301
|
Sushnitha M, Evangelopoulos M, Tasciotti E, Taraballi F. Cell Membrane-Based Biomimetic Nanoparticles and the Immune System: Immunomodulatory Interactions to Therapeutic Applications. Front Bioeng Biotechnol 2020; 8:627. [PMID: 32626700 PMCID: PMC7311577 DOI: 10.3389/fbioe.2020.00627] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Nanoparticle-based drug delivery systems have been synthesized from a wide array of materials. The therapeutic success of these platforms hinges upon their ability to favorably interact with the biological environment (both systemically and locally) and recognize the diseased target tissue. The immune system, composed of a highly coordinated organization of cells trained to recognize foreign bodies, represents a key mediator of these interactions. Although components of this system may act as a barrier to nanoparticle (NP) delivery, the immune system can also be exploited to target and trigger signaling cues that facilitate the therapeutic response stemming from systemic administration of NPs. The nano-bio interface represents the key facilitator of this communication exchange, where the surface properties of NPs govern their in vivo fate. Cell membrane-based biomimetic nanoparticles have emerged as one approach to achieve targeted drug delivery by actively engaging and communicating with the biological milieu. In this review, we will highlight the relationship between these biomimetic nanoparticles and the immune system, emphasizing the role of tuning the nano-bio interface in the immunomodulation of diseases. We will also discuss the therapeutic applications of this approach with biomimetic nanoparticles, focusing on specific diseases ranging from cancer to infectious diseases. Lastly, we will provide a critical evaluation on the current state of this field of cell membrane-based biomimetic nanoparticles and its future directions in immune-based therapy.
Collapse
Affiliation(s)
- Manuela Sushnitha
- Department of Bioengineering, Rice University, Houston, TX, United States
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
302
|
Sun M, Yang D, Fanqi W, Wang Z, Ji H, Liu Z, Gai S, Zhang F, Yang P. SiO 2@Cu 7S 4 nanotubes for photo/chemodynamic and photo-thermal dual-mode synergistic therapy under 808 nm laser irradiation. J Mater Chem B 2020; 8:5707-5721. [PMID: 32510093 DOI: 10.1039/d0tb00696c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Photodynamic therapy (PDT) is a light-based modality for tumor treatment that involves the generation of reactive oxygen species (ROS) by the combination of light, a photosensitizer, and molecular oxygen. Nevertheless, the therapeutic effects of PDT are limited by hypoxic conditions that worsen with oxygen consumption during the PDT process. Photo/chemodynamic therapy (PCDT) based on the Fenton reaction is one strategy to improve ROS generation, provided a highly effective Fenton reagent is developed. In this research, SiO2@Cu7S4 nanotubes (NTs) were synthesized as a PCDT agent. This double-valence metal-sulfide composite material can react with H2O2 at the tumor site. SiO2@Cu7S4 NTs can produce more ROS than the traditional PDT agents, and besides, they can also be used as a photothermal therapy (PTT) agent. SiO2@Cu7S4 NTs will trigger the PTT effect under 808 nm irradiation and generate a large amount of heat to eradicate cancer cells. This heat will also promote the PCDT effect by increasing the reaction rate. Thus, the SiO2@Cu7S4 NT is a suitable material for PCDT and PTT synergistic oncotherapy. The 808 nm laser is selected as the appropriate excitation source, providing adequate penetration and minimal harm to normal cells. The experimental data presented herein demonstrate the promising photosensitive, Fenton-like, and photothermal performance of SiO2@Cu7S4 NTs. Furthermore, the findings could promote the development of PCDT and PTT synergistic therapy. Thus, this research provides a feasible method to design a single, multifunctional material for cancer treatment.
Collapse
Affiliation(s)
- Mingdi Sun
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
303
|
Gao J, Wang F, Wang S, Liu L, Liu K, Ye Y, Wang Z, Wang H, Chen B, Jiang J, Ou J, van Hest JCM, Peng F, Tu Y. Hyperthermia-Triggered On-Demand Biomimetic Nanocarriers for Synergetic Photothermal and Chemotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903642. [PMID: 32537410 PMCID: PMC7284223 DOI: 10.1002/advs.201903642] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/22/2020] [Accepted: 03/08/2020] [Indexed: 05/03/2023]
Abstract
Nanoparticle-based drug delivery systems with low side effects and enhanced efficacy hold great potential in the treatment of various malignancies, in particular cancer; however, they are still challenging to attain. Herein, an anticancer drug delivery system based on a cisplatin (CDDP) containing nanogel, functionalized with photothermal gold nanorods (GNRs) which are electrostatically decorated with doxorubicin (DOX) is reported. The nanoparticles are formed via the crosslinking reaction of hyaluronic acid with the ancillary anticarcinogen CDDP in the presence of DOX-decorated GNRs. The nanogel is furthermore cloaked with a cancer cell membrane, and the resulting biomimetic nanocarrier (4T1-HANG-GNR-DC) shows efficient accumulation by homologous tumor targeting and possesses long-time retention in the tumor microenvironment. Upon near-infrared (NIR) laser irradiation, in situ photothermal therapy is conducted which further induces hyperthermia-triggered on-demand drug release from the nanogel reservoir to achieve a synergistic photothermal/chemo-therapy. The as-developed biomimetic nanocarriers, with their dual-drug delivery features, homotypic tumor targeting and synergetic photothermal/chemo-therapy, show much promise as a potential platform for cancer treatment.
Collapse
Affiliation(s)
- Junbin Gao
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Fei Wang
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Shuanghu Wang
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Lu Liu
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Kun Liu
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Yicheng Ye
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Zhen Wang
- School of Materials Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Hong Wang
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Bin Chen
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Jiamiao Jiang
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Juanfeng Ou
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| | - Jan C. M. van Hest
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600, MBThe Netherlands
| | - Fei Peng
- School of Materials Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Yingfeng Tu
- School of Pharmaceutical ScienceGuangdong Provincial Key Laboratory of New Drug ScreeningSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
304
|
Valcourt DM, Kapadia CH, Scully MA, Dang MN, Day ES. Best Practices for Preclinical In Vivo Testing of Cancer Nanomedicines. Adv Healthc Mater 2020; 9:e2000110. [PMID: 32367687 PMCID: PMC7473451 DOI: 10.1002/adhm.202000110] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/17/2020] [Indexed: 01/06/2023]
Abstract
Significant advances have been made in the development of nanoparticles for cancer treatment in recent years. Despite promising results in preclinical animal models, cancer nanomedicines often fail in clinical trials. This failure rate could be reduced by defining stringent criteria for testing and quality control during the design and development stages, and by performing carefully planned preclinical studies in relevant animal models. This article discusses best practices for the evaluation of nanomedicines in murine tumor models. First, a recommended set of experiments to perform is introduced, including discussion of the types of data to collect during these studies. This is followed by an outline of various tumor models and their clinical relevance. Next, different routes of nanoparticle administration are overviewed, followed by a summary of important controls to include in in vivo studies of nanomedicine. Finally, animal welfare considerations are discussed, and an overview of the steps involved in achieving US Food and Drug Administration approval after animal studies are completed is provided. Researchers should use this report as a guideline for effective preclinical evaluation of cancer nanomedicine. As the community adopts best practices for in vivo testing, the rate of clinical translation of cancer nanomedicines is likely to improve.
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Chintan H Kapadia
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Mackenzie A Scully
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
- Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, DE, 19713, USA
| |
Collapse
|
305
|
Wang H, Wang K, He L, Liu Y, Dong H, Li Y. Engineering antigen as photosensitiser nanocarrier to facilitate ROS triggered immune cascade for photodynamic immunotherapy. Biomaterials 2020; 244:119964. [DOI: 10.1016/j.biomaterials.2020.119964] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/25/2020] [Accepted: 03/10/2020] [Indexed: 12/19/2022]
|
306
|
Jiang Q, Wang K, Zhang X, Ouyang B, Liu H, Pang Z, Yang W. Platelet Membrane-Camouflaged Magnetic Nanoparticles for Ferroptosis-Enhanced Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2001704. [PMID: 32338436 DOI: 10.1002/smll.202001704] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 04/01/2020] [Indexed: 06/11/2023]
Abstract
Although cancer immunotherapy has emerged as a tremendously promising cancer therapy method, it remains effective only for several cancers. Photoimmunotherapy (e.g., photodynamic/photothermal therapy) could synergistically enhance the immune response of immunotherapy. However, excessively generated immunogenicity will cause serious inflammatory response syndrome. Herein, biomimetic magnetic nanoparticles, Fe3 O4 -SAS @ PLT, are reported as a novel approach to sensitize effective ferroptosis and generate mild immunogenicity, enhancing the response rate of non-inflamed tumors for cancer immunotherapy. Fe3 O4 -SAS@PLT are built from sulfasalazine (SAS)-loaded mesoporous magnetic nanoparticles (Fe3 O4 ) and platelet (PLT) membrane camouflage and triggered a ferroptotic cell death via inhibiting the glutamate-cystine antiporter system Xc- pathway. Fe3 O4 -SAS @ PLT-mediated ferroptosis significantly improves the efficacy of programmed cell death 1 immune checkpoint blockade therapy and achieves a continuous tumor elimination in a mouse model of 4T1 metastatic tumors. Proteomics studies reveal that Fe3 O4 -SAS @ PLT-mediated ferroptosis could not only induce tumor-specific immune response but also efficiently repolarize macrophages from immunosuppressive M2 phenotype to antitumor M1 phenotype. Therefore, the concomitant of Fe3 O4 -SAS @ PLT-mediated ferroptosis with immunotherapy are expected to provide great potential in the clinical treatment of tumor metastasis.
Collapse
Affiliation(s)
- Qin Jiang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, P. R. China
| | - Kuang Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, P. R. China
| | - Xingyu Zhang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Boshu Ouyang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Haixia Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, P. R. China
| | - Zhiqing Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
307
|
Wang J, Sun J, Hu W, Wang Y, Chou T, Zhang B, Zhang Q, Ren L, Wang H. A Porous Au@Rh Bimetallic Core-Shell Nanostructure as an H 2 O 2 -Driven Oxygenerator to Alleviate Tumor Hypoxia for Simultaneous Bimodal Imaging and Enhanced Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001862. [PMID: 32329171 PMCID: PMC7386557 DOI: 10.1002/adma.202001862] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 05/19/2023]
Abstract
In treatment of hypoxic tumors, oxygen-dependent photodynamic therapy (PDT) is considerably limited. Herein, a new bimetallic and biphasic Rh-based core-shell nanosystem (Au@Rh-ICG-CM) is developed to address tumor hypoxia while achieving high PDT efficacy. Such porous Au@Rh core-shell nanostructures are expected to exhibit catalase-like activity to efficiently catalyze oxygen generation from endogenous hydrogen peroxide in tumors. Coating Au@Rh nanostructures with tumor cell membrane (CM) enables tumor targeting via homologous binding. As a result of the large pores of Rh shells and the trapping ability of CM, the photosensitizer indocyanine green (ICG) is successfully loaded and retained in the cavity of Au@Rh-CM. Au@Rh-ICG-CM shows good biocompatibility, high tumor accumulation, and superior fluorescence and photoacoustic imaging properties. Both in vitro and in vivo results demonstrate that Au@Rh-ICG-CM is able to effectively convert endogenous hydrogen peroxide into oxygen and then elevate the production of tumor-toxic singlet oxygen to significantly enhance PDT. As noted, the mild photothermal effect of Au@Rh-ICG-CM also improves PDT efficacy. By integrating the superiorities of hypoxia regulation function, tumor accumulation capacity, bimodal imaging, and moderate photothermal effect into a single nanosystem, Au@Rh-ICG-CM can readily serve as a promising nanoplatform for enhanced cancer PDT.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Wei Hu
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Yuhao Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Tsengming Chou
- Laboratory for Multiscale Imaging, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Qiang Zhang
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian, 361005, P. R. China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian, 361005, P. R. China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| |
Collapse
|
308
|
Guo J, Huang L. Membrane-core nanoparticles for cancer nanomedicine. Adv Drug Deliv Rev 2020; 156:23-39. [PMID: 32450105 DOI: 10.1016/j.addr.2020.05.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
Cancer is one of the most severe disease burdens in modern times, with an estimated increase in the number of patients diagnosed globally from 18.1 million in 2018 to 23.6 million in 2030. Despite a significant progress achieved by conventional therapies, they have limitations and are still far from ideal. Therefore, safe, effective and widely-applicable treatments are urgently needed. Over the past decades, the development of novel delivery approaches based on membrane-core (MC) nanostructures for transporting chemotherapeutics, nucleic acids and immunomodulators has significantly improved anticancer efficacy and reduced side effects. In this review, the formulation strategies based on MC nanostructures for delivery of anticancer drug are described, and recent advances in the application of MC nanoformulations to overcome the delivery hurdles for clinical translation are discussed.
Collapse
|
309
|
Meng T, Jiang R, Wang S, Li J, Zhang F, Lee JH, Jiang J, Zhu M. Stem Cell Membrane-Coated Au-Ag-PDA Nanoparticle-Guided Photothermal Acne Therapy. Colloids Surf B Biointerfaces 2020; 192:111145. [PMID: 32480049 DOI: 10.1016/j.colsurfb.2020.111145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/25/2020] [Accepted: 05/18/2020] [Indexed: 01/29/2023]
Abstract
The polydopamine coating on Au-Ag nanoparticles (Au-Ag-PDA) possess excellent photothermal conversion efficiency after absorbing near-infrared laser light. After the stem cell membrane (STCM) encapsulates Au-Ag-PDA (Au-Ag-PDA@STCM), the nanoparticles (NPs) exhibit less cytotoxicity, and further optimizing their efficiency in photothermal therapy. The photothermal activity of Au-Ag-PDA@STCM has not yet been reported. Therefore, in this study, the sebaceous gland cell line SZ95 and the golden hamsters were used to observe the photothermal effects of the Au-Ag-PDA@STCM. SZ95 cells were treated with various concen-trations of Au-Ag-PDA@STCM NPs. The photothermal effect on cell proliferation was analyzed after irradiating the cells with a 808 nm laser. After laser treatment of golden hamsters, the flank organs were observed at 4 different time points. Histological analysis was performed to observe tissue damage. The results suggest that Au-Ag-PDA@STCM NPs significantly inhibited the proliferation of sebaceous gland cells in vitro, and reduced the size of sebaceous glands and sebum secretion in vivo. Therefore, NPs can be used to treat acne by thermally injuring sebaceous gland cells.
Collapse
Affiliation(s)
- Tianqi Meng
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Rihua Jiang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Shiyi Wang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jing Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Fuqiang Zhang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jeung-Hoon Lee
- Department of Dermatology, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| | - Mingji Zhu
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
310
|
Zhao X, Tang D, Wu Y, Chen S, Wang C. An artificial cell system for biocompatible gene delivery in cancer therapy. NANOSCALE 2020; 12:10189-10195. [PMID: 32355942 DOI: 10.1039/c9nr09131a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The gene therapy of cancer is generally recognized as a promising approach for the reversal of neoplastic progress of cancer cells. However, the lack of biocompatibility as well as highly available carriers is the bottleneck in gene therapy. In recent years, with the advances of understanding in cell signaling and cell based functions, the development of cell mimic carriers is showing great potential in evaluating the anticancer efficacy of drugs. Here in our study, an artificial cell (AC) system was fabricated to mimic the gene protection and transfection functions of cancer cells using cancer cell derived histone and membranes. As expected, the AC showed high biocompatibility as well as preferable gene transfection capability both in vitro and in vivo, and might be a promising tool for the flexible assembly of cell mimic systems in cancer therapy.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacy, Xinxiang Central Hospital, Xinxiang, Henan, P. R. China
| | | | | | | | | |
Collapse
|
311
|
Zhang L, Deng S, Zhang Y, Peng Q, Li H, Wang P, Fu X, Lei X, Qin A, Yu X. Homotypic Targeting Delivery of siRNA with Artificial Cancer Cells. Adv Healthc Mater 2020; 9:e1900772. [PMID: 32181988 DOI: 10.1002/adhm.201900772] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 01/27/2020] [Accepted: 02/25/2020] [Indexed: 01/10/2023]
Abstract
The camouflage with cell membrane bestows nanoparticles with cell-like functions, such as specific recognition, long blood circulation, and immune escaping. For cancer therapy, the nanoparticles camouflaged with cancer cell membrane (CCM) from homologous cells show homotypic targeting delivery of small molecule compounds, photosensitizers, or enzymes to the tumors. However, effective gene therapy encounters difficulties by this approach due to the properties of nucleic acids. Herein, a cancer cell-like gene delivery system is developed using an excellent polymer poly(β-amino ester) (PBAE) to condense small interfering RNA (siRNA) (targeting to Plk1 gene) into nanoparticles (PBAE/siPlk1) as the core, which is further camouflaged with CCM. These novel biomimetic nanoparticles CCM/PBAE/siPlk1 (CCMPP) demonstrate highly specific targeting to homotypic cancer cells, effective downregulation of PLK1 level, and inducing apoptosis of cancer cells. Based on the homotypic binding adhesion molecules on the CCM, the cellular internalization and homotypic-targeting accumulation to the tumors are clearly improved. CCMPP induces highly efficient apoptosis of cancer cells both in vitro and in vivo and results in significant tumor inhibition. The artificial cancer cells with homotypic properties can serve as a biomimetic delivery system for cancer-targeted gene therapy.
Collapse
Affiliation(s)
- Lingmin Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Sai Deng
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Yanfen Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Qingsheng Peng
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Huan Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Ping Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Xiaomei Fu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Xueping Lei
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Aiping Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical University Guangzhou Guangdong 511436 China
| |
Collapse
|
312
|
Vaughan HJ, Green JJ, Tzeng SY. Cancer-Targeting Nanoparticles for Combinatorial Nucleic Acid Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901081. [PMID: 31222852 PMCID: PMC6923623 DOI: 10.1002/adma.201901081] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/13/2019] [Indexed: 05/03/2023]
Abstract
Nucleic acids are a promising type of therapeutic for the treatment of a wide range of conditions, including cancer, but they also pose many delivery challenges. For efficient and safe delivery to cancer cells, nucleic acids must generally be packaged into a vehicle, such as a nanoparticle, that will allow them to be taken up by the target cells and then released in the appropriate cellular compartment to function. As with other types of therapeutics, delivery vehicles for nucleic acids must also be designed to avoid unwanted side effects; thus, the ability of such carriers to target their cargo to cancer cells is crucial. Classes of nucleic acids, hurdles that must be overcome for effective intracellular delivery, types of nonviral nanomaterials used as delivery vehicles, and the different strategies that can be employed to target nucleic acid delivery specifically to tumor cells are discussed. Additonally, nanoparticle designs that facilitate multiplexed delivery of combinations of nucleic acids are reviewed.
Collapse
Affiliation(s)
- Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Translational Tissue Engineering Center and Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, 400 North Broadway, Smith Building 5001, Baltimore, MD, 21231, USA
| |
Collapse
|
313
|
Ding X, Yin C, Zhang W, Sun Y, Zhang Z, Yang E, Sun D, Wang W. Designing Aptamer-Gold Nanoparticle-Loaded pH-Sensitive Liposomes Encapsulate Morin for Treating Cancer. NANOSCALE RESEARCH LETTERS 2020; 15:68. [PMID: 32232589 PMCID: PMC7105578 DOI: 10.1186/s11671-020-03297-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/11/2020] [Indexed: 05/10/2023]
Abstract
This study proposes the synthesis of a type of anticancer nanoparticle, aptamers and Au nanoparticle (Apt-Au)-modified Morin pH-sensitive liposome (MSL), which exhibits targeting properties. Tumors are difficult to cure because their microenvironment varies from that of normal tissue; its pH is lower than that of normal tissue, which generally impedes the effectiveness of drugs. Thus, pH-responsive drugs have attracted extensive attention. Gold nanoparticles (AuNPs) show potential as drug carriers because of their small size, good biocompatibility, easy surface modification, and strong cell penetration. Apt-Au@MSL exhibits excellent monodispersity and tumor-targeting properties and can be released in partly acidic environment via dialysis. We screened our model cancer cell by MTT assay and found that SGC-7901 cells can effectively suppress proliferation. In vivo results demonstrate that the administration of Apt-Au@MSL could inhibit tumor growth in xenograft mouse models. H&E staining and TUNEL assay further confirmed that Apt-Au@MSL can promote tumor apoptosis. Apt-Au@MSL may induce apoptosis by triggering overproduction of reactive oxygen species (ROS) and regulating multiple signal crosstalk. Both blood biochemistry tests and H&E staining suggested that these materials exhibit negligible acute toxicity and good biocompatibility in vivo. With its powerful function, Apt-Au@MSL can be used as a target-based anticancer material for future clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyuan Ding
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Chenyang Yin
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Weiwei Zhang
- School of Biochemical Engineering, Anhui Polytechnic University, 8 Zheshan Road, Wuhu, 241000, Anhui, China
| | - Yu Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Zhenzhen Zhang
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Endong Yang
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China
| | - Dongdong Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China.
| | - Weiyun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
314
|
Cao X, Tan T, Zhu D, Yu H, Liu Y, Zhou H, Jin Y, Xia Q. Paclitaxel-Loaded Macrophage Membrane Camouflaged Albumin Nanoparticles for Targeted Cancer Therapy. Int J Nanomedicine 2020; 15:1915-1928. [PMID: 32256068 PMCID: PMC7090179 DOI: 10.2147/ijn.s244849] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Melanoma is the most common symptom of aggressive skin cancer, and it has become a serious health concern worldwide in recent years. The metastasis rate of malignant melanoma remains high, and it is highly difficult to cure with the currently available treatment options. Effective yet safe therapeutic options are still lacking. Alternative treatment options are in great demand to improve the therapeutic outcome against advanced melanoma. This study aimed to develop albumin nanoparticles (ANPs) coated with macrophage plasma membranes (RANPs) loaded with paclitaxel (PTX) to achieve targeted therapy against malignant melanoma. METHODS Membrane derivations were achieved by using a combination of hypotonic lysis, mechanical membrane fragmentation, and differential centrifugation to empty the harvested cells of their intracellular contents. The collected membrane was then physically extruded through a 400 nm porous polycarbonate membrane to form macrophage cell membrane vesicles. Albumin nanoparticles were prepared through a well-studied nanoprecipitation process. At last, the two components were then coextruded through a 200 nm porous polycarbonate membrane. RESULTS Using paclitaxel as the model drug, PTX-loaded RANPs displayed significantly enhanced cytotoxicity and apoptosis rates compared to albumin nanoparticles without membrane coating in the murine melanoma cell line B16F10. RANPs also exhibited significantly higher internalization efficiency in B16F10 cells than albumin nanoparticles without a membrane coating. Next, a B16F10 tumor xenograft mouse model was established to explore the biodistribution profiles of RANPs, which showed prolonged blood circulation and selective accumulation at the tumor site. PTX-loaded RANPs also demonstrated greatly improved antitumor efficacy in B16F10 tumor-bearing mouse xenografts. CONCLUSION Albumin-based nanoscale delivery systems coated with macrophage plasma membranes offer a highly promising approach to achieve tumor-targeted therapy following systemic administration.
Collapse
Affiliation(s)
- Xi Cao
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Tingfei Tan
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Dongchun Zhu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Haixia Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Yaru Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Haiyun Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Yong Jin
- Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, People’s Republic of China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| |
Collapse
|
315
|
Xu CH, Ye PJ, Zhou YC, He DX, Wei H, Yu CY. Cell membrane-camouflaged nanoparticles as drug carriers for cancer therapy. Acta Biomater 2020; 105:1-14. [PMID: 32001369 DOI: 10.1016/j.actbio.2020.01.036] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/18/2019] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
The translocation of natural cell membranes to the surface of synthetic nanoparticles, which allows man-made vectors to share merits and functionalities created by nature, has been a hot subject of research in the past decade. The resulting biomimetic nanoparticles not only retain the physicochemical properties of nanomaterials, but also inherit the advantageous functions of source cells. Combined with the preponderances of both synthetic and natural platforms, the optimized biomimetic systems can maximize the drug delivery efficiency. In this review, we first summarize the preparation strategies of the biomimetic systems from the perspective of the correlation between the properties of nanoparticles and cell membranes. Six types of cell membrane-camouflaged nanoparticles are further introduced with an emphasis on their properties and performance. Finally, a concluding remark regarding the primary challenges and opportunities associated with these nanoparticles is presented. STATEMENT OF SIGNIFICANCE: Translocation of natural cell membranes to the surface of synthetic nanoparticles has been repeatedly highlighted in the past decade to endow man-made vectors with merits and functionalities created by nature; therefore, the resulting biomimetic systems not only retain the physicochemical properties of nanomaterials but also inherit the biological functions of source cells for efficient drug delivery. To provide a timely review on this hot and rapidly developing subject of research, this paper summarized recent progress on the cell membrane-camouflaged nanoparticles as drug carriers for cancer therapy, and focused primarily on six different types of cell membrane-coated nanoparticles with an emphasis on the preparation strategies from the perspective of the correlation between the properties of nanoparticles and cell membrane.
Collapse
Affiliation(s)
- Cheng-Hui Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Peng-Ju Ye
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Yang-Chun Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Dong-Xiu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| |
Collapse
|
316
|
Han Z, Lv W, Li Y, Chang J, Zhang W, Liu C, Sun J. Improving Tumor Targeting of Exosomal Membrane-Coated Polymeric Nanoparticles by Conjugation with Aptamers. ACS APPLIED BIO MATERIALS 2020; 3:2666-2673. [DOI: 10.1021/acsabm.0c00181] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ziwei Han
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100149, China
| | - Wenxing Lv
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yike Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100149, China
| | - Jianqiao Chang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Wei Zhang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100149, China
| | - Chao Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100149, China
| | - Jiashu Sun
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100149, China
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, School of Chemistry and Molecular Engineering, Shanghai 200062, China
| |
Collapse
|
317
|
Guo J, Zeng H, Chen Y. Emerging Nano Drug Delivery Systems Targeting Cancer-Associated Fibroblasts for Improved Antitumor Effect and Tumor Drug Penetration. Mol Pharm 2020; 17:1028-1048. [PMID: 32150417 DOI: 10.1021/acs.molpharmaceut.0c00014] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jian Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Huating Zeng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
318
|
Tanita K, Koseki Y, Kamishima T, Kasai H. Tropone Skeleton Enhances the Dispersion Stability of Nano-prodrugs. CHEM LETT 2020. [DOI: 10.1246/cl.190876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Keita Tanita
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Yoshitaka Koseki
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Takaaki Kamishima
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| | - Hitoshi Kasai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8577, Japan
| |
Collapse
|
319
|
Nie D, Dai Z, Li J, Yang Y, Xi Z, Wang J, Zhang W, Qian K, Guo S, Zhu C, Wang R, Li Y, Yu M, Zhang X, Shi X, Gan Y. Cancer-Cell-Membrane-Coated Nanoparticles with a Yolk-Shell Structure Augment Cancer Chemotherapy. NANO LETTERS 2020; 20:936-946. [PMID: 31671946 DOI: 10.1021/acs.nanolett.9b03817] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite rapid advancements in antitumor drug delivery, insufficient intracellular transport and subcellular drug accumulation are still issues to be addressed. Cancer cell membrane (CCM)-camouflaged nanoparticles (NPs) have shown promising potential in tumor therapy due to their immune escape and homotypic binding capacities. However, their efficacy is still limited due to inefficient tumor penetration and compromised intracellular transportation. Herein, a yolk-shell NP with a mesoporous silica nanoparticle (MSN)-supported PEGylated liposome yolk and CCM coating, CCM@LM, was developed for chemotherapy and exhibited a homologous tumor-targeting effect. The yolk-shell structure endowed CCM@LM with moderate rigidity, which might contribute to the frequent transformation into an ellipsoidal shape during infiltration, leading to facilitated penetration throughout multicellular spheroids in vitro (up to a 23.3-fold increase compared to the penetration of membrane vesicles). CCM@LM also exhibited a cellular invasion profile mimicking an enveloped virus invasion profile. CCM@LM was directly internalized by membrane fusion, and the PEGylated yolk (LM) was subsequently released into the cytosol, indicating the execution of an internalization pathway similar to that of an enveloped virus. The incoming PEGylated LM further underwent efficient trafficking throughout the cytoskeletal filament network, leading to enhanced perinuclear aggregation. Ultimately, CCM@LM, which co-encapsulated low-dose doxorubicin and the poly(ADP-ribose) polymerase inhibitor, mefuparib hydrochloride, exhibited a significantly stronger antitumor effect than the first-line chemotherapeutic drug Doxil. Our findings highlight that NPs that can undergo facilitated tumor penetration and robust intracellular trafficking have a promising future in cancer chemotherapy.
Collapse
Affiliation(s)
- Di Nie
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Zhuo Dai
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Jialin Li
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Yiwei Yang
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Ziyue Xi
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Jie Wang
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Wei Zhang
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- School of Pharmacy , Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Kun Qian
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Shiyan Guo
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
| | - Chunliu Zhu
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
| | - Rui Wang
- School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Yiming Li
- School of Pharmacy , Shanghai University of Traditional Chinese Medicine , Shanghai 201203 , China
| | - Miaorong Yu
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xinxin Zhang
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xinghua Shi
- University of Chinese Academy of Sciences , Beijing 100049 , China
- CAS Key Laboratory for Nanosystem and Hierarchy Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Chinese Academy of Sciences , Beijing 100190 , China
| | - Yong Gan
- Shanghai Institute of Materia Medica , Chinese Academy of Sciences , Shanghai 201203 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
320
|
Jin J, Bhujwalla ZM. Biomimetic Nanoparticles Camouflaged in Cancer Cell Membranes and Their Applications in Cancer Theranostics. Front Oncol 2020; 9:1560. [PMID: 32039028 PMCID: PMC6985278 DOI: 10.3389/fonc.2019.01560] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/23/2019] [Indexed: 01/10/2023] Open
Abstract
Nanoparticles (NPs) camouflaged in cell membranes represent novel biomimetic platforms that can mimic some of the membrane functions of the cells from which these membranes are derived, in biological systems. Studies using cell membrane coated NPs cover a large repertoire of membranes derived from cells such as red blood cells, immune cells, macrophages, and cancer cells. Cancer cell membrane coated nanoparticles (CCMCNPs) typically consist of a NP core with a cancer cell plasma membrane coat that can carry tumor-specific receptors and antigens for cancer targeting. The NP core can serve as a vehicle to carry imaging and therapeutic moieties. As a result, these CCMCNPs are being investigated for multiple purposes including cancer theranostics. Here we have discussed the key steps and major issues in the synthesis and characterization of CCMCNPs. We have highlighted the homologous binding mechanisms of CCMCNPs that are being investigated for cancer targeting, and have presented our data that identify BT474 CCMCNPs as binding to multiple cancer cell lines. Current preclinical applications of CCMCNPs for cancer theranostics and their advantages and limitations are discussed.
Collapse
Affiliation(s)
- Jiefu Jin
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States.,Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
321
|
Ma W, Zhu D, Li J, Chen X, Xie W, Jiang X, Wu L, Wang G, Xiao Y, Liu Z, Wang F, Li A, Shao D, Dong W, Liu W, Yuan Y. Coating biomimetic nanoparticles with chimeric antigen receptor T cell-membrane provides high specificity for hepatocellular carcinoma photothermal therapy treatment. Theranostics 2020; 10:1281-1295. [PMID: 31938065 PMCID: PMC6956810 DOI: 10.7150/thno.40291] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Rationale: Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies in the world. Apart from traditional surgical resection, radiotherapy, and chemotherapy, more recent techniques such as nano-photothermal therapy and biotherapy are gradually being adopted for the treatment of HCC. This project intends to combine the advantages of nanoscale drug delivery systems with the targeting ability of CAR-T cells. Method: Based on cell membrane-coated nanoparticles and cell membrane-targeting modifications, a novel nanomaterial was prepared by coating CAR-T cell membranes specifically recognizing GPC3+ HCC cells onto mesoporous silica containing IR780 nanoparticles. Subsequently, the physical properties were characterized, and the in vitro and in vivo targeting abilities of this nanoparticle were verified. Results: CAR-T cells were constructed which could recognize GPC3 expressed on the cell surface of HCC cells. Then the isolated CAR-T cell membrane was successfully coated on the IR780 loaded mesoporous silica materials, as verified by transmission electron microscopy. The superior targeting ability of CAR-T cell membrane coated nanoparticles compared to IR780 loaded mesoporous silica nanoparticles was verified, both in vitro and in vivo. Conclusion: This new nanomaterial exhibits photothermal antitumor abilities along with enhanced targeting abilities, suggesting a promising strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Daoming Zhu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Jinghua Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xi Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Xie
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Xiang Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Long Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ganggang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yusha Xiao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhisu Liu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Andrew Li
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| | - Dan Shao
- Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wenfei Dong
- Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
322
|
He Z, Zhang Y, Feng N. Cell membrane-coated nanosized active targeted drug delivery systems homing to tumor cells: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 106:110298. [DOI: 10.1016/j.msec.2019.110298] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 09/08/2019] [Accepted: 10/07/2019] [Indexed: 01/14/2023]
|
323
|
Li Y, Gan Y, Li C, Yang YY, Yuan P, Ding X. Cell membrane-engineered hybrid soft nanocomposites for biomedical applications. J Mater Chem B 2020; 8:5578-5596. [DOI: 10.1039/d0tb00472c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
An overview of various cell membrane-engineered hybrid soft nanocomposites for medical applications.
Collapse
Affiliation(s)
- Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen 518107
- China
| | - Yingying Gan
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen 518107
- China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen 518107
- China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology
- Singapore 138669
- Singapore
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen 518107
- China
| | - Xin Ding
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen 518107
- China
| |
Collapse
|
324
|
Jiang L, Lee HW, Loo SCJ. Therapeutic lipid-coated hybrid nanoparticles against bacterial infections. RSC Adv 2020; 10:8497-8517. [PMID: 35497832 PMCID: PMC9050015 DOI: 10.1039/c9ra10921h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022] Open
Abstract
One of the most important health concerns in society is the development of pathogen-causing nosocomial infections. Since the first discovery of antibiotics, bacterial infections have been highly treatable. However, with evolution and the nondiscretionary usage of antibiotics, pathogens have also found new ways to survive the onslaught of antibiotics by surviving intracellularly or through the formation of obstinate biofilms, and through these, the outcomes of regular antibiotic treatments may now be unsatisfactory. Lipid-coated hybrid nanoparticles (LCHNPs) are the next-generation core–shell structured nanodelivery system, where an inorganic or organic core, loaded with antimicrobials, is enveloped by lipid layers. This core–shell structure, with multifarious decorations, not only improves the loading capabilities of therapeutics but also has the potential to improve therapeutic delivery, especially for targeting biofilm-based and intracellular bacterial infections. Although there has been significant interest in the development of LCHNPs, they have yet to be widely exploited for bacterial infections. In this review, we will provide an overview on the latest development of LCHNPs and the various approaches in synthesizing this nano-delivery system. In addition, a discussion on future perspectives of LCHNPs, in combination with other novel anti-bacterial technologies, will be provided towards the end of this review. Lipid-coated hybrid nanoparticles are next-generation core–shell structured nanodelivery systems, which improve the loading capabilities of therapeutics and can improve therapeutic delivery, especially for targeting biofilm-based and intracellular bacterial infections.![]()
Collapse
Affiliation(s)
- Lai Jiang
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore
| | - Hiang Wee Lee
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore
| | - Say Chye Joachim Loo
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore
- Singapore Centre for Environmental Life Sciences Engineering
- Nanyang Technological University
| |
Collapse
|
325
|
Choi B, Park W, Park SB, Rhim WK, Han DK. Recent trends in cell membrane-cloaked nanoparticles for therapeutic applications. Methods 2019; 177:2-14. [PMID: 31874237 DOI: 10.1016/j.ymeth.2019.12.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022] Open
Abstract
Synthetic nanoparticles are extensively utilized in various biomedical engineering fields because of their unique physicochemical properties. However, their exogenous characteristics result in synthetic nanosystem invaders that easily induce the passive immune clearance mechanism, thereby increasing the retention effect caused by reticuloendothelial system (RES), resulting in low therapeutic efficacy and toxic effects. Recently, a cell membrane cloaking has been emerging technique as a novel interfacing approach from the biological/immunological perspective. This has been considered as useful technique for improving the performance of synthetic nanocarriers in vivo. By cell membrane cloaking, nanoparticles acquire the biological functions of natural cell membranes due to the presence of membrane-anchored proteins, antigens, and immunological moieties as well as physicochemical property of natural cell membrane. Due to cell membrane cloaking, the derived biological properties and functions of nanoparticles such as their immunosuppressive capability, long circulation time, and disease targeting ability have enhanced their future potential in biomedicine. Here, we review the cell membrane-cloaked nanosystems, highlight their novelty, introduce the preparation and characterization methods with relevant biomedical applications, and describe the prospects for using this novel biomimetic system that was developed from a combination of cell membranes and synthetic nanomaterials.
Collapse
Affiliation(s)
- Bogyu Choi
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Wooram Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Sung-Bin Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea.
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea.
| |
Collapse
|
326
|
Xiao T, Li D, Shi X, Shen M. PAMAM Dendrimer‐Based Nanodevices for Nuclear Medicine Applications. Macromol Biosci 2019; 20:e1900282. [DOI: 10.1002/mabi.201900282] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/03/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Tingting Xiao
- Key Laboratory of Science & Technology of Eco‐TextileMinistry of EducationCollege of ChemistryChemical Engineering and BiotechnologyDonghua University Shanghai 201620 P. R. China
| | - Du Li
- Key Laboratory of Science & Technology of Eco‐TextileMinistry of EducationCollege of ChemistryChemical Engineering and BiotechnologyDonghua University Shanghai 201620 P. R. China
| | - Xiangyang Shi
- Key Laboratory of Science & Technology of Eco‐TextileMinistry of EducationCollege of ChemistryChemical Engineering and BiotechnologyDonghua University Shanghai 201620 P. R. China
| | - Mingwu Shen
- Key Laboratory of Science & Technology of Eco‐TextileMinistry of EducationCollege of ChemistryChemical Engineering and BiotechnologyDonghua University Shanghai 201620 P. R. China
| |
Collapse
|
327
|
Harris JC, Scully MA, Day ES. Cancer Cell Membrane-Coated Nanoparticles for Cancer Management. Cancers (Basel) 2019; 11:E1836. [PMID: 31766360 PMCID: PMC6966582 DOI: 10.3390/cancers11121836] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer is a global health problem in need of transformative treatment solutions for improved patient outcomes. Many conventional treatments prove ineffective and produce undesirable side effects because they are incapable of targeting only cancer cells within tumors and metastases post administration. There is a desperate need for targeted therapies that can maximize treatment success and minimize toxicity. Nanoparticles (NPs) with tunable physicochemical properties have potential to meet the need for high precision cancer therapies. At the forefront of nanomedicine is biomimetic nanotechnology, which hides NPs from the immune system and provides superior targeting capabilities by cloaking NPs in cell-derived membranes. Cancer cell membranes expressing "markers of self" and "self-recognition molecules" can be removed from cancer cells and wrapped around a variety of NPs, providing homotypic targeting and circumventing the challenge of synthetically replicating natural cell surfaces. Compared to unwrapped NPs, cancer cell membrane-wrapped NPs (CCNPs) provide reduced accumulation in healthy tissues and higher accumulation in tumors and metastases. The unique biointerfacing capabilities of CCNPs enable their use as targeted nanovehicles for enhanced drug delivery, localized phototherapy, intensified imaging, or more potent immunotherapy. This review summarizes the state-of-the-art in CCNP technology and provides insight to the path forward for clinical implementation.
Collapse
Affiliation(s)
- Jenna C. Harris
- Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA;
| | | | - Emily S. Day
- Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA;
- Biomedical Engineering, University of Delaware, Newark, DE 19716, USA;
- Helen F. Graham Cancer Center and Research Institute, Newark, DE 19713, USA
| |
Collapse
|
328
|
Liu Y, Luo J, Chen X, Liu W, Chen T. Cell Membrane Coating Technology: A Promising Strategy for Biomedical Applications. NANO-MICRO LETTERS 2019; 11:100. [PMID: 34138027 PMCID: PMC7770915 DOI: 10.1007/s40820-019-0330-9] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/14/2019] [Indexed: 05/02/2023]
Abstract
Cell membrane coating technology is an approach to the biomimetic replication of cell membrane properties, and is an active area of ongoing research readily applicable to nanoscale biomedicine. Nanoparticles (NPs) coated with cell membranes offer an opportunity to unite natural cell membrane properties with those of the artificial inner core material. The coated NPs not only increase their biocompatibility but also achieve effective and extended circulation in vivo, allowing for the execution of targeted functions. Although cell membrane-coated NPs offer clear advantages, much work remains before they can be applied in clinical practice. In this review, we first provide a comprehensive overview of the theory of cell membrane coating technology, followed by a summary of the existing preparation and characterization techniques. Next, we focus on the functions and applications of various cell membrane types. In addition, we collate model drugs used in cell membrane coating technology, and review the patent applications related to this technology from the past 10 years. Finally, we survey future challenges and trends pertaining to this technology in an effort to provide a comprehensive overview of the future development of cell membrane coating technology.
Collapse
Affiliation(s)
- Yao Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Jingshan Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, People's Republic of China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| |
Collapse
|
329
|
Liu C, Zhang W, Li Y, Chang J, Tian F, Zhao F, Ma Y, Sun J. Microfluidic Sonication To Assemble Exosome Membrane-Coated Nanoparticles for Immune Evasion-Mediated Targeting. NANO LETTERS 2019; 19:7836-7844. [PMID: 31597431 DOI: 10.1021/acs.nanolett.9b02841] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Using natural membranes to coat nanoparticles (NPs) provides an efficient means to reduce the immune clearance of NPs and improve their tumor-specific targeting. However, fabrication of these drug-loaded biomimetic NPs, such as exosome membrane (EM)- or cancer cell membrane (CCM)-coated poly(lactic-co-glycolic acid) (PLGA) NPs, remains a challenging task owing to the heterogeneous nature of biomembranes and labor-intensive procedures. Herein, we report a microfluidic sonication approach to produce EM-, CCM-, and lipid-coated PLGA NPs encapsulated with imaging agents in a one-step and straightforward manner. Tumor cell-derived EM-coated PLGA NPs consisting of both endosomal and plasma membrane proteins show superior homotypic targeting as compared to CCM-PLGA NPs of similar sizes and core-shell structures in both in vitro and in vivo models. The underlying mechanism is associated with a significantly reduced uptake of EM-PLGA NPs by macrophages and peripheral blood monocytes, revealing an immune evasion-mediated targeting of EM-PLGA NPs to homologous tumors. Overall, this work illustrates the promise of using microfluidic sonication approach to fabricate biomimetic NPs for better biocompatibility and targeting efficacy.
Collapse
Affiliation(s)
- Chao Liu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100149 , China
| | - Wei Zhang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100149 , China
| | - Yike Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100149 , China
| | - Jianqiao Chang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
| | - Fei Tian
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100149 , China
| | - Fanghao Zhao
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
| | - Yao Ma
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100149 , China
| | - Jiashu Sun
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100149 , China
| |
Collapse
|
330
|
Wu P, Yin D, Liu J, Zhou H, Guo M, Liu J, Liu Y, Wang X, Liu Y, Chen C. Cell membrane based biomimetic nanocomposites for targeted therapy of drug resistant EGFR-mutated lung cancer. NANOSCALE 2019; 11:19520-19528. [PMID: 31573595 DOI: 10.1039/c9nr05791a] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The therapeutic efficacy of anti-cancer nanomedicines is generally constrained due to limited accumulation in the solid tumors. In this study, we developed a biomimetic nano-carrier to enhance the chemo-therapeutic efficacy of doxorubicin and icotinib in a chemo-resistant non-small cell lung cancer (NSCLC) cell line harboring a mutation in the epidermal growth factor receptor (EGFR). The unique nanomedicine was prepared by coating with targeting cancer cell membrane proteins as highly specific ligands. The resulting biomimetic nanoparticles were highly stable and exhibited superior homologous targeting ability in vitro compared with control groups. In a mouse EGFR-mutated NSCLC xenograft model, intravenous injection of the biomimetic nanomedicine led to a high tumour inhibition rate (87.56%). Histopathological analysis demonstrated that the biomimetic nanomedicine had minimal side effects. Taken together, a cancer cell membrane-based biomimetic drug carrier can significantly enhance drug accumulation and improve therapeutic efficacy in cancers.
Collapse
Affiliation(s)
- Pengying Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China. and Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Dongtao Yin
- Department of Thoracic Surgery, General Hospital of the Chinese People's Liberation Army, Beijing, 100853, China and Department of Thoracic Surgery, Rocket Force Characteristic Medical Center of the Chinese People's Liberation Army, Beijing, 100088, China
| | - Jiaming Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China. and The College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yang Liu
- Department of Thoracic Surgery, General Hospital of the Chinese People's Liberation Army, Beijing, 100853, China
| | - Xiaobing Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
331
|
Wang Z, Zhang F, Shao D, Chang Z, Wang L, Hu H, Zheng X, Li X, Chen F, Tu Z, Li M, Sun W, Chen L, Dong W. Janus Nanobullets Combine Photodynamic Therapy and Magnetic Hyperthermia to Potentiate Synergetic Anti-Metastatic Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901690. [PMID: 31763151 PMCID: PMC6864517 DOI: 10.1002/advs.201901690] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/16/2019] [Indexed: 05/08/2023]
Abstract
Photodynamic therapy (PDT) is clinically promising in destructing primary tumors but ineffective against distant metastases. This study reports the use of immunogenic nanoparticles mediated combination of PDT and magnetic hyperthermia to synergistically augment the anti-metastatic efficacy of immunotherapy. Janus nanobullets integrating chlorine e6 (Ce6) loaded, disulfide-bridged mesoporous organosilica bodies with magnetic heads (M-MONs@Ce6) are tailored for redox/pH-triggered photosensitizer release accompanying their matrix degradation. Cancer cell membrane cloaking enables favorable tumor-targeted accumulation and prolonged blood circulation time of M-MONs@Ce6. The combination of PDT and magnetic hyperthermia has a strong synergy anticancer activity and simultaneously elicits a sequence of immunogenic cell death, resulting in synergistically tumor-specific immune responses. When combined with anti-CTLA-4 antibody, the biomimetic and biodegradable nanoparticle enables the notable eradication of primary and deeply metastatic tumors with low systematic toxicity, thus potentially advancing the development of combined hyperthermia, PDT, and checkpoint blockade immunotherapy to combat cancer metastasis.
Collapse
Affiliation(s)
- Zheng Wang
- CAS Key Laboratory of Bio Medical DiagnosticsSuzhou Institute of BiomedicalEngineering and TechnologyChinese Academy of SciencesSuzhou215163China
| | - Fan Zhang
- CAS Key Laboratory of Bio Medical DiagnosticsSuzhou Institute of BiomedicalEngineering and TechnologyChinese Academy of SciencesSuzhou215163China
- Department of PharmacologyNanomedicine Engineering Laboratory of Jilin ProvinceCollege of Basic Medical SciencesJilin UniversityChangchun130021China
| | - Dan Shao
- CAS Key Laboratory of Bio Medical DiagnosticsSuzhou Institute of BiomedicalEngineering and TechnologyChinese Academy of SciencesSuzhou215163China
- Department of PharmacologyNanomedicine Engineering Laboratory of Jilin ProvinceCollege of Basic Medical SciencesJilin UniversityChangchun130021China
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Zhimin Chang
- CAS Key Laboratory of Bio Medical DiagnosticsSuzhou Institute of BiomedicalEngineering and TechnologyChinese Academy of SciencesSuzhou215163China
| | - Lei Wang
- State Key Laboratory of Fine ChemicalsDalian University of TechnologyDalian116024China
| | - Hanze Hu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Xiao Zheng
- Department of PharmacologyNanomedicine Engineering Laboratory of Jilin ProvinceCollege of Basic Medical SciencesJilin UniversityChangchun130021China
| | - Xuezhao Li
- State Key Laboratory of Fine ChemicalsDalian University of TechnologyDalian116024China
| | - Fangman Chen
- CAS Key Laboratory of Bio Medical DiagnosticsSuzhou Institute of BiomedicalEngineering and TechnologyChinese Academy of SciencesSuzhou215163China
| | - Zhaoxu Tu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Mingqiang Li
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Wen Sun
- State Key Laboratory of Fine ChemicalsDalian University of TechnologyDalian116024China
| | - Li Chen
- Department of PharmacologyNanomedicine Engineering Laboratory of Jilin ProvinceCollege of Basic Medical SciencesJilin UniversityChangchun130021China
| | - Wen‐Fei Dong
- CAS Key Laboratory of Bio Medical DiagnosticsSuzhou Institute of BiomedicalEngineering and TechnologyChinese Academy of SciencesSuzhou215163China
| |
Collapse
|
332
|
Zhang D, Ye Z, Wei L, Luo H, Xiao L. Cell Membrane-Coated Porphyrin Metal-Organic Frameworks for Cancer Cell Targeting and O 2-Evolving Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:39594-39602. [PMID: 31577410 DOI: 10.1021/acsami.9b14084] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Photodynamic therapy (PDT) has attracted great attention as an alternative tumor treatment method. Unfortunately, it suffers from some limitations like poor targeting capability and insufficient therapeutic efficiency caused by tumor hypoxia. In this work, we introduce a novel O2-evolving PDT nanoparticle for homologous cancer cell targeting as well as dual-mode imaging [i.e., magnetic resonance imaging (MRI) and fluorescence imaging]. Specifically, the nanostructure consists of a MnO2 nanosheet-coated metal-organic framework core and cancer cell membrane shell (defined as CM-MMNPs). The MnO2 layer displays H+ and H2O2 responsiveness, which can produce O2 to enhance O2-mediated singlet oxygen (1O2) generation for PDT. Moreover, the resulted Mn2+ can also be used as an optimal MRI contrast agent. The introduction of cell membrane and membrane proteins endow the CM-MMNPs with good stability and integrity in the process of cellular endocytosis, as well as strong homologous cell-targeting ability. This multifunctional nanoparticle has the potential to overcome the hypoxia of cancer cells in PDT, and provides a new paradigm for tumor targeting, detection, and therapy, which is promising for biomedical applications in the future.
Collapse
Affiliation(s)
- Di Zhang
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering , Hunan Normal University , Changsha 410082 , China
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| | - Zhongju Ye
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| | - Lin Wei
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Key Laboratory of Phytochemical R&D of Hunan Province, College of Chemistry and Chemical Engineering , Hunan Normal University , Changsha 410082 , China
| | - Haibin Luo
- School of Pharmaceutical Science , Sun Yat-Sen University , Guangzhou 510006 , China
| | - Lehui Xiao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, College of Chemistry , Nankai University , Tianjin 300071 , China
| |
Collapse
|
333
|
Wang J, Wang Z, Zhong Y, Zou Y, Wang C, Wu H, Lee A, Yang W, Wang X, Liu Y, Zhang D, Yan J, Hao M, Zheng M, Chung R, Bai F, Shi B. Central metal-derived co-assembly of biomimetic GdTPP/ZnTPP porphyrin nanocomposites for enhanced dual-modal imaging-guided photodynamic therapy. Biomaterials 2019; 229:119576. [PMID: 31704467 DOI: 10.1016/j.biomaterials.2019.119576] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023]
Abstract
Dual-modal imaging guided photodynamic therapy (PDT) of multifunctional nanocomposites holds great promise for precision tumor theranostics. However, poor heterogeneous interfacial compatibility between functional components, low hydrophilicity and complicated preparation of nanocomposites remain major obstacles for further bioapplication. Herein, a facile central metal-derived co-assembly strategy is developed to effectively integrate gadolinium porphyrin (GdTPP) contrast agent and Zinc porphyrin (ZnTPP) photosensitizer into a homogeneous GdTPP/ZnTPP nanocomposites (GZNs). GZNs possesses the following advantages: (1) Greatly improved interfacial compatibility facilitated by incorporating two metalporphyrins with same group (phenyl-) and different central metal atoms (Zn and Gd) leading to higher yield (4.7-5 fold) than either monocomponent nanoparticles. (2) Poor dispersity of GdTPP nanoparticles is greatly improved after integrating with ZnTPP blocks. (3) The GZNs inherit excellent fluorescence imaging, high relaxation rate (8.18 mM-1 s-1) and singlet oxygen production from two raw metalporphyrins. After camouflaging with homotypic cancer cell membrane for immunologic escape, the HeLa membrane coated GZNs (mGZNs) show enhanced in vivo MR/FL imaging guided anti-tumor targeting efficiency of 80.6% for HeLa cells. Our new strategy using central metal-derived co-assembly of homogeneous building blocks greatly improves interfacial compatibility to achieve combined functions for visualized cancer theranostics.
Collapse
Affiliation(s)
- Jiefei Wang
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Zhongjie Wang
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Yong Zhong
- Key Laboratory for Special Functional Materials of Ministry of Education, National & Local Joint Engineering Research Center for High-efficiency Display and Lighting Technology, School of Materials Science and Engineering, Collaborative Innovation Center of Nano Functional Materials and Applications, Henan University, Kaifeng, 475004, China
| | - Yan Zou
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China; Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Chong Wang
- Luohe Central Hospital, Luohe, 462000, China
| | - Haigang Wu
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Albert Lee
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Weitao Yang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Xiao Wang
- Key Laboratory for Special Functional Materials of Ministry of Education, National & Local Joint Engineering Research Center for High-efficiency Display and Lighting Technology, School of Materials Science and Engineering, Collaborative Innovation Center of Nano Functional Materials and Applications, Henan University, Kaifeng, 475004, China
| | - Yanjie Liu
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Dongya Zhang
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Jiliang Yan
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Mingcong Hao
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Meng Zheng
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China
| | - Roger Chung
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Feng Bai
- Key Laboratory for Special Functional Materials of Ministry of Education, National & Local Joint Engineering Research Center for High-efficiency Display and Lighting Technology, School of Materials Science and Engineering, Collaborative Innovation Center of Nano Functional Materials and Applications, Henan University, Kaifeng, 475004, China.
| | - Bingyang Shi
- International Joint Center for Biomedical Innovation, School of Life Sciences Henan University, Kaifeng, 475004, China; Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
334
|
Safarov T, Kiran B, Bagirova M, Allahverdiyev AM, Abamor ES. An overview of nanotechnology-based treatment approaches against Helicobacter Pylori. Expert Rev Anti Infect Ther 2019; 17:829-840. [PMID: 31591930 DOI: 10.1080/14787210.2019.1677464] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tural Safarov
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| | - Bukre Kiran
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| | - Melahat Bagirova
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| | - Adil M Allahverdiyev
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| | - Emrah Sefik Abamor
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| |
Collapse
|
335
|
Yang MY, Zhao RR, Fang YF, Jiang JL, Yuan XT, Shao JW. Carrier-free nanodrug: A novel strategy of cancer diagnosis and synergistic therapy. Int J Pharm 2019; 570:118663. [DOI: 10.1016/j.ijpharm.2019.118663] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 01/08/2023]
|
336
|
Sun D, Chen J, Wang Y, Ji H, Peng R, Jin L, Wu W. Advances in refunctionalization of erythrocyte-based nanomedicine for enhancing cancer-targeted drug delivery. Theranostics 2019; 9:6885-6900. [PMID: 31660075 PMCID: PMC6815958 DOI: 10.7150/thno.36510] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer remains a daunting and cureless disease, which is responsible for one-sixth of human deaths worldwide. These mortality rates have been expected to rise in the future due to the side effects of conventional treatments (chemotherapy, radiotherapy, and surgery), which can be addressed by applying nanomedicine. In order to escape from biological barriers, such nanomedicine should be mimicked and designed to be stealthy while navigating in the bloodstream. To achieve this, scientists take advantage of erythrocytes (red blood cells; RBCs) as drug carriers and develop RBC membrane (RBCm) coating nanotechnology. Thanks to the significant advances in nanoengineering, various facile surface functionalization methods can be applied to arm RBCm with not only targeting moieties, but also imaging agents, therapeutic agents, and nanoparticles, which are useful for theranostic nanomedicine. This review focuses on refunctionalization of erythrocyte-based nanomedicine for enhancing cancer-targeted drug delivery.
Collapse
Affiliation(s)
- Da Sun
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
- Biomedical Collaborative Innovation Center of Zhejiang Province & Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, Zhejiang, 325035, China
| | - Jia Chen
- Sichuan Provincial Center for Mental Health, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Yuan Wang
- Chongqing Business Vocational College, Chongqing, 401331, China
| | - Hao Ji
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Renyi Peng
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Libo Jin
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
- Biomedical Collaborative Innovation Center of Zhejiang Province & Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, Zhejiang, 325035, China
| | - Wei Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| |
Collapse
|
337
|
Hu Q, Bu Y, Cao R, Zhang G, Xie X, Wang S. Stability Designs of Cell Membrane Cloaked Magnetic Carbon Nanotubes for Improved Life Span in Screening Drug Leads. Anal Chem 2019; 91:13062-13070. [DOI: 10.1021/acs.analchem.9b03268] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Qi Hu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi’an 710061, China
| | - Yusi Bu
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi’an 710061, China
| | - Ruiqi Cao
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi’an 710061, China
| | - Gao Zhang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi’an 710061, China
| | - Xiaoyu Xie
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi’an 710061, China
| | - Sicen Wang
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
- Shaanxi Engineering Research Center of Cardiovascular Drugs Screening & Analysis, Xi’an 710061, China
| |
Collapse
|
338
|
Li A, Zhao J, Fu J, Cai J, Zhang P. Recent advances of biomimetic nano-systems in the diagnosis and treatment of tumor. Asian J Pharm Sci 2019; 16:161-174. [PMID: 33995611 PMCID: PMC8105416 DOI: 10.1016/j.ajps.2019.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/22/2019] [Accepted: 08/31/2019] [Indexed: 12/18/2022] Open
Abstract
The lack of effective methods of diagnosis and treatment presents a major barrier to combat against tumor. The biomimetic concept is an emerging field that expresses great application potential in tumor fighting. Strategy for combining nano-systems with biomimetic technology has gained increasing attention that is proved bioinspired, environmentally benign, and promising. Herein, we provide an up-to-date review of biomimetic nano-systems as well as their applications in tumor therapy. In addition, the challenges and future directions of biomimetic nano-systems to achieve clinical translation are also pointed out.
Collapse
Affiliation(s)
- Anning Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiawei Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingru Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jia Cai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
339
|
Zhang M, Ye J, Li C, Xia Y, Wang Z, Feng J, Zhang X. Cytomembrane-Mediated Transport of Metal Ions with Biological Specificity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900835. [PMID: 31508286 PMCID: PMC6724363 DOI: 10.1002/advs.201900835] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/10/2019] [Indexed: 05/17/2023]
Abstract
Metal ions are of significant importance in biomedical science. This study reports a new concept of cytomembrane-mediated biospecific transport of metal ions without using any other materials. For the first time, cytomembranes are exploited for two-step conjugation with metal ions to provide hybrid nanomaterials. The innate biofunction of cell membranes renders the hybrids with superior advantages over common vehicles for metal ions, including excellent biocompatibility, low immunogenic risk, and particularly specific biotargeting functionality. As a proof-of-concept demonstration, cancer cell membranes are used for in vivo delivery of various metal ions, including ruthenium, europium, iron, and manganese, providing a series of tumor-targeted nanohybrids capable of photothermal therapy/imaging, magnetic resonance imaging, photoacoustic imaging, and fluorescence imaging with improved performances. In addition, the special structure of the cell membrane allows easy accommodation of small-molecular agents within the nanohybrids for effective chemotherapy. This study provides a new class of metal-ion-included nanomaterials with versatile biofunctions and offers a novel solution to address the important challenge in the field of in vivo targeted delivery of metal ions.
Collapse
Affiliation(s)
- Ming‐Kang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| | - Jing‐Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| | - Chu‐Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| | - Yu Xia
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| | - Zi‐Yang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| | - Xian‐Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P.R. China
| |
Collapse
|
340
|
Yong T, Zhang X, Bie N, Zhang H, Zhang X, Li F, Hakeem A, Hu J, Gan L, Santos HA, Yang X. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat Commun 2019; 10:3838. [PMID: 31444335 PMCID: PMC6707218 DOI: 10.1038/s41467-019-11718-4] [Citation(s) in RCA: 540] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Developing biomimetic nanoparticles without loss of the integrity of proteins remains a major challenge in cancer chemotherapy. Here, we develop a biocompatible tumor-cell-exocytosed exosome-biomimetic porous silicon nanoparticles (PSiNPs) as drug carrier for targeted cancer chemotherapy. Exosome-sheathed doxorubicin-loaded PSiNPs (DOX@E-PSiNPs), generated by exocytosis of the endocytosed DOX-loaded PSiNPs from tumor cells, exhibit enhanced tumor accumulation, extravasation from blood vessels and penetration into deep tumor parenchyma following intravenous administration. In addition, DOX@E-PSiNPs, regardless of their origin, possess significant cellular uptake and cytotoxicity in both bulk cancer cells and cancer stem cells (CSCs). These properties endow DOX@E-PSiNPs with great in vivo enrichment in total tumor cells and side population cells with features of CSCs, resulting in anticancer activity and CSCs reduction in subcutaneous, orthotopic and metastatic tumor models. These results provide a proof-of-concept for the use of exosome-biomimetic nanoparticles exocytosed from tumor cells as a promising drug carrier for efficient cancer chemotherapy.
Collapse
Affiliation(s)
- Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China.,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Xiaoqiong Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Center for Biotechnology, Åbo Akademi University, 20520, Turku, Finland
| | - Xuting Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Fuying Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Abdul Hakeem
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China. .,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, 430074, Wuhan, China.
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, FI-00014, Helsinki, Finland. .,Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074, Wuhan, China. .,Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, 430074, Wuhan, China.
| |
Collapse
|
341
|
Internalization Characterization of Si Nanorod with Camouflaged Cell Membrane Proteins Reveals ATXN2 as a Negative Regulator. Cells 2019; 8:cells8080931. [PMID: 31430912 PMCID: PMC6721741 DOI: 10.3390/cells8080931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
The fabrication of shape-controlled nanocarriers is critical for efficient delivery of biomolecules across the cell membrane. Surface coating of the nanocarrier can improve internalization efficiency. Here, we developed a facile method of silicon nanorod fabrication leading to a controlled size and shape. We then systematically evaluated five surface modifications with membrane proteins from different cancer cell lines including MCF7, MD231, Hela, Panc-PDX, and Panc-1. We demonstrated that silicon nanorods coated with either a homolytic or heterolytic membrane protein coating have significantly improved internalization efficiency as compared with uncoated Si nanorods. To elucidate the molecular mechanism of the improved efficiency associated with a modified coating, we analyzed the coating membrane proteins derived from five cell lines with proteomics and identified 601 proteins shared by different cell sources. These proteins may function as cell-substrate adhesion molecules that contribute to the enhanced internalization. We also tested the internalization efficiency of nanorods with different coatings in each of the five cell lines to determine the influencing factors from target cells. We found that the internalization efficiency varied among different target cells, and the ranking of the average efficiency was as follows: Hela > Panc-PDX > MD231 > MCF7 > Panc-1. The bioinformatics analysis suggested that the low internalization efficiency in Panc-1 cells might be associated with the upregulation of ATXN2, which is a negative regulator of endocytosis. We further demonstrated that ATXN2 knockdown with specific siRNA significantly improved nanorod internalization efficiency in Panc-1 cells suggesting that ATXN2 can be a reference for efficiency prediction of nanoparticle delivery to tumor cells. Thus, we studied the effect of different cancer cell membrane proteins on nanorod uptake efficiencies. These results can improve nanorod internalization to cancer cells, including a fundamental understanding of the internalization efficiency of cancer cells.
Collapse
|
342
|
Kim HY, Kang M, Choo YW, Go SH, Kwon SP, Song SY, Sohn HS, Hong J, Kim BS. Immunomodulatory Lipocomplex Functionalized with Photosensitizer-Embedded Cancer Cell Membrane Inhibits Tumor Growth and Metastasis. NANO LETTERS 2019; 19:5185-5193. [PMID: 31298024 DOI: 10.1021/acs.nanolett.9b01571] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Liposomes are clinically used as drug carriers for cancer therapy; however, unwanted leakage of the encapsulated anticancer drug and poor tumor-targeting efficiency of liposomes may generate toxic side effects on healthy cells and lead to failure of tumor eradication. To overcome these limitations, we functionalized liposomes with a photosensitizer (KillerRed, KR)-embedded cancer cell membrane (CCM). A lipid adjuvant was also embedded in the lipocomplex to promote the anticancer immune response. KR proteins were expressed on CCM and did not leak from the lipocomplex. Owing to the homotypic affinity of the CCM for the source cancer cells, the lipocomplex exhibited a 3.3-fold higher cancer-targeting efficiency in vivo than a control liposome. The liposome functionalized with KR-embedded CCM and lipid adjuvant generated cytotoxic reactive oxygen species in photodynamic therapy and effectively induced anticancer immune responses, inhibiting primary tumor growth and lung metastasis in homotypic tumor-bearing mice. Taken together, the lipocomplex technology may improve liposome-based cancer therapy.
Collapse
Affiliation(s)
- Han Young Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Yeon Woong Choo
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seok-Hyeong Go
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul 08826 , Republic of Korea
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul 08826 , Republic of Korea
- Institute of Chemical Processes , Seoul National University , Seoul 08826 , Republic of Korea
| |
Collapse
|
343
|
Meng QF, Cheng YX, Huang Q, Zan M, Xie W, Sun Y, Li R, Wei X, Guo SS, Zhao XZ, Rao L, Liu W. Biomimetic Immunomagnetic Nanoparticles with Minimal Nonspecific Biomolecule Adsorption for Enhanced Isolation of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:28732-28739. [PMID: 31339033 DOI: 10.1021/acsami.9b10318] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunomagnetic micro/nanoparticles (IMNs) have been widely used to isolate rare circulating tumor cells (CTCs) from blood samples for early diagnosis of cancers. However, when entering into biofluids, IMNs nonspecifically adsorb biomolecules and the in situ formed biomolecule corona covers IMN surface ligands and weakens the targeting capabilities of IMNs. In this work, we demonstrated that by surface coating of IMNs with red blood cell (RBC)-derived vesicles, the obtained biomimetic particles (RBC-IMNs) basically adsorb no biomolecules and maintain the CTC targeting ability when exposed to plasma. Compared to IMNs, RBC-IMNs exhibited an excellent cell isolation efficiency in spiked blood samples, which was improved to 95.71% from 60.22%. Furthermore, by using RBC-IMNs, we successfully isolated CTCs in 28 out of 30 prostate cancer patient blood samples and further showed the robustness of RBC-IMNs in downstream cell sequencing. The work presented here provides a new insight into developing targeted nanomaterials for biological and medical applications.
Collapse
Affiliation(s)
- Qian-Fang Meng
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology , Renming Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Qinqin Huang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
- Department of Obstetrics and Gynecology , Renming Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Minghui Zan
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
- Department of Obstetrics and Gynecology , Renming Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Wei Xie
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Yue Sun
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Rui Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Xiaoyun Wei
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| | - Lang Rao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB) , National Institutes of Health (NIH) , Bethesda , Maryland 20892 , United States
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology , Wuhan University , Wuhan , Hubei 430072 , China
| |
Collapse
|
344
|
Yan H, Shao D, Lao Y, Li M, Hu H, Leong KW. Engineering Cell Membrane-Based Nanotherapeutics to Target Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900605. [PMID: 31406672 PMCID: PMC6685500 DOI: 10.1002/advs.201900605] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/28/2019] [Indexed: 05/10/2023]
Abstract
Inflammation is ubiquitous in the body, triggering desirable immune response to defend against dangerous signals or instigating undesirable damage to cells and tissues to cause disease. Nanomedicine holds exciting potential in modulating inflammation. In particular, cell membranes derived from cells involved in the inflammatory process may be used to coat nanotherapeutics for effective targeted delivery to inflammatory tissues. Herein, the recent progress of rationally engineering cell membrane-based nanotherapeutics for inflammation therapy is highlighted, and the challenges and opportunities presented in realizing the full potential of cell-membrane coating in targeting and manipulating the inflammatory microenvironment are discussed.
Collapse
Affiliation(s)
- Huize Yan
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Dan Shao
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Yeh‐Hsing Lao
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Mingqiang Li
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Guangdong Provincial Key Laboratory of Liver DiseaseThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510630China
| | - Hanze Hu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Institutes of Life SciencesSchool of Biomedical Science and Engineering and National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510006China
- Department of System BiologyColumbia University Medical CenterNew YorkNY10032USA
| |
Collapse
|
345
|
Zhang J, Lin Y, Zhou H, He H, Ma J, Luo M, Zhang Z, Pang D. Cell Membrane-Camouflaged NIR II Fluorescent Ag 2 Te Quantum Dots-Based Nanobioprobes for Enhanced In Vivo Homotypic Tumor Imaging. Adv Healthc Mater 2019; 8:e1900341. [PMID: 31125518 DOI: 10.1002/adhm.201900341] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/12/2019] [Indexed: 11/11/2022]
Abstract
The advantages of fluorescence bioimaging in the second near-infrared (NIR II, 1000-1700 nm) window are well known; however, current NIR II fluorescent probes for in vivo tumor imaging still have many shortcomings, such as low fluorescence efficiency, unstable performance under in vivo environments, and inefficient enrichment at tumor sites. In this study, Ag2 Te quantum dots (QDs) that emit light at a wavelength of 1300 nm are assembled with poly(lactic-co-glycolic acid) and further encapsulated within cancer cell membranes to overcome the shortcomings mentioned above. The as-prepared ≈100 nm biomimetic nanobioprobes exhibit ultrabright (≈60 times greater than that of free Ag2 Te QDs) and highly stable (≈97% maintenance after laser radiation for 1 h) fluorescence in the NIR II window. By combining the active homotypic tumor targeting capability derived from the source cell membrane with the passive enhanced permeation and retention effect, improved accumulation at tumor sites ((31 ± 2)% injection dose per gram of tumor) and a high tumor-to-normal tissue ratio (13.3 ± 0.7) are achieved. In summary, a new biomimetic NIR II fluorescent nanobioprobe with ultrabright and stable fluorescence, homotypic targeting and good biocompatibility for enhanced in vivo tumor imaging is developed in this study.
Collapse
Affiliation(s)
- Jing‐Jing Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
| | - Yi Lin
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
| | - Hui Zhou
- School of Pharmaceutical SciencesWuhan University Wuhan 430072 P. R. China
| | - He He
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
| | - Jiao‐Jiao Ma
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
| | - Meng‐Yao Luo
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
| | - Zhi‐Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
| | - Dai‐Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)College of Chemistry and Molecular SciencesState Key Laboratory of VirologyThe Institute for Advanced Studies, and Wuhan Institute of BiotechnologyWuhan University Wuhan 430072 P. R. China
- College of ChemistryNankai University Tianjin 300071 P. R. China
| |
Collapse
|
346
|
Qiu C, Han HH, Sun J, Zhang HT, Wei W, Cui SH, Chen X, Wang JC, Zhang Q. Regulating intracellular fate of siRNA by endoplasmic reticulum membrane-decorated hybrid nanoplexes. Nat Commun 2019; 10:2702. [PMID: 31221991 PMCID: PMC6586638 DOI: 10.1038/s41467-019-10562-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 05/17/2019] [Indexed: 01/07/2023] Open
Abstract
Most cationic vectors are difficult to avoid the fate of small interfering RNA (siRNA) degradation following the endosome-lysosome pathway during siRNA transfection. In this study, the endoplasmic reticulum (ER) membrane isolated from cancer cells was used to fabricate an integrative hybrid nanoplexes (EhCv/siRNA NPs) for improving siRNA transfection. Compared to the undecorated Cv/siEGFR NPs, the ER membrane-decorated EhCv/siRNA NPs exhibits a significantly higher gene silencing effect of siRNA in vitro and a better antitumor activity in nude mice bearing MCF-7 human breast tumor in vivo. Further mechanistic studies demonstrate that functional proteins on the ER membrane plays important roles on improving cellular uptake and altering intracellular trafficking pathway of siRNA. It is worth to believe that the ER membrane decoration on nanoplexes can effectively transport siRNA through the endosome-Golgi-ER pathway to evade lysosomal degradation and enhance the silencing effects of siRNA.
Collapse
Affiliation(s)
- Chong Qiu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Hu-Hu Han
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jing Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Hai-Tao Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, 410013, Changsha, China
| | - Wei Wei
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Shi-He Cui
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Xin Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| |
Collapse
|
347
|
Lu G, Lv C, Bao W, Li F, Zhang F, Zhang L, Wang S, Gao X, Zhao D, Wei W, Xie HY. Antimonene with two-orders-of-magnitude improved stability for high-performance cancer theranostics. Chem Sci 2019; 10:4847-4853. [PMID: 31183034 PMCID: PMC6520929 DOI: 10.1039/c9sc00324j] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
Although the antimonene (AM) nanomaterial is recently emerging as a new photothermal therapy (PTT) agent, its rapid degradation in physiological medium immensely limits its direct utilization. To this end, we herein engineered AM by the cooperation of dimension optimization, size control, and cell membrane (CM) camouflage. Compared with traditional AM nanosheets, the resulting AM nanoparticles (∼55 nm) cloaked with the CM (denoted as CmNPs) exhibited significantly improved stability and increased photothermal efficacy as well as superior tumor targeting capacity. After intravenous injection, the CmNPs enabled satisfactory photoacoustic/photothermal multimodal imaging at tumor sites. Meanwhile, the PTT together with the newly explored function of photodynamic therapy (PDT) achieved a potent combination therapy with few side effects. The maximized theranostic performance thus strongly recommends CmNPs as a safe and highly reliable modality for anticancer therapy.
Collapse
Affiliation(s)
- Guihong Lu
- School of Life Science , Beijing Institute of Technology , No. 5 South Zhong Guan Cun Street , Beijing 100081 , China . ;
| | - Chengliang Lv
- School of Life Science , Beijing Institute of Technology , No. 5 South Zhong Guan Cun Street , Beijing 100081 , China . ;
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering , Institute of Process Engineering , Chinese Academy of Sciences , 1 North 2nd Street, Zhong Guan Cun , Beijing 100190 , China
| | - Feng Li
- School of Life Science , Beijing Institute of Technology , No. 5 South Zhong Guan Cun Street , Beijing 100081 , China . ;
| | - Fan Zhang
- School of Life Science , Beijing Institute of Technology , No. 5 South Zhong Guan Cun Street , Beijing 100081 , China . ;
| | - Lijun Zhang
- State Key Laboratory of Biochemical Engineering , Institute of Process Engineering , Chinese Academy of Sciences , 1 North 2nd Street, Zhong Guan Cun , Beijing 100190 , China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering , Institute of Process Engineering , Chinese Academy of Sciences , 1 North 2nd Street, Zhong Guan Cun , Beijing 100190 , China
| | - Xiaoyong Gao
- State Key Laboratory of Biochemical Engineering , Institute of Process Engineering , Chinese Academy of Sciences , 1 North 2nd Street, Zhong Guan Cun , Beijing 100190 , China
| | - Dongxu Zhao
- School of Life Science , Beijing Institute of Technology , No. 5 South Zhong Guan Cun Street , Beijing 100081 , China . ;
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering , Institute of Process Engineering , Chinese Academy of Sciences , 1 North 2nd Street, Zhong Guan Cun , Beijing 100190 , China
| | - Hai-Yan Xie
- School of Life Science , Beijing Institute of Technology , No. 5 South Zhong Guan Cun Street , Beijing 100081 , China . ;
| |
Collapse
|
348
|
Liu WL, Zou MZ, Liu T, Zeng JY, Li X, Yu WY, Li CX, Ye JJ, Song W, Feng J, Zhang XZ. Expandable Immunotherapeutic Nanoplatforms Engineered from Cytomembranes of Hybrid Cells Derived from Cancer and Dendritic Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1900499. [PMID: 30907473 DOI: 10.1002/adma.201900499] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/07/2019] [Indexed: 05/18/2023]
Abstract
Using the cytomembranes (FMs) of hybrid cells acquired from the fusion of cancer and dendritic cells (DCs), this study offers a biologically derived platform for the combination of immunotherapy and traditional oncotherapy approaches. Due to the immunoactivation implicated in the cellular fusion, FMs can effectively express whole cancer antigens and immunological co-stimulatory molecules for robust immunotherapy. FMs share the tumor's self-targeting character with the parent cancer cells. In bilateral tumor-bearing mouse models, the FM-coated nanophotosensitizer causes durable immunoresponse to inhibit the rebound of primary tumors post-nanophotosensitizer-induced photodynamic therapy (PDT). The FM-induced immunotherapy displays ultrahigh antitumor effects even comparable to that of PDT. On the other hand, PDT toward primary tumors enhances the immunotherapy-caused regression of the irradiation-free distant tumors. Consequently, both the primary and the distant tumors are almost completely eliminated. This tumor-specific immunotherapy-based nanoplatform is potentially expandable to multiple tumor types and readily equipped with diverse functions owing to the flexible nanoparticle options.
Collapse
Affiliation(s)
- Wen-Long Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Mei-Zhen Zou
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
| | - Tao Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Jin-Yue Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xue Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Wu-Yang Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Chu-Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Wen Song
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
349
|
Xuan M, Shao J, Li J. Cell membrane-covered nanoparticles as biomaterials. Natl Sci Rev 2019; 6:551-561. [PMID: 34691904 PMCID: PMC8291551 DOI: 10.1093/nsr/nwz037] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/12/2019] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
Surface engineering of synthetic carriers is an essential and important strategy for drug delivery in vivo. However, exogenous properties make synthetic nanosystems invaders that easily trigger the passive immune clearance mechanism, increasing the retention effect caused by the reticuloendothelial systems and bioadhesion, finally leading to low therapeutic efficacy and toxic effects. Recently, a cell membrane cloaking technique has been reported as a novel interfacing approach from the biological/immunological perspective, and has proved useful for improving the performance of synthetic nanocarriers in vivo. After cell membrane cloaking, nanoparticles not only acquire the physiochemical properties of natural cell membranes but also inherit unique biological functions due to the presence of membrane-anchored proteins, antigens, and immunological moieties. The derived biological properties and functions, such as immunosuppressive capability, long circulation time, and targeted recognition integrated in synthetic nanosystems, have enhanced their potential in biomedicine in the future. Here, we review the cell membrane-covered nanosystems, highlight their novelty, introduce relevant biomedical applications, and describe the future prospects for the use of this novel biomimetic system constructed from a combination of cell membranes and synthetic nanomaterials.
Collapse
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| |
Collapse
|
350
|
Min H, Wang J, Qi Y, Zhang Y, Han X, Xu Y, Xu J, Li Y, Chen L, Cheng K, Liu G, Yang N, Li Y, Nie G. Biomimetic Metal-Organic Framework Nanoparticles for Cooperative Combination of Antiangiogenesis and Photodynamic Therapy for Enhanced Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1808200. [PMID: 30773718 DOI: 10.1002/adma.201808200] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/27/2019] [Indexed: 05/25/2023]
Abstract
Photodynamic therapy (PDT) is a promising anticancer treatment and is clinically approved for different types of tumors. However, current PDT suffers several obstacles, including its neutralization by excess glutathione (GSH) in the tumor tissue and its strongly proangiogenic tumor response. In this work, a biomimic, multifunctional nanoparticle-based PDT agent, combining a tumor-targeted photosensitizer with GSH scavenging and antiangiogenesis therapy, is developed. A porphyrinic Zr-metal-organic framework nanoparticle is used simultaneously as the photosensitizer and the delivery vehicle of vascular endothelial growth factor receptor 2 (VEGFR2) inhibitor apatinib. The core nanoparticles are wrapped in MnO2 to consume the intratumoral GSH and then decorated with a tumor cell membrane camouflage. After intravenous administration, the nanoparticles selectively accumulate in tumor through homotypic targeting mediated by the biomimic decoration, and the combination of enhanced PDT and antiangiogenic drug significantly improves their tumor inhibition efficiency. This study provides an integrated solution for mechanism-based enhancement of PDT and demonstrates the encouraging potential for multifunctional nanosystem applicable for tumor therapy.
Collapse
Affiliation(s)
- Huan Min
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Jing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingqiu Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, P. R. China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuexiang Han
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Long Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Na Yang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|