are summarized, and the biomedical applications that could be tackled using micronanoswarms are introduced, such as targeted drug delivery, hyperthermia, imaging and sensing, and thrombolysis. Moreover, potential challenges and promising trends of future developments are discussed. It is envisioned that the future success of these promising tools will have a significant impact on clinical treatment.
Collapse
Affiliation(s)
- Hui Chen
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
| | - Huimin Zhang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Tiantian Xu
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- Guangdong Provincial Key Laboratory of Robotics and Intelligent System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518126, China
| | - Jiangfan Yu
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
| |
Collapse
|
65
|
Kawassaki RK, Romano M, Dietrich N, Araki K. Titanium and Iron Oxide Nanoparticles for Cancer Therapy: Surface Chemistry and Biological Implications. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.735434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Currently, cancer is among the most challenging diseases due to its ability to continuously evolve into a more complex muldimentional system, in addition to its high capability to spread to other organs and tissues. In this context, the relevance of nanobiomaterials (NBMs) for the development of new more effective and less harmful treatments is increasing. NBMs provide the possibility of combining several functionalities on a single system, expectedly in a synergic way, to better perform the treatment and cure. However, the control of properties such as colloidal stability, circulation time, pharmacokinetics, and biodistribution, assuring the concentration in specific target tissues and organs, while keeping all desired properties, tends to be dependent on subtle changes in surface chemistry. Hence, the behavior of such materials in different media/environments is of uttermost relevance and concern since it can compromise their efficiency and safety on application. Given the bright perspectives, many efforts have been focused on the development of nanomaterials fulfilling the requirements for real application. These include robust and reproducible preparation methods to avoid aggregation while preserving the interaction properties. The possible impact of nanomaterials in different forms of diagnosis and therapy has been demonstrated in the past few years, given the perspectives on how revolutionary they can be in medicine and health. Considering the high biocompatibility and suitability, this review is focused on titanium dioxide– and iron oxide–based nanoagents highlighting the current trends and main advancements in the research for cancer therapies. The effects of phenomena, such as aggregation and agglomeration, the formation of the corona layer, and how they can compromise relevant properties of nanomaterials and their potential applicability, are also addressed. In short, this review summarizes the current understanding and perspectives on such smart nanobiomaterials for diagnostics, treatment, and theranostics of diseases.
Collapse
|
66
|
Deng L, Zhang H, Zhang Y, Luo S, Du Z, Lin Q, Zhang Z, Zhang L. An exosome-mimicking membrane hybrid nanoplatform for targeted treatment toward Kras-mutant pancreatic carcinoma. Biomater Sci 2021; 9:5599-5611. [PMID: 34250995 DOI: 10.1039/d1bm00446h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pancreatic carcinoma elevates quickly and thus has a high mortality rate. Therefore, early treatment is essential for treating pancreatic carcinoma. KRAS is the most frequently identified and one of the earliest mutations in pancreatic tumorigenesis. Thus, the KRAS-mutant cell is an ideal target for the treatment of pancreatic carcinoma, especially at the early stage. KRAS mutation increases macropinocytosis in pancreatic cancer cells, enhancing the internalization of exosomes. Because acquiring natural exosomes could be laborious and their encapsulation efficiency is often unsatisfactory, we aimed to develop a delivery system that mimics the Kras-mutant cell targeting capability of exosomes but is easier to generate and has better loading efficiency. For this purpose, we constructed a hybrid nanoplatform by fusing CLT (Celastrol)-Loaded PEGylated lipids with the DC2.4 cell membrane (M-LIP-CLT) to achieve targeted treatment of Kras-mutant pancreatic cancer. This hybrid nanoplatform improved CLT tumor accumulation and showed excellent anti-cancer efficiency both in vitro and in vivo with increased safety. These results suggest that M-LIP-CLT is an effective drug delivery system for targeted therapy against pancreatic carcinoma, and the fusion strategy showed attractive potential for further development.
Collapse
Affiliation(s)
- Lang Deng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Hanming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Yu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Shi Luo
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Zhengwu Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610041, P. R. China.
| |
Collapse
|
67
|
Zhang Y, Xia Q, Wu T, He Z, Li Y, Li Z, Hou X, He Y, Ruan S, Wang Z, Sun J, Feng N. A novel multi-functionalized multicellular nanodelivery system for non-small cell lung cancer photochemotherapy. J Nanobiotechnology 2021; 19:245. [PMID: 34391438 PMCID: PMC8364713 DOI: 10.1186/s12951-021-00977-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A red blood cell membrane (RBCm)-derived drug delivery system allows prolonged circulation of an antitumor treatment and overcomes the issue of accelerated blood clearance induced by PEGylation. However, RBCm-derived drug delivery systems are limited by low drug-loading capacities and the lack of tumor-targeting ability. Thus, new designs of RBCm-based delivery systems are needed. RESULTS Herein, we designed hyaluronic acid (HA)-hybridized RBCm (HA&RBCm)-coated lipid multichambered nanoparticles (HA&RBCm-LCNPs) to remedy the limitations of traditional RBCm drug delivery systems. The inner core co-assembled with phospholipid-regulated glycerol dioleate/water system in HA&RBCm-LCNPs met the required level of blood compatibility for intravenous administration. These newly designed nanocarriers had a honeycomb structure with abundant spaces that efficiently encapsulated paclitaxel and IR780 for photochemotherapy. The HA&RBCm coating allowed the nanocarriers to overcome the reticuloendothelial system barrier and enhanced the nanocarriers specificity to A549 cells with high levels of CD44. These properties enhanced the combinatorial antitumor effects of paclitaxel and IR780 associated with microtubule destruction and the mitochondrial apoptotic pathway. CONCLUSIONS The multifunctional HA&RBCm-LCNPs we designed expanded the functionality of RBCm and resulted in a vehicle for safe and efficient antitumor treatment.
Collapse
Affiliation(s)
- Yongtai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qing Xia
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Wu
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zehui He
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yanyan Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhe Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xuefeng Hou
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuanzhi He
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shuyao Ruan
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhi Wang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia Sun
- Teaching Experiment Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
68
|
Le QV, Lee J, Lee H, Shim G, Oh YK. Cell membrane-derived vesicles for delivery of therapeutic agents. Acta Pharm Sin B 2021; 11:2096-2113. [PMID: 34522579 PMCID: PMC8424219 DOI: 10.1016/j.apsb.2021.01.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/02/2021] [Accepted: 01/14/2021] [Indexed: 12/18/2022] Open
Abstract
Cell membranes have recently emerged as a new source of materials for molecular delivery systems. Cell membranes have been extruded or sonicated to make nanoscale vesicles. Unlike synthetic lipid or polymeric nanoparticles, cell membrane-derived vesicles have a unique multicomponent feature, comprising lipids, proteins, and carbohydrates. Because cell membrane-derived vesicles contain the intrinsic functionalities and signaling networks of their parent cells, they can overcome various obstacles encountered in vivo. Moreover, the different natural combinations of membranes from various cell sources expand the range of cell membrane-derived vesicles, creating an entirely new category of drug-delivery systems. Cell membrane-derived vesicles can carry therapeutic agents within their interior or can coat the surfaces of drug-loaded core nanoparticles. Cell membranes typically come from single cell sources, including red blood cells, platelets, immune cells, stem cells, and cancer cells. However, recent studies have reported hybrid sources from two different types of cells. This review will summarize approaches for manufacturing cell membrane-derived vesicles and treatment applications of various types of cell membrane-derived drug-delivery systems, and discuss challenges and future directions.
Collapse
Key Words
- Blood cells
- CAR-T, chimeric antigen receptor-engineered T cell
- CRISPR, clustered regularly interspaced short palindromic repeats
- CXCR4, C-X-C chemokine receptor type 4
- Cancer cells
- Cell membrane-derived vesicles
- DC, dendritic cell
- Drug-delivery systems
- Immune cells
- Manufacturing
- Membrane engineering
- NF-κB, nuclear factor kappa B
- NIR, near infrared
- PEG, polyethylene glycol
- PLGA, poly(lactic-co-glycolic acid)
- RBC, red blood cell
- Stem cells
- TCR, T-cell receptor
- TRAIL, tumor necrosis factor-related apoptosis-inducing ligand
Collapse
Affiliation(s)
- Quoc-Viet Le
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hobin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
69
|
Liu X, Zhong X, Li C. Challenges in cell membrane-camouflaged drug delivery systems: Development strategies and future prospects. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.03.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
70
|
Zhang R, Wu S, Ding Q, Fan Q, Dai Y, Guo S, Ye Y, Li C, Zhou M. Recent advances in cell membrane-camouflaged nanoparticles for inflammation therapy. Drug Deliv 2021; 28:1109-1119. [PMID: 34121563 PMCID: PMC8205088 DOI: 10.1080/10717544.2021.1934188] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
During inflammation, inflammatory cells are rapidly recruited to sites of infection or injury, where they cross physiological barriers around the infected site and further infiltrate into the tissues. Other cells, such as erythrocytes, endothelial cells and stem cells, also play prominent roles in host defense and tissue repair. In recent years, nanotechnology has been exploited to deliver drugs to sites of inflammation. For example, nanoparticles camouflaged with a cell membrane are a novel drug-delivery platform that can interact with the immune system and that show great potential for treating inflammation. Encapsulating drugs inside plasma membranes derived from various cells involved in inflammatory processes can be effective against inflammation. This review describes the preparation, characterization, and properties of various types of cell membrane-camouflaged biomimetic nanoparticles. It also summarizes preclinical research into their efficacy against inflammation.
Collapse
Affiliation(s)
- Rongtao Zhang
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China.,School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqiong Wu
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China.,School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qian Ding
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China.,School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qingze Fan
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Dai
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shiwei Guo
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yun Ye
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Meiling Zhou
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
71
|
Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: Improved pharmacokinetics, cell internalization and anticancer efficacy. J Control Release 2021; 335:130-157. [PMID: 34015400 DOI: 10.1016/j.jconrel.2021.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/13/2023]
Abstract
Despite enormous advancements in the field of oncology, the innocuous and effectual treatment of various types of malignancies remained a colossal challenge. The conventional modalities such as chemotherapy, radiotherapy, and surgery have been remained the most viable options for cancer treatment, but lacking of target-specificity, optimum safety and efficacy, and pharmacokinetic disparities are their impliable shortcomings. Though, in recent decades, numerous encroachments in the field of onco-targeted drug delivery have been adapted but several limitations (i.e., short plasma half-life, early clearance by reticuloendothelial system, immunogenicity, inadequate internalization and localization into the onco-tissues, chemoresistance, and deficient therapeutic efficacy) associated with these onco-targeted delivery systems limits their clinical viability. To abolish the aforementioned inadequacies, a promising approach has been emerged in which stealthing of synthetic nanocarriers has been attained by cloaking them into the natural cell membranes. These biomimetic nanomedicines not only retain characteristics features of the synthetic nanocarriers but also inherit the cell-membrane intrinsic functionalities. In this review, we have summarized preparation methods, mechanism of cloaking, and pharmaceutical and therapeutic superiority of cell-membrane camouflaged nanomedicines in improving the bio-imaging and immunotherapy against various types of malignancies. These pliable adaptations have revolutionized the current drug delivery strategies by optimizing the plasma circulation time, improving the permeation into the cancerous microenvironment, escaping the immune evasion and rapid clearance from the systemic circulation, minimizing the immunogenicity, and enabling the cell-cell communication via cell membrane markers of biomimetic nanomedicines. Moreover, the preeminence of cell-membrane cloaked nanomedicines in improving the bio-imaging and theranostic applications, alone or in combination with phototherapy or radiotherapy, have also been pondered.
Collapse
|
72
|
Chen X, Liu B, Tong R, Zhan L, Yin X, Luo X, Huang Y, Zhang J, He W, Wang Y. Orchestration of biomimetic membrane coating and nanotherapeutics in personalized anticancer therapy. Biomater Sci 2021; 9:590-625. [PMID: 33305765 DOI: 10.1039/d0bm01617a] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nanoparticle-based therapeutic and detectable modalities can augment anticancer efficiency, holding potential in capable target and suppressive metastases post administration. However, the individual discrepancies of the current "one-size-fits-all" strategies for anticancer nanotherapeutics have heralded the need for "personalized therapy". Benefiting from the special inherency of various cells, diverse cell membrane-coated nanoparticles (CMCNs) were established on a patient-by-patient basis, which would facilitate the personalized treatment of individual cancer patients. CMCNs in a complex microenvironment can evade the immune system and target homologous tumors with a suppressed immune response, as well as a prolonged circulation time, consequently increasing the drug accumulation at the tumor site and anticancer therapeutic efficacy. This review focuses on the emerging strategies and advances of CMCNs to synergistically integrate the merit of source cells with nanoparticulate delivery systems for the orchestration of personalized anticancer nanotherapeutics, thus discussing their rationalities in facilitating chemotherapy, imaging, immunotherapy, phototherapy, radiotherapy, sonodynamic, magnetocaloric, chemodynamic and gene therapy. Furthermore, the mechanism, challenges and opportunities of CMCNs in personalized anticancer therapy were highlighted to further boost cooperation from different fields, including materials science, chemistry, medicine, pharmacy and biology for the lab-to-clinic translation of CMCNs combined with the individual advantages of source cells and nanotherapeutics.
Collapse
Affiliation(s)
- Xuerui Chen
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Bingbing Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China
| | - Rongliang Tong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lin Zhan
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xuelian Yin
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xin Luo
- Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanan Huang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Wen He
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yanli Wang
- Tumor Precision Targeting Research Center, School of Medicine & School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China. and Institution of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
73
|
Hanley T, Vankayala R, Lee CH, Tang JC, Burns JM, Anvari B. Phototheranostics Using Erythrocyte-Based Particles. Biomolecules 2021; 11:729. [PMID: 34068081 PMCID: PMC8152750 DOI: 10.3390/biom11050729] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
There has been a recent increase in the development of delivery systems based on red blood cells (RBCs) for light-mediated imaging and therapeutic applications. These constructs are able to take advantage of the immune evasion properties of the RBC, while the addition of an optical cargo allows the particles to be activated by light for a number of promising applications. Here, we review some of the common fabrication methods to engineer these constructs. We also present some of the current light-based applications with potential for clinical translation, and offer some insight into future directions in this exciting field.
Collapse
Affiliation(s)
- Taylor Hanley
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; (T.H.); (R.V.); (J.C.T.); (J.M.B.)
| | - Raviraj Vankayala
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; (T.H.); (R.V.); (J.C.T.); (J.M.B.)
- Radoptics, Limited Liability Company, 1002 Health Sciences Road, East, Suite P214, Irvine, CA 92612, USA
| | - Chi-Hua Lee
- Department of Biochemistry, University of California, Riverside, CA 92521, USA;
| | - Jack C. Tang
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; (T.H.); (R.V.); (J.C.T.); (J.M.B.)
| | - Joshua M. Burns
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; (T.H.); (R.V.); (J.C.T.); (J.M.B.)
| | - Bahman Anvari
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; (T.H.); (R.V.); (J.C.T.); (J.M.B.)
| |
Collapse
|
74
|
Chen H, Zheng D, Pan W, Li X, Lv B, Gu W, Machuki JO, Chen J, Liang W, Qin K, Greven J, Hildebrand F, Yu Z, Zhang X, Guo K. Biomimetic Nanotheranostics Camouflaged with Cancer Cell Membranes Integrating Persistent Oxygen Supply and Homotypic Targeting for Hypoxic Tumor Elimination. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19710-19725. [PMID: 33890760 DOI: 10.1021/acsami.1c03010] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Treatment resistance of the tumors to photodynamic therapy (PDT) owing to O2 deficiency largely compromised the therapeutic efficacy, which could be addressed via modulating oxygen levels by using O2 self-enriched nanosystems. Here, we report on augmenting the O2-evolving strategy based on a biomimetic, catalytic nanovehicle (named as N/P@MCC), constructed by the catalase-immobilized hollow mesoporous nanospheres by enveloping a cancer cell membrane (CCM), which acts as an efficient nanocontainer to accommodate nitrogen-doped graphene quantum dots (N-GQDs) and protoporphyrin IX (PpIX). Inheriting the virtues of biomimetic CCM cloaking, the CCM-derived shell conferred N/P@MCC nanovehicles with highly specific self-recognition and homotypic targeting toward cancerous cells, ensuring tumor-specific accumulation and superior circulation durations. N-GQDs, for the first time, have been evidenced as a new dual-functional nanoagents with PTT and PDT capacities, enabling the generation of 1O2 for PDT and inducing local low-temperature hyperthermia for thermally ablating cancer cells and infrared thermal imaging (IRT). Leveraging the intrinsic catalytic features of catalase, such N/P@MCC nanovehicles effectively scavenged the excessive H2O2 to sustainably evolve oxygen for a synchronous O2 self-supply and hypoxia alleviation, with an additional benefit because the resulting O2 bubbles could function as an echo amplifier, leading to the sufficient echogenic reflectivity for ultrasound imaging. Concurrently, the elevated O2 reacted with N-GQDs and PpIX to elicit a maximally increased 1O2 output for augmented PDT. Significantly, the ultrasound imaging coupled with fluorescence imaging, IRT, performs a tumor-modulated trimodal bioimaging effect. Overall, this offers a paradigm to rationally explore O2 self-supply strategies focused on versatile nanotheranostics for hypoxic tumor elimination.
Collapse
Affiliation(s)
- Hongliang Chen
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Donghui Zheng
- Department of Nephrology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P. R. China
| | - Wenzhen Pan
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Xiang Li
- Department of Nephrology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P. R. China
- Department of Clinical Laboratory, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223002, P. R. China
| | - Bin Lv
- Department of Orthopedics, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province 212002, P. R. China
| | - Wenxiang Gu
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Jeremiah Ong'achwa Machuki
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| | - Jiahui Chen
- Key Laboratory of Animal Products Processing, Ministry of Agriculture, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Weiqiang Liang
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Kang Qin
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Johannes Greven
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xing Zhang
- Department of Trauma and Reconstructive Surgery, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Kaijin Guo
- Institute of Orthopedics, Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P. R. China
| |
Collapse
|
75
|
Zhang J, Wei K, Shi J, Zhu Y, Guan M, Fu X, Zhang Z. Biomimetic Nanoscale Erythrocyte Delivery System for Enhancing Chemotherapy via Overcoming Biological Barriers. ACS Biomater Sci Eng 2021; 7:1496-1505. [PMID: 33651596 DOI: 10.1021/acsbiomaterials.1c00008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Overcoming multiple biological barriers, including circulation time in vivo, tumor vascular endothelium, reticuloendothelial system (RES), extracellular matrix (ECM), etc., is the key to improve the therapeutic efficacy of drug delivery systems in treating tumors. Inspired by the ability of natural erythrocytes to cross multiple barriers, in this study, a biomimetic delivery system named NE@DOX-Ang2 was developed for enhancing the chemotherapy of breast cancer, which employed nano-erythrocyte (NE) encapsulating doxorubicin (DOX) and surface modification with a targeted angiopep-2 peptide (Ang2). NE@DOX-Ang2 enhanced the capacity to cross biological barriers in a three-dimensional (3D) tumor spheroid model and in vivo in mice. Compared with a conventional drug delivery system of liposomes, the half-life of NE@DOX-Ang2 increased approximately 2.5 times. Moreover, NE@DOX-Ang2 exhibited excellent tumor-targeting ability and antitumor effects in vitro and in vivo. Briefly, the prepared nano-erythrocyte drug carrier has features of favorable biocompatibility and low immunogenicity and the advantage of prolonging the half-life of drugs, which may provide a novel perspective for development of clinically available nanomedicines.
Collapse
Affiliation(s)
- Junli Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China
| | - Kaiyan Wei
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, 3 Kangfu Road, Zhengzhou 450052, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China
| | - Yifan Zhu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China
| | - Mengting Guan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China
| | - Xudong Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, 3 Kangfu Road, Zhengzhou 450052, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, China
| |
Collapse
|
76
|
Song R, Ruan M, Dai J, Xue W. Biomimetic magnetofluorescent ferritin nanoclusters for magnetic resonance and fluorescence-dual modal imaging and targeted tumor therapy. J Mater Chem B 2021; 9:2494-2504. [PMID: 33656039 DOI: 10.1039/d0tb02175j] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multiple imaging by combining magnetic resonance (MR) and fluorescence imaging into a single nanosystem displays distinctive merits, which is desirable for precise in vivo imaging. In this work, we proposed a new tumor-targeting dual-modal diagnosis strategy by designing and fabricating a biocompatible nano-erythrocyte and successfully delivering it into in vivo tumors. The novel nano-contrast agent (CMR) was prepared by encapsulating human heavy-chain ferritin (HFn) nanoparticles with Cy5.5 binding and mineralized iron oxide nanoparticles (Fe3O4 NPs) into erythrocyte membranes (RBCs). We demonstrated that the as-prepared CMR displayed excellent biocompatibility with low hepatotoxicity and long blood circulation time. More importantly, by functionalizing the CMR with different types of targeting moieties, the nanosystem could precisely target both subcutaneous and orthotopic tumors, and exhibited excellent MR and fluorescence dual-model imaging ability. Moreover, we demonstrated that the CMR was able to modulate the tumor microenvironment to achieve an efficient antitumor effect.
Collapse
Affiliation(s)
- Rongguang Song
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Miaoliang Ruan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Jian Dai
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China. and Institute of Life and Health Engineering, Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Jinan University, Guangzhou 510632, China and The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| |
Collapse
|
77
|
Oroojalian F, Beygi M, Baradaran B, Mokhtarzadeh A, Shahbazi MA. Immune Cell Membrane-Coated Biomimetic Nanoparticles for Targeted Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006484. [PMID: 33577127 DOI: 10.1002/smll.202006484] [Citation(s) in RCA: 209] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Nanotechnology has provided great opportunities for managing neoplastic conditions at various levels, from preventive and diagnostic to therapeutic fields. However, when it comes to clinical application, nanoparticles (NPs) have some limitations in terms of biological stability, poor targeting, and rapid clearance from the body. Therefore, biomimetic approaches, utilizing immune cell membranes, are proposed to solve these issues. For example, macrophage or neutrophil cell membrane coated NPs are developed with the ability to interact with tumor tissue to suppress cancer progression and metastasis. The functionality of these particles largely depends on the surface proteins of the immune cells and their preserved function during membrane extraction and coating process on the NPs. Proteins on the outer surface of immune cells can render a wide range of activities to the NPs, including prolonged blood circulation, remarkable competency in recognizing antigens for enhanced targeting, better cellular interactions, gradual drug release, and reduced toxicity in vivo. In this review, nano-based systems coated with immune cells-derived membranous layers, their detailed production process, and the applicability of these biomimetic systems in cancer treatment are discussed. In addition, future perspectives and challenges for their clinical translation are also presented.
Collapse
Affiliation(s)
- Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
| | - Mohammad Beygi
- Department of Agricultural Engineering, Isfahan University of Technology (IUT), Isfahan, 84156-83111, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 51666-14731, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 51666-14731, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, 45139-56184, Iran
| |
Collapse
|
78
|
Castro F, Martins C, Silveira MJ, Moura RP, Pereira CL, Sarmento B. Advances on erythrocyte-mimicking nanovehicles to overcome barriers in biological microenvironments. Adv Drug Deliv Rev 2021; 170:312-339. [PMID: 32946921 DOI: 10.1016/j.addr.2020.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/29/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
Although nanocarriers offer many advantages as drug delivery systems, their poor stability in circulation, premature drug release and nonspecific uptake in non-target organs have prompted biomimetic approaches using natural cell membranes to camouflage nanovehicles. Among them, erythrocytes, representing the most abundant blood circulating cells, have been extensively investigated for biomimetic coating on artificial nanocarriers due to their upgraded biocompatibility, biodegradability, non-immunogenicity and long-term blood circulation. Due to the cell surface mimetic properties combined with customized core material, erythrocyte-mimicking nanovehicles (EM-NVs) have a wide variety of applications, including drug delivery, imaging, phototherapy, immunomodulation, sensing and detection, that foresee a huge potential for therapeutic and diagnostic applications in several diseases. In this review, we summarize the recent advances in the biomedical applications of EM-NVs in cancer, infection, heart-, autoimmune- and CNS-related disorders and discuss the major challenges and opportunities in this research area.
Collapse
Affiliation(s)
- Flávia Castro
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria José Silveira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Rui Pedro Moura
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite Pereira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
79
|
Seaberg J, Montazerian H, Hossen MN, Bhattacharya R, Khademhosseini A, Mukherjee P. Hybrid Nanosystems for Biomedical Applications. ACS NANO 2021; 15:2099-2142. [PMID: 33497197 PMCID: PMC9521743 DOI: 10.1021/acsnano.0c09382] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Inorganic/organic hybrid nanosystems have been increasingly developed for their versatility and efficacy at overcoming obstacles not readily surmounted by nonhybridized counterparts. Currently, hybrid nanosystems are implemented for gene therapy, drug delivery, and phototherapy in addition to tissue regeneration, vaccines, antibacterials, biomolecule detection, imaging probes, and theranostics. Though diverse, these nanosystems can be classified according to foundational inorganic/organic components, accessory moieties, and architecture of hybridization. Within this Review, we begin by providing a historical context for the development of biomedical hybrid nanosystems before describing the properties, synthesis, and characterization of their component building blocks. Afterward, we introduce the architectures of hybridization and highlight recent biomedical nanosystem developments by area of application, emphasizing hybrids of distinctive utility and innovation. Finally, we draw attention to ongoing clinical trials before recapping our discussion of hybrid nanosystems and providing a perspective on the future of the field.
Collapse
Affiliation(s)
- Joshua Seaberg
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
| | - Hossein Montazerian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA 90095, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Md Nazir Hossen
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
80
|
Zhang M, Cheng S, Jin Y, Zhang N, Wang Y. Membrane engineering of cell membrane biomimetic nanoparticles for nanoscale therapeutics. Clin Transl Med 2021; 11:e292. [PMID: 33635002 PMCID: PMC7819108 DOI: 10.1002/ctm2.292] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, cell membrane camouflaging technology has emerged as an important strategy of nanomedicine, and the modification on the membranes is also a promising approach to enhance the properties of the nanoparticles, such as cancer targeting, immune evasion, and phototherapy sensitivity. Indeed, diversified approaches have been exploited to re-engineer the membranes of nanoparticles in several studies. In this review, first we discuss direct modification strategy of cell membrane camouflaged nanoparticles (CM-NP) via noncovalent, covalent, and enzyme-involved methods. Second, we explore how the membranes of CM-NPs can be re-engineered at the cellular level using strategies such as genetic engineering and membranes fusion. Due to the innate biological properties and excellent biocompatibility, the functionalized cell membrane-camouflaged nanoparticles have been widely applied in the fields of drug delivery, imaging, detoxification, detection, and photoactivatable therapy.
Collapse
Affiliation(s)
- Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Shanshan Cheng
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yue Jin
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Nan Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yu Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghaiChina
- Shanghai Key Laboratory of Gynecologic OncologyShanghaiChina
| |
Collapse
|
81
|
Xie M, Deng T, Li J, Shen H. The camouflage of graphene oxide by red blood cell membrane with high dispersibility for cancer chemotherapy. J Colloid Interface Sci 2021; 591:290-299. [PMID: 33609896 DOI: 10.1016/j.jcis.2021.01.088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/05/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023]
Abstract
Cancer is a serious threat to human health. Graphene oxide (GO) is a good carrier for cancer treatment due to its large surface area and high drug loading, while it's unstable under physiological conditions with a high tendency to be uptaken by macrophages in the body. This paper constructs a red blood cell (RBC) membrane modified GO nanocarrier system for cancer chemotherapy. After the modification of RBC, the stability and hemolysis performance of GO were greatly improved, which is beneficial to the biological application. Moreover, DOX-loaded RBC-GO still able to maintain good stability with a pH-dependent DOX release profile. RBC-GO can be uptaken by MCF-7 cells and DOX-loaded RBC-GO nanocomposites have strong concentration-dependent cytotoxicity. More importantly, in vivo study showed that RBC-GO can accumulate at the tumor site in a large quantity, and among all the experimental groups, RBC-GO-DOX had the best anti-tumor effect after tail vein injection in mice and the lowest systemic toxicity. Experiments have proved that RBC-GO can be used as a drug carrier to achieve targeted drug delivery.
Collapse
Affiliation(s)
- Meng Xie
- School of Pharmacy, Jiangsu University, 212013, China.
| | - Tongtong Deng
- School of Pharmacy, Jiangsu University, 212013, China
| | - Jiaqian Li
- School of Pharmacy, Jiangsu University, 212013, China
| | - Haijun Shen
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 212013, China.
| |
Collapse
|
82
|
Luo GF, Chen WH, Zeng X, Zhang XZ. Cell primitive-based biomimetic functional materials for enhanced cancer therapy. Chem Soc Rev 2021; 50:945-985. [PMID: 33226037 DOI: 10.1039/d0cs00152j] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cell primitive-based functional materials that combine the advantages of natural substances and nanotechnology have emerged as attractive therapeutic agents for cancer therapy. Cell primitives are characterized by distinctive biological functions, such as long-term circulation, tumor specific targeting, immune modulation etc. Moreover, synthetic nanomaterials featuring unique physical/chemical properties have been widely used as effective drug delivery vehicles or anticancer agents to treat cancer. The combination of these two kinds of materials will catalyze the generation of innovative biomaterials with multiple functions, high biocompatibility and negligible immunogenicity for precise cancer therapy. In this review, we summarize the most recent advances in the development of cell primitive-based functional materials for cancer therapy. Different cell primitives, including bacteria, phages, cells, cell membranes, and other bioactive substances are introduced with their unique bioactive functions, and strategies in combining with synthetic materials, especially nanoparticulate systems, for the construction of function-enhanced biomaterials are also summarized. Furthermore, foreseeable challenges and future perspectives are also included for the future research direction in this field.
Collapse
Affiliation(s)
- Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | | | | | | |
Collapse
|
83
|
Fontana F, Bartolo R, Santos HA. Biohybrid Nanosystems for Cancer Treatment: Merging the Best of Two Worlds. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:135-162. [PMID: 33543459 DOI: 10.1007/978-3-030-58174-9_7] [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: 01/30/2023]
Abstract
During the last 20+ years, research into the biomedical application of nanotechnology has helped in reshaping cancer treatment. The clinical use of several passively targeted nanosystems resulted in improved quality of care for patients. However, the therapeutic efficacy of these systems is not superior to the original drugs. Moreover, despite extensive investigations into actively targeted nanocarriers, numerous barriers still remain before their successful clinical translation, including sufficient bloodstream circulation time and efficient tumor targeting. The combination of synthetic nanomaterials with biological elements (e.g., cells, cell membranes, and macromolecules) is presently the cutting-edge research in cancer nanotechnology. The features provided by the biological moieties render the particles with prolonged bloodstream circulation time and homotopic targeting to the tumor site. Moreover, cancer cell membranes serve as sources of neoantigens, useful in the formulation of nanovaccines. In this chapter, we will discuss the advantages of biohybrid nanosystems in cancer chemotherapy, immunotherapy, and combined therapy, as well as highlight their preparation methods and clinical translatability.
Collapse
Affiliation(s)
- Flavia Fontana
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Raquél Bartolo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
84
|
Li M, Cui X, Wei F, Wang Z, Han X. Red blood cell membrane-coated biomimetic upconversion nanoarchitectures for synergistic chemo-photodynamic therapy. NEW J CHEM 2021. [DOI: 10.1039/d1nj04305f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The red blood cell (RBC) membrane camouflaged core–shell upconversion/mesoporous silica nanoparticles (UCNPs@mSiO2 NPs) were constructed for synergistic chemo-photodynamic therapy.
Collapse
Affiliation(s)
- Minghui Li
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Xinyu Cui
- Department of Public Health, Mudanjiang Medical University, Mudanjiang, China
| | - Feng Wei
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Zhao Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Xiaojun Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
85
|
Thangaraju K, Neerukonda SN, Katneni U, Buehler PW. Extracellular Vesicles from Red Blood Cells and Their Evolving Roles in Health, Coagulopathy and Therapy. Int J Mol Sci 2020; 22:E153. [PMID: 33375718 PMCID: PMC7796437 DOI: 10.3390/ijms22010153] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Red blood cells (RBCs) release extracellular vesicles (EVs) including both endosome-derived exosomes and plasma-membrane-derived microvesicles (MVs). RBC-derived EVs (RBCEVs) are secreted during erythropoiesis, physiological cellular aging, disease conditions, and in response to environmental stressors. RBCEVs are enriched in various bioactive molecules that facilitate cell to cell communication and can act as markers of disease. RBCEVs contribute towards physiological adaptive responses to hypoxia as well as pathophysiological progression of diabetes and genetic non-malignant hematologic disease. Moreover, a considerable number of studies focus on the role of EVs from stored RBCs and have evaluated post transfusion consequences associated with their exposure. Interestingly, RBCEVs are important contributors toward coagulopathy in hematological disorders, thus representing a unique evolving area of study that can provide insights into molecular mechanisms that contribute toward dysregulated hemostasis associated with several disease conditions. Relevant work to this point provides a foundation on which to build further studies focused on unraveling the potential roles of RBCEVs in health and disease. In this review, we provide an analysis and summary of RBCEVs biogenesis, composition, and their biological function with a special emphasis on RBCEV pathophysiological contribution to coagulopathy. Further, we consider potential therapeutic applications of RBCEVs.
Collapse
Affiliation(s)
- Kiruphagaran Thangaraju
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.); (P.W.B.)
| | - Sabari Nath Neerukonda
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA;
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Upendra Katneni
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.); (P.W.B.)
| | - Paul W. Buehler
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.); (P.W.B.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
86
|
Bush LM, Healy CP, Javdan SB, Emmons JC, Deans TL. Biological Cells as Therapeutic Delivery Vehicles. Trends Pharmacol Sci 2020; 42:106-118. [PMID: 33342562 DOI: 10.1016/j.tips.2020.11.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022]
Abstract
One of the significant challenges remaining in the field of drug delivery is insufficient targeting of diseased tissues or cells. While efforts to perform targeted drug delivery by engineered nanoparticles have shown some success, there are underlying targeting, toxicity, and immunogenicity challenges. By contrast, live cells usually have innate targeting mechanisms, and can be used as drug-delivery vehicles to increase the efficiency with which a drug accumulates to act on the intended tissue. In some cases, when no native cell types exhibit the desired therapeutic phenotype, preferred outcomes can be achieved by genetically modifying and reprogramming cells with gene circuits. This review highlights recent advances in the use of cells to deliver therapeutics. Specifically, we discuss how red blood cells (RBCs), platelets, neutrophils, mesenchymal stem cells (MSCs), and bacteria have been utilized to advance drug delivery.
Collapse
Affiliation(s)
- Lucas M Bush
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Connor P Healy
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Shwan B Javdan
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Jonathan C Emmons
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Tara L Deans
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
87
|
Jha A, Nikam AN, Kulkarni S, Mutalik SP, Pandey A, Hegde M, Rao BSS, Mutalik S. Biomimetic nanoarchitecturing: A disguised attack on cancer cells. J Control Release 2020; 329:413-433. [PMID: 33301837 DOI: 10.1016/j.jconrel.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
With the changing face of healthcare, there is a demand for drug delivery systems that have increased efficacy and biocompatibility. Nanotechnology derived drug carrier systems were found to be ideal candidates to meet these demands. Among the vast number of nanosized delivery systems, biomimetic nanoparticles have been researched at length. These nanoparticles mimic cellular functions and are highly biocompatible. They are also able to avoid clearance by the reticuloendothelial system which increases the time spent by them in the systemic circulation. Additionally, their low immunogenicity and targeting ability increase their significance as drug carriers. Based on their core material we have summarized them as biomimetic inorganic nanoparticles, biomimetic polymeric nanoparticles, and biomimetic lipid nanoparticles. The core then may be coated using membranes derived from erythrocytes, cancer cells, leukocytes, stem cells, and other membranes to endow them with biomimetic properties. They can be used for personalized therapy and diagnosis of a large number of diseases, primarily cancer. This review summarizes the various therapeutic approaches using biomimetic nanoparticles along with their applications in the field of cancer imaging, nucleic acid therapy and theranostic properties. A brief overview about toxicity concerns related to these nanoconstructs has been added to provide knowledge about biocompatibility of such nanoparticles.
Collapse
Affiliation(s)
- Adrija Jha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Sadhana P Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | - Manasa Hegde
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India
| | | | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka, India.
| |
Collapse
|
88
|
Jiménez-Jiménez C, Manzano M, Vallet-Regí M. Nanoparticles Coated with Cell Membranes for Biomedical Applications. BIOLOGY 2020; 9:biology9110406. [PMID: 33218092 PMCID: PMC7698879 DOI: 10.3390/biology9110406] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Simple Summary Nanomedicine has developed a new technology based on nanoparticles for drug delivery coated with different cell membranes. Although they were originally developed to increase their blood circulation time and stability though the use of red blood cell membranes, the versatility of this technology has extended to membranes from different cell types, such as white blood cells, platelets, cancer cells, mesenchymal stem cells, and beta cells, among others. Therefore, this cellular diversity and its unique properties, together with the possibility of using a wide range of nanoparticles and different drug dosage forms, has opened a new area for the manufacture of nanoparticles, with many potential applications in the clinic. Abstract Nanoparticles designed for diagnosing and treating different diseases have impacted the scientific research in biomedicine, and are expected to revolutionize the clinic in the near future through a new area called nanomedicine. In the last few years, a new approach in this field has emerged: the use of cell membranes for coating nanoparticles in an attempt to mimic the ability of cells to interface and interact with physiological environments. Although such functions have been replicated through synthetic techniques, many research groups are now employing naturally derived cell membranes to coat different types of nanoparticles in an attempt to improve their performance for a wide range of applications. This review summarizes the literature on nanoparticles coated with cell membranes and, more importantly, aims at inspiring and encouraging new developments to this technology in the biomedical area.
Collapse
Affiliation(s)
- Carla Jiménez-Jiménez
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, UCM, Instituto Investigación Sanitaria Hospital 12 de Octubre, imas12, 28040 Madrid, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Miguel Manzano
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, UCM, Instituto Investigación Sanitaria Hospital 12 de Octubre, imas12, 28040 Madrid, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (M.M.); (M.V.-R.)
| | - María Vallet-Regí
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, UCM, Instituto Investigación Sanitaria Hospital 12 de Octubre, imas12, 28040 Madrid, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (M.M.); (M.V.-R.)
| |
Collapse
|
89
|
Kermanizadeh A, Jacobsen NR, Murphy F, Powell L, Parry L, Zhang H, Møller P. A Review of the Current State of Nanomedicines for Targeting and Treatment of Cancers: Achievements and Future Challenges. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Fiona Murphy
- Heriot Watt University School of Engineering and Physical Sciences Edinburgh EH14 4AS UK
| | - Leagh Powell
- Heriot Watt University School of Engineering and Physical Sciences Edinburgh EH14 4AS UK
| | - Lee Parry
- Cardiff University European Cancer Stem Cell Research Institute, School of Biosciences Cardiff CF24 4HQ UK
| | - Haiyuan Zhang
- Changchun Institute of Applied Chemistry Laboratory of Chemical Biology Changchun 130022 China
| | - Peter Møller
- University of Copenhagen Department of Public Health Copenhagen DK1014 Denmark
| |
Collapse
|
90
|
Dash P, Piras AM, Dash M. Cell membrane coated nanocarriers - an efficient biomimetic platform for targeted therapy. J Control Release 2020; 327:546-570. [DOI: 10.1016/j.jconrel.2020.09.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 01/08/2023]
|
91
|
Liu Q, Fang H, Gai Y, Lan X. pH-Triggered Assembly of Natural Melanin Nanoparticles for Enhanced PET Imaging. Front Chem 2020; 8:755. [PMID: 33134253 PMCID: PMC7579405 DOI: 10.3389/fchem.2020.00755] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/21/2020] [Indexed: 02/03/2023] Open
Abstract
Natural melanin nanoplatforms have attracted attention in molecular imaging. Natural melanin can be made into small-sized nanoparticles, which penetrate tumor sites deeply, but unfortunately, the particles continue to backflow into the blood or are cleared into the surrounding tissues, leading to loss of retention within tumors. Here, we report a pH-triggered approach to aggregate natural melanin nanoparticles by introducing a hydrolysis-susceptible citraconic amide on the surface. Triggered by pH values lower than 7.0, such as the tumor acid environment, the citraconic amide moiety tended to hydrolyze abruptly, resulting in both positive and negative surface charges. The electrostatic attractions between nanoparticles drove nanoparticle aggregation, which increased accumulation in the tumor site because backflow was blocked by the increased size. Melanin nanoparticles have the natural ability to bind metal ions, which can be labeled with isotopes for nuclear medicine imaging. When the melanin nanoparticles were labeled by 68Ga, we observed that the pH-induced physical aggregation in tumor sites resulted in enhanced PET imaging. The pH-triggered assembly of natural melanin nanoparticles could be a practical strategy for efficient tumor targeted imaging.
Collapse
Affiliation(s)
- Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
92
|
Chen HY, Deng J, Wang Y, Wu CQ, Li X, Dai HW. Hybrid cell membrane-coated nanoparticles: A multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater 2020; 112:1-13. [PMID: 32470527 DOI: 10.1016/j.actbio.2020.05.028] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/06/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Biomimetic nanotechnology through camouflaging synthetic nanoparticles (NPs) with natural cell membranes, which bestows with immune evasion and superior targeting capacity, has been extensively used in drug delivery systems (DDS) over the last decades. These biomimetic NPs not only retain the physicochemical features of the synthetic vehicles but also inherit the cell membranes' intrinsic functionalities. Combined with these benefits, optimized nano-biomimetic DDS allow maximum delivery efficacy. Compared to erythrocyte/cancer single cell membrane, the hybrid cell membrane expressing CD47 membrane protein and self-recognition molecules, from erythrocytes and cancer cells, provides remarkable features to the synthetic vehicles, such as immune evasion, long-term circulation, and homotypic targeting. In this review, we describe the preparation strategies, the camouflaging mechanism, and the antitumor applications of hybrid cell membrane-camouflaged NPs. Moreover, we discuss further modification of the hybrid cell membrane and the surface properties of fusion cellular membranes. Finally, we summarize the primary challenges and opportunities associated with these NPs. STATEMENT OF SIGNIFICANCE: Camouflaging synthetic nanoparticles with hybrid cell membrane has been extensively highlighted in recent years. The resultant biomimetic nanoparticles not only reserve the physicochemical properties of the synthetic nanoparticles but also inherit the biological functions of source cells. Compared with single cell membrane, hybrid cell membrane can endow synthetic nanoparticles with multiple biofunctions derived from the original source cells. To provide a timely review of this rapidly developing subject of research, this paper summarized recent progress on the hybrid cell membrane-camouflaged nanoparticles as drug delivery systems for cancer diagnosis and treatment. In this review, we focused primarily on five different types of hybrid cell membrane-camouflaged nanoparticles with the preparation strategies, the camouflaging mechanism, and the antitumor applications. Moreover, further modification of the hybrid cell membrane was also discussed for isolating effectively circulating tumor cells.
Collapse
|
93
|
Xu Y, Lu Q, Sun L, Feng S, Nie Y, Ning X, Lu M. Nanosized Phase-Changeable "Sonocyte" for Promoting Ultrasound Assessment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002950. [PMID: 32697421 DOI: 10.1002/smll.202002950] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/16/2020] [Indexed: 05/13/2023]
Abstract
Despite the ability of microbubble contrast agents to improve ultrasound diagnostic performance, their application potential is limited due to low stability, fast clearance, and poor tissue permeation. This study presents a promising nanosized phase-changeable erythrocyte (Sonocyte), composed of liposomal dodecafluoropentane coated with multilayered red blood cell membranes (RBCm), for improving ultrasound assessments. Sonocyte is the first RBCm-functionalized ultrasound contrast agent with uniform nanosized morphology, and exhibits good stability, systemic circulation, target-tissue accumulation, and even ultrasound-responsive phase transition, thereby satisfying the inherent requirement of ultrasound imaging. It is identified that Sonocyte displays similar sensitivity as microbubble SonoVue, a clinical ultrasound contrast agent, for effectively detecting normal parenchyma and hepatic necrosis. Importantly, compared with SonoVue lacking of ability to detect tumors, Sonocyte can identify tumors with high sensitivity and specificity due to superior tumor accumulation and penetration. Therefore, Sonocyte exhibits superior capabilities over SonoVue, endowing with a great clinical application potential.
Collapse
Affiliation(s)
- Yurui Xu
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Qiangbing Lu
- National Laboratory of Solid State Microstructures, Department of Materials Science and Engineering, Nanjing University, Nanjing, 210093, China
| | - Lei Sun
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Shujun Feng
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Yuanyuan Nie
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210093, China
| | - Minghui Lu
- National Laboratory of Solid State Microstructures, Department of Materials Science and Engineering, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
94
|
Hou Z, Liu Y, Xu J, Zhu J. Surface engineering of magnetic iron oxide nanoparticles by polymer grafting: synthesis progress and biomedical applications. NANOSCALE 2020; 12:14957-14975. [PMID: 32648868 DOI: 10.1039/d0nr03346d] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Magnetic iron oxide nanoparticles (IONPs) have wide applications in magnetic resonance imaging (MRI), biomedicine, drug delivery, hyperthermia therapy, catalysis, magnetic separation, and others. However, these applications are usually limited by irreversible agglomeration of IONPs in aqueous media because of their dipole-dipole interactions, and their poor stability. A protecting polymeric shell provides IONPs with not only enhanced long-term stability, but also the functionality of polymer shells. Therefore, polymer-grafted IONPs have recently attracted much attention of scientists. In this tutorial review, we will present the current strategies for grafting polymers onto the surface of IONPs, basically including "grafting from" and "grafting to" methods. Available functional groups and chemical reactions, which could be employed to bind polymers onto the IONP surface, are comprehensively summarized. Moreover, the applications of polymer-grafted IONPs will be briefly discussed. Finally, future challenges and perspectives in the synthesis and application of polymer-grafted IONPs will also be discussed.
Collapse
Affiliation(s)
- Zaiyan Hou
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| | - Yijing Liu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| | - Jiangping Xu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| | - Jintao Zhu
- Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education (HUST), School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| |
Collapse
|
95
|
Chen S, Ren Y, Duan P. Biomimetic nanoparticle loading obatoclax mesylate for the treatment of non-small-cell lung cancer (NSCLC) through suppressing Bcl-2 signaling. Biomed Pharmacother 2020; 129:110371. [PMID: 32563984 DOI: 10.1016/j.biopha.2020.110371] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Lung cancer still remains a leading cause of cancer mortality in the world. Obatoclax mesylate (OM), a B cell chronic lymphocytic leukemia/lymphoma 2 (Bcl-2) family antagonist, is a potential antitumor drug. However, its poor aqueous solubility restricts its clinical application. Although these inherent defects, nanotechnology can be used to improve the solubility and tumor target of OM, promoting its antitumor efficiency. In the present study, the poly(lactic-coglycolic acid) (PLGA) was used and combined with red blood-cell membrane (RBCm) to explore if OM-loaded RBCm nanoparticles could improve the antitumor efficacy of OM for the treatment of lung cancer with relatively lower side effects compared with the free OM. The good physicochemical stability of the prepared RBCm-OM/PLGA nanoparticles was confirmed, and the optimal size of 153 nm was screened out, along with sustained drug release behavior. We found that RBCm-OM/PLGA nanoparticles effectively reduced the proliferation of lung cancer cells. Additionally, RBCm-OM/PLGA nanoparticles considerably induced apoptosis in lung cancer cells by reducing Bcl-2 expression levels, accompanied with the improved Cyto-c releases in cytoplasm and Caspase-3 activation. Mitochondrial membrane potential was also obviously impaired in lung cancer cells incubated with RBCm-OM/PLGA nanoparticles. Compared with free OM, RBCm-OM/PLGA nanoparticles could greatly prolong the drug circulation time in vivo and upgraded the drug concentration accumulated in tumor tissue. Furthermore, RBCm-OM/PLGA nanoparticles exerted stronger antitumor efficacy in vivo against lung cancer progression with superior safety. Therefore, RBCm-OM/PLGA nanoparticles provided new potential for lung cancer therapy with the improved safety and therapeutic effect.
Collapse
Affiliation(s)
- Song Chen
- Department of Radiology, XD Group Hospital, Xi'an City, Shaanxi Province, 710077, China
| | - Yujie Ren
- Department of CT Room, Dongying People's Hospital, Dongying City, Shandong Province, 257091, China
| | - Peng Duan
- Department of Oncology, The Third People's Hospital of Qingdao, Qingdao City, Shandong Province, 266041, China.
| |
Collapse
|
96
|
Gong C, Yu X, You B, Wu Y, Wang R, Han L, Wang Y, Gao S, Yuan Y. Macrophage-cancer hybrid membrane-coated nanoparticles for targeting lung metastasis in breast cancer therapy. J Nanobiotechnology 2020; 18:92. [PMID: 32546174 PMCID: PMC7298843 DOI: 10.1186/s12951-020-00649-8] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/11/2020] [Indexed: 12/27/2022] Open
Abstract
Cell membrane- covered drug-delivery nanoplatforms have been garnering attention because of their enhanced bio-interfacing capabilities that originate from source cells. In this top-down technique, nanoparticles (NPs) are covered by various membrane coatings, including membranes from specialized cells or hybrid membranes that combine the capacities of different types of cell membranes. Here, hybrid membrane-coated doxorubicin (Dox)-loaded poly(lactic-co-glycolic acid) (PLGA) NPs (DPLGA@[RAW-4T1] NPs) were fabricated by fusing membrane components derived from RAW264.7(RAW) and 4T1 cells (4T1). These NPs were used to treat lung metastases originating from breast cancer. This study indicates that the coupling of NPs with a hybrid membrane derived from macrophage and cancer cells has several advantages, such as the tendency to accumulate at sites of inflammation, ability to target specific metastasis, homogenous tumor targeting abilities in vitro, and markedly enhanced multi-target capability in a lung metastasis model in vivo. The DPLGA@[RAW-4T1] NPs exhibited excellent chemotherapeutic potential with approximately 88.9% anti-metastasis efficacy following treatment of breast cancer-derived lung metastases. These NPs were robust and displayed the multi-targeting abilities of hybrid membranes. This study provides a promising biomimetic nanoplatform for effective treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Chunai Gong
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Xiaoyan Yu
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Benming You
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yan Wu
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Rong Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Lu Han
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Yujie Wang
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Shen Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China.
| |
Collapse
|
97
|
Rodríguez-Rodríguez H, Salas G, Arias-Gonzalez JR. Heat Generation in Single Magnetic Nanoparticles under Near-Infrared Irradiation. J Phys Chem Lett 2020; 11:2182-2187. [PMID: 32119551 DOI: 10.1021/acs.jpclett.0c00143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Heat generation by pointlike structures is an appealing concept for its implications in nanotechnology and biomedicine. The way to pump energy that excites heat locally and the synthesis of nanostructures that absorb such energy are key issues in this endeavor. High-frequency alternating magnetic or near-infrared optical fields are used to induce heat in iron oxide nanoparticles, a combined solution that is being exploited in hyperthermia treatments. However, the temperature determination around a single iron oxide nanoparticle remains a challenge. We study the heat released from iron oxide nanostructures under near-infrared illumination on a one-by-one basis by optical tweezers. To measure the temperature, we follow the medium viscosity changes around the trapped particle as a function of the illuminating power, thus avoiding the use of thermal probes. Our results help interpret temperature, a statistical parameter, in the nanoscale and the concept of heat production by nanoparticles under thermal agitation.
Collapse
Affiliation(s)
- Héctor Rodríguez-Rodríguez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA-Nanoscience), Cantoblanco 28049, Madrid, Spain
- Departamento de Quı́mica-Fı́sica Aplicada, Universidad Autónoma de Madrid, 28049 Cantoblanco, Madrid, Spain
| | - Gorka Salas
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA-Nanoscience), Cantoblanco 28049, Madrid, Spain
- CNB-CSIC-IMDEA Nanociencia Associated Unit "Unidad de Nanobiotecnologı́a", Madrid, Spain
| | - J Ricardo Arias-Gonzalez
- Centro de Tecnologı́as Fı́sicas, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| |
Collapse
|
98
|
A Review on the Optimal Design of Magnetic Nanoparticle-Based T2 MRI Contrast Agents. MAGNETOCHEMISTRY 2020. [DOI: 10.3390/magnetochemistry6010011] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Relaxivity r2 and thus the contrast efficacy of superparamagnetic nanoparticles (NPs) can be enhanced via either NP’s magnetic properties or coating optimization. Numerous reports can be found about the investigation of the optimal iron oxide nanoparticles (IO NPs) size, shape, crystallinity and composition that yield high saturation magnetization (ms) values and, consequently, high r2 values. Although the use of an appropriate coating can boost up the NPs MRI contrast agent efficiency, this topic has been largely understudied. Therefore, in this review, the factors affording r2 enhancement of spherical magnetic NPs are discussed. Based on the literature, the requirements for an optimal surface coating that may increase r2 values and ensure stability and biocompatibility of NPs are listed. One of the best candidates that fulfil these requirements are liposomes with embedded magnetic NPs, so-called magneto-liposomes. The analysis of the literature elucidated the most appropriate phospholipid compositions for the relaxivity enhancement and for magneto-liposomes in vivo stability. Finally, the future directions in the development of NP-based contrast agents are given. For example, most of the synthetic NPs are recognized and eliminated as a foreign substance by the immune system. To overcome this issue, a design of a biomimetic, cell-membrane-based nanocarrier for contrast agents is proposed. Disguised with cell membranes, NPs or other active components can act as autogenous cells and thus ensure the inherent biocompatibility.
Collapse
|
99
|
Zou S, Wang B, Wang C, Wang Q, Zhang L. Cell membrane-coated nanoparticles: research advances. Nanomedicine (Lond) 2020; 15:625-641. [PMID: 32098564 DOI: 10.2217/nnm-2019-0388] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell membranes have been continuously imitated and used for the modification of nanoparticles (NPs) to improve NP biological properties. Cell membrane-coated NPs, where core NPs are wrapped with plasma membrane vesicles, show high biocompatibility, targeting specificity and low side effects. Compared with conventional strategies, this novel approach directly leverages intact and natural functions of cell membranes, instead of replicating these features via synthetic techniques. This top-down technique bestows NPs with enhanced biointerfacing capabilities with potential in the diagnosis and treatment of cancer, infection and other diseases. Herein, we report on the advances in cell membrane-coated NPs, including the preparation process, source cell membranes for wrapping and potential applications of these cell membrane-coated NPs.
Collapse
Affiliation(s)
- Shuaijun Zou
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Beilei Wang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Chao Wang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Qianqian Wang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| | - Liming Zhang
- Marine Bio-pharmaceutical Institute, Naval Medical University, Shanghai, 200433, PR China
| |
Collapse
|
100
|
Daniyal M, Jian Y, Xiao F, Sheng W, Fan J, Xiao C, Wang Z, Liu B, Peng C, Yuhui Q, Wang W. Development of a nanodrug-delivery system camouflaged by erythrocyte membranes for the chemo/phototherapy of cancer. Nanomedicine (Lond) 2020; 15:691-709. [PMID: 32043430 DOI: 10.2217/nnm-2019-0454] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Development of a new drug-delivery system using a compound derived from Pronephrium penangianum (J5) for the treatment of cervical cancer. Materials & methods: The delivery system was developed using Prussian blue nanoparticles, camouflaged by red blood cell membrane and with folic acid surface modifications. Results: Our results showed the successful development of a nanodrug-delivery system, which increases the half-life and immune evasion ability of the drug. The mechanism of this system was through suppressing B-cell lymphoma 2 and increasing B-cell lymphoma 2-associated X protein and the cleaved caspase level. An in vivo study also confirmed good antitumor activity without any side effects to normal tissue. Conclusion: This drug-delivery system provides a good alternative for the treatment of cervical cancer using J5.
Collapse
Affiliation(s)
- Muhammad Daniyal
- TCM & Ethnomedicine Innovative & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan ,410208, PR China
| | - YuQing Jian
- TCM & Ethnomedicine Innovative & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan ,410208, PR China
| | - Feng Xiao
- College of Biology, Hunan University, Changsha, 410082, PR China
| | - Wenbing Sheng
- TCM & Ethnomedicine Innovative & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan ,410208, PR China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, 410082, PR China
| | - Chang Xiao
- College of Biology, Hunan University, Changsha, 410082, PR China
| | - Zhou Wang
- College of Biology, Hunan University, Changsha, 410082, PR China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, PR China
| | - Caiyun Peng
- TCM & Ethnomedicine Innovative & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan ,410208, PR China
| | - Qin Yuhui
- TCM & Ethnomedicine Innovative & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan ,410208, PR China
| | - Wei Wang
- TCM & Ethnomedicine Innovative & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan ,410208, PR China
| |
Collapse
|