351
|
Pasto A, Giordano F, Evangelopoulos M, Amadori A, Tasciotti E. Cell membrane protein functionalization of nanoparticles as a new tumor-targeting strategy. Clin Transl Med 2019; 8:8. [PMID: 30877412 PMCID: PMC6420595 DOI: 10.1186/s40169-019-0224-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Nanoparticles have seen considerable popularity as effective tools for drug delivery. However, non-specific targeting continues to remain a challenge. Recently, biomimetic nanoparticles have emerged as an innovative solution that exploits biologically-derived components to improve therapeutic potential. Specifically, cell membrane proteins extracted from various cells (i.e., leukocytes, erythrocytes, platelets, mesenchymal stem cells, cancer) have shown considerable promise in bestowing nanoparticles with increased circulation and targeting efficacy. Traditional nanoparticles can be detected and removed by the immune system which significantly hinders their clinical success. Biomimicry has been proposed as a promising approach to overcome these limitations. In this review, we highlight the current trends in biomimetic nanoparticles and describe how they are being used to increase their chemotherapeutic effect in cancer treatment.
Collapse
Affiliation(s)
- Anna Pasto
- Veneto Institute of Oncology-IRCCS, Padua, Italy.,Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Federica Giordano
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Alberto Amadori
- Veneto Institute of Oncology-IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX, 77030, USA. .,Houston Methodist Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
352
|
Zou H, Zhu J, Huang DS. Cell membrane capsule: a novel natural tool for antitumour drug delivery. Expert Opin Drug Deliv 2019; 16:251-269. [PMID: 30742557 DOI: 10.1080/17425247.2019.1581762] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chemotherapy plays an important role in antitumour therapy, but causes serious adverse reactions. So, drug delivery system (DDS) with cell-targeting ability is an important method to reduce adverse reactions while ensuring the effectiveness of chemotherapy. Synthetic drug carriers and DDSs based on cells have proven safety and efficacy, but they also have many deficiencies or limitations. Cell membrane capsules (CMCs), which are based on extracellular vesicles (EVs), are a promising biomimetic DDS that retains some cell membrane channels and cytoplasmic functions, with escape macrophage phagocytosis. AREAS COVERED The EVs for constructing CMCs can be prepared by natural secretion, chemical-induced budding, nanofilter membrane extrusion and similar methods and are isolated and purified by a variety of methods such as centrifugation and liquid chromatography. CMCs can target the tumour cells either spontaneously or through targeting modifications using proteins or aptamers to actively target the tumour cells. CMCs can be directly wrapped with chemicals, photosensitizers, RNA, proteins and other ingredients, or they can be loaded with antitumour agent-loaded synthetic nanoparticles, which are delivered to the target cells to play a specific role. EXPERT OPINION This review describes the concept, function, characteristics, origins, and manufacturing methods of CMCs and their application in antitumour therapy.
Collapse
Affiliation(s)
- Hai Zou
- a Clinical Research Institute , Zhejiang Provincial People's Hospital , Hangzhou , China.,b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , PR China.,c People's Hospital of Hangzhou Medical College , Hangzhou , Zhejiang Province , China.,d Medical College , Hangzhou , China
| | - Jing Zhu
- c People's Hospital of Hangzhou Medical College , Hangzhou , Zhejiang Province , China.,d Medical College , Hangzhou , China.,e Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Dong-Sheng Huang
- c People's Hospital of Hangzhou Medical College , Hangzhou , Zhejiang Province , China.,f Department of Hepatobiliary Surgery , Zhejiang Provincial People's Hospital , Hangzhou , China
| |
Collapse
|
353
|
Jin J, Krishnamachary B, Barnett JD, Chatterjee S, Chang D, Mironchik Y, Wildes F, Jaffee EM, Nimmagadda S, Bhujwalla ZM. Human Cancer Cell Membrane-Coated Biomimetic Nanoparticles Reduce Fibroblast-Mediated Invasion and Metastasis and Induce T-Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:7850-7861. [PMID: 30707559 PMCID: PMC6628902 DOI: 10.1021/acsami.8b22309] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Biomimetic nanoparticles (NPs) combine the flexibility and reproducibility of synthetic materials with the functionality of biological materials. Here, we developed and characterized biomimetic poly(lactic- co-glycolic acid) (PLGA) NPs coated with human cancer cell membrane fractions (CCMFs) to form CCMF-coated PLGA (CCMF-PLGA) NPs. We evaluated the ability of these CCMF-PLGA NPs to disrupt cancer cell-stromal cell interactions and to induce an immune response. Western blot analysis verified the plasma membrane purity of CCMFs. Confocal fluorescence microscopy and flow cytometry confirmed the presence of intact membrane-associated proteins including CXCR4 and CD44 following membrane derivation and coating. CCMFs and CCMF-PLGA NPs were capable of inhibiting cancer cell migration toward human mammary fibroblasts. Intravenous injection of CCMF-PLGA NPs significantly reduced experimental metastasis in vivo. Following immunization of Balb/c mice, near-infrared fluorescence imaging confirmed the migration of NPs to proximal draining lymph nodes (LNs). A higher percentage of CD8+ and CD4+ cytotoxic T-lymphocyte populations was observed in spleens and LNs of CCMF-PLGA NP-immunized mice. Splenocytes isolated from CCMF-PLGA NP-immunized mice had the highest number of interferon gamma-producing T-cells as detected by the ELISpot assay. CCMF-PLGA NPs hold promise for disrupting cancer cell-stromal cell interactions and for priming the immune system in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiefu Jin
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Correspondence should be addressed to: (ZMB); (JJ)
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James D. Barnett
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samit Chatterjee
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Di Chang
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Flonne Wildes
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sridhar Nimmagadda
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zaver M. Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Correspondence should be addressed to: (ZMB); (JJ)
| |
Collapse
|
354
|
Zhang KL, Wang YJ, Sun J, Zhou J, Xing C, Huang G, Li J, Yang H. Artificial chimeric exosomes for anti-phagocytosis and targeted cancer therapy. Chem Sci 2019; 10:1555-1561. [PMID: 30809374 PMCID: PMC6357862 DOI: 10.1039/c8sc03224f] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/23/2018] [Indexed: 12/28/2022] Open
Abstract
Development of exosome-based delivery systems is still facing some formidable challenges, including the lack of standardized isolation and purification methods, non-large-scale production and low drug-loading efficiency. Inspired by biomimetic technologies, we turned to the design of artificial chimeric exosomes (ACEs) constructed by integrating cell membrane proteins from multiple cell types into synthetic phospholipid bilayers. For benchmarking, hybrid membrane proteins derived from red blood cells (RBCs) and MCF-7 cancer cells were selected as models. The resulting ACEs were engineered much like "Emperor Qin's Terra-Cotta Warriors", simultaneously equipped with armor (anti-phagocytosis capability from RBCs) and dagger-axes (homologous targeting ability from cancer cells). ACEs demonstrated higher tumor accumulation, lower interception and better antitumor therapeutic effect than plain liposomes in vivo, alongside large-scale standardized preparation, stable structure, high drug-loading capacity and custom-tailored functionality, highlighting the suitability of ACEs as promising alternatives of exosomes in clinical applications.
Collapse
Affiliation(s)
- Kai-Long Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology , Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety , State Key Laboratory of Photocatalysis on Energy and Environment , College of Chemistry , Fuzhou University , Fuzhou 350116 , P. R. China . ;
| | - Ying-Jie Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology , Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety , State Key Laboratory of Photocatalysis on Energy and Environment , College of Chemistry , Fuzhou University , Fuzhou 350116 , P. R. China . ;
| | - Jin Sun
- Institute of Molecular Medicine , Renji Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai Jiao Tong University , Shanghai , 200240 , P. R. China
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , P. R. China
| | - Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology , Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety , State Key Laboratory of Photocatalysis on Energy and Environment , College of Chemistry , Fuzhou University , Fuzhou 350116 , P. R. China . ;
| | - Chao Xing
- MOE Key Laboratory for Analytical Science of Food Safety and Biology , Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety , State Key Laboratory of Photocatalysis on Energy and Environment , College of Chemistry , Fuzhou University , Fuzhou 350116 , P. R. China . ;
| | - Guoming Huang
- College of Biological Science and Engineering , Fuzhou University , Fuzhou 350116 , P. R. China
| | - Juan Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology , Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety , State Key Laboratory of Photocatalysis on Energy and Environment , College of Chemistry , Fuzhou University , Fuzhou 350116 , P. R. China . ;
- Institute of Molecular Medicine , Renji Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai Jiao Tong University , Shanghai , 200240 , P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology , Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety , State Key Laboratory of Photocatalysis on Energy and Environment , College of Chemistry , Fuzhou University , Fuzhou 350116 , P. R. China . ;
| |
Collapse
|
355
|
Lang T, Liu Y, Zheng Z, Ran W, Zhai Y, Yin Q, Zhang P, Li Y. Cocktail Strategy Based on Spatio-Temporally Controlled Nano Device Improves Therapy of Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1806202. [PMID: 30516854 DOI: 10.1002/adma.201806202] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/04/2018] [Indexed: 06/09/2023]
Abstract
Metastatic breast cancer may be resistant to chemo-immunotherapy due to the existence of cancer stem cells (CSC). Also, the control of particle size and drug release of a drug carrier for multidrug combination is a key issue influencing the therapy effect. Here, a cocktail strategy is reported, in which chemotherapy against both bulk tumor cells and CSC and immune checkpoint blockade therapy are intergraded into one drug delivery system. The chemotherapeutic agent paclitaxel (PTX), the anti-CSC agent thioridazine (THZ), and the PD-1/PD-L1 inhibitor HY19991 (HY) are all incorporated into an enzyme/pH dual-sensitive nanoparticle with a micelle-liposome double-layer structure. The particle size shrinks when the nanoparticle transfers from circulation to tumor tissues, favoring both pharmacokinetics and cellular uptake, meanwhile achieving sequential drug release where needed. This nano device, named PM@THL, increases the intratumoral drug concentrations in mice and exhibits significant anticancer efficacy, with tumor inhibiting rate of 93.45% and lung metastasis suppression rate of 97.64%. It also reduces the proportion of CSC and enhances the T cells infiltration in tumor tissues, and thus prolongs the survival of mice. The cocktail therapy based on the spatio-temporally controlled nano device will be a promising strategy for treating breast cancer.
Collapse
Affiliation(s)
- Tianqun Lang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiran Liu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Zhong Zheng
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- College of Life Sciences, Jilin University, Changchun, 130012, China
| | - Wei Ran
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
356
|
Jia Y, Wang X, Hu D, Wang P, Liu Q, Zhang X, Jiang J, Liu X, Sheng Z, Liu B, Zheng H. Phototheranostics: Active Targeting of Orthotopic Glioma Using Biomimetic Proteolipid Nanoparticles. ACS NANO 2019; 13:386-398. [PMID: 30576599 DOI: 10.1021/acsnano.8b06556] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Advances in phototheranostics revolutionized glioma intraoperative fluorescence imaging and phototherapy. However, the lack of desired active targeting agents for crossing the blood-brain barrier (BBB) significantly compromises the theranostic efficacy. In this study, biomimetic proteolipid nanoparticles (NPs) with U.S. Food and Drug Administration (FDA)-approved indocyanine green (ICG) were constructed to allow fluorescence imaging, tumor margin detection, and phototherapy of orthotopic glioma in mice. By embedding glioma cell membrane proteins into NPs, the obtained biomimetic ICG-loaded liposome (BLIPO-ICG) NPs could cross BBB and actively reach glioma at the early stage thanks to their specific binding to glioma cells due to their excellent homotypic targeting and immune escaping characteristics. High accumulation in the brain tumor with a signal to background ratio of 8.4 was obtained at 12 h post-injection. At this time point, the glioma and its margin were clearly visualized by near-infrared fluorescence imaging. Under the imaging guidance, the glioma tissue could be completely removed as a proof of concept. In addition, after NIR laser irradiation (1 W/cm2, 5 min), the photothermal effect exerted by BLIPO-ICG NPs efficiently suppressed glioma cell proliferation with a 94.2% tumor growth inhibition. No photothermal damages of normal brain tissue and treatment-induced side effects were observed. These results suggest that the biomimetic proteolipid NP is a promising phototheranostic nanoplatform for brain-tumor-specific imaging and therapy.
Collapse
Affiliation(s)
- Yali Jia
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences , Shaanxi Normal University , Xi'an 710119 , China
| | - Xiaobing Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences , Shaanxi Normal University , Xi'an 710119 , China
| | - Dehong Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Pan Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences , Shaanxi Normal University , Xi'an 710119 , China
| | - Quanhong Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences , Shaanxi Normal University , Xi'an 710119 , China
| | - Xuanjun Zhang
- Faculty of Health Sciences , University of Macau , Taipa , Macau SAR , China
| | - Jingying Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering , National University of Singapore , 4 Engineering Drive 4 , Singapore 117585 , Singapore
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering , Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences , Shenzhen 518055 , China
| |
Collapse
|
357
|
Cheng H, Jiang XY, Zheng RR, Zuo SJ, Zhao LP, Fan GL, Xie BR, Yu XY, Li SY, Zhang XZ. A biomimetic cascade nanoreactor for tumor targeted starvation therapy-amplified chemotherapy. Biomaterials 2019; 195:75-85. [PMID: 30616030 DOI: 10.1016/j.biomaterials.2019.01.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/08/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022]
Abstract
Targeted drug delivery with precisely controlled drug release and activation is highly demanding and challenging for tumor precision therapy. Herein, a biomimetic cascade nanoreactor (designated as Mem@GOx@ZIF-8@BDOX) is constructed for tumor targeted starvation therapy-amplified chemotherapy by assembling tumor cell membrane cloak and glucose oxidase (GOx) onto zeolitic imidazolate framework (ZIF-8) with the loading prodrug of hydrogen peroxide (H2O2)-sensitive BDOX. Biomimetic membrane camouflage affords superior immune evasion and homotypic binding capacities, which significantly enhance the tumor preferential accumulation and uptake for targeted drug delivery. Moreover, GOx-induced glycolysis would cut off glucose supply and metabolism pathways for tumor starvation therapy with the transformation of tumor microenvironments. Importantly, this artificial adjustment could trigger the site-specific BDOX release and activation for cascade amplified tumor chemotherapy regardless of the complexity and variability of tumor physiological environments. Both in vitro and in vivo investigations indicate that the biomimetic cascade nanoreactor could remarkably improve the therapeutic efficacy with minimized side effects through the synergistic starvation therapy and chemotherapy. This biomimetic cascade strategy would contribute to developing intelligent drug delivery systems for tumor precision therapy.
Collapse
Affiliation(s)
- Hong Cheng
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, PR China.
| | - Xue-Yan Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Rong-Rong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Sheng-Jia Zuo
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Lin-Ping Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Gui-Ling Fan
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Bo-Ru Xie
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Shi-Ying Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
358
|
Sun M, Yang D, Wang C, Bi H, Zhou Y, Wang X, Xu J, He F, Gai S, Yang P. AgBiS2-TPP nanocomposite for mitochondrial targeting photodynamic therapy, photothermal therapy and bio-imaging under 808 nm NIR laser irradiation. Biomater Sci 2019; 7:4769-4781. [DOI: 10.1039/c9bm01077g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AgBiS2 nanodots as an NIR light-excited photosensitizer produce a PDT effect, which is reported for the first time.
Collapse
|
359
|
Pan W, Zhang X, Gao P, Li N, Tang B. An anti-inflammatory nanoagent for tumor-targeted photothermal therapy. Chem Commun (Camb) 2019; 55:9645-9648. [DOI: 10.1039/c9cc04486h] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An anti-inflammatory nanoagent based on polydopamine nanospheres was developed to achieve highly efficient photothermal therapy, which can effectively kill cancer cells and simultaneously eliminate photothermal therapy-induced inflammation.
Collapse
Affiliation(s)
- Wei Pan
- College of Chemistry
- Chemical Engineering and Materials Science
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Institute of Molecular and Nano Science
| | - Xinhao Zhang
- College of Chemistry
- Chemical Engineering and Materials Science
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Institute of Molecular and Nano Science
| | - Peng Gao
- College of Chemistry
- Chemical Engineering and Materials Science
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Institute of Molecular and Nano Science
| | - Na Li
- College of Chemistry
- Chemical Engineering and Materials Science
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Institute of Molecular and Nano Science
| | - Bo Tang
- College of Chemistry
- Chemical Engineering and Materials Science
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Institute of Molecular and Nano Science
| |
Collapse
|
360
|
Sun Y, Wang H, Wang P, Zhang K, Geng X, Liu Q, Wang X. Tumor targeting DVDMS-nanoliposomes for an enhanced sonodynamic therapy of gliomas. Biomater Sci 2019; 7:985-994. [DOI: 10.1039/c8bm01187g] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
UTMD-assisted intelligent DVDMS encapsulate iRGD-Liposomes mediate SDT with deep tumor penetration and specific targeting ability enhanced anti-glioma efficacy.
Collapse
Affiliation(s)
- Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| | - Haiping Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| | - Kun Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| | - Xiaorui Geng
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| | - Quanhong Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China
- The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry
- The Ministry of Education
- College of Life Sciences
- Shaanxi Normal University
| |
Collapse
|
361
|
Xu X, Yang G, Xue X, Lu H, Wu H, Huang Y, Jing D, Xiao W, Tian J, Yao W, Pan CX, Lin TY, Li Y. A polymer-free, biomimicry drug self-delivery system fabricated via a synergistic combination of bottom-up and top-down approaches. J Mater Chem B 2018; 6:7842-7853. [PMID: 31380107 PMCID: PMC6676892 DOI: 10.1039/c8tb01464g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Compared to conventional carrier-assistant drug delivery systems (DDSs), drug self-delivery systems (DSDSs) have advantages of unprecedented drug loading capacity, minimized carrier-related toxicity and ease of preparation. However, the colloidal stability and blood circulation time of DSDSs still need to be improved. Here we report on the development of a novel biomimicry drug self-delivery system by the integration of a top-down cell membrane complexing technique into our self-delivery multifunctional nano-platform made from bottom-up approach that contains 100% active pharmaceutical ingredients (API) of Pheophorbide A and Irinotecan conjugates (named PI). Compared to conventional cell membrane coated nanoparticles with polymer framework as core and relatively low drug loading, this system consisting of red blood cell membrane vesicles complexed PI (RBC-PI) is polymer-free with up to 50% API loading. RBC-PI exhibited 10 times higher area under curve in pharmacokinetic study and much lower macrophage uptake compared with the parent PI nanoparticles. RBC-PI retained the excellent chemophototherapeutic effects of the PI nanoparticles, but possessed superior anti-cancer efficacy with prolonged blood circulation, improved tumor delivery, and enhanced photothermal effects in animal models. This system represents a novel example of using cell membrane complexing technique for effective surface modification of DSDSs. This is also an innovative study to form a polymer-free cell membrane nanoparticle complexing with positive surface charged materials. This biomimicry DSDS takes advantages of the best features from both systems to make up for each other's shortcomings and posed all the critical features for an ideal drug delivery system.
Collapse
Affiliation(s)
- Xiaobao Xu
- College of Biomedical Engineering & Instrument Science,
Zhejiang University, Hangzhou 310027, China
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Gaomai Yang
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Hongwei Lu
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Hao Wu
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Yee Huang
- Institute of Animal Husbandry and Veterinary Science,
Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, China
| | - Di Jing
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| | - Jingkui Tian
- College of Biomedical Engineering & Instrument Science,
Zhejiang University, Hangzhou 310027, China
| | - Wei Yao
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
| | - Chong-xian Pan
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
| | - Tzu-yin Lin
- Department of Internal Medicine, University of California
Davis, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis
Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817,
USA
| |
Collapse
|
362
|
Mesenchymal stem cell-based drug delivery strategy: from cells to biomimetic. J Control Release 2018; 294:102-113. [PMID: 30553849 DOI: 10.1016/j.jconrel.2018.12.019] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Owing to the diversity and ease of preparation of nanomaterials, the rational nanocarriers with a rational design have become increasingly popular in medical researches. Although nanoparticle-based drug delivery exhibits great potential, there are some challenges facing like rapid plasma clearance, triggering or aggravation of immune response, etc. Herein, cell-based targeted drug delivery systems have drawn more and more attention owing to low immunogenicity and intrinsic mutation rate, and innate ability to allow targeted delivery. Mesenchymal stem cells (MSCs) have been used in gene and drug delivery. The use of MSCs is a promising approach for the development of gene transfer systems and drug loading strategies because of their intrinsic properties, including homing ability and tumor tropism. By combining the inherent cell properties and merits of synthetic nanoparticles (NPs), cell membrane coated NPs emerge as the time requires. Overall, we provide a comprehensive overview of the utility of MSCs in drug and gene delivery as well as MSC membrane coated nanoparticles for therapy and drug delivery, aiming to figure out the significant room for development and highlight the potential future directions.
Collapse
|
363
|
Li Z, Hu S, Cheng K. Platelets and their biomimetics for regenerative medicine and cancer therapies. J Mater Chem B 2018; 6:7354-7365. [PMID: 31372220 PMCID: PMC6675472 DOI: 10.1039/c8tb02301h] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Platelets, circulating blood cells derived from megakaryocytes, play a key role in various physical activities, including coagulation, hemostasis, the body's innate immune response, and cancer metastasis. By taking advantage of their key traits, researchers have developed strategies to exploit platelets and platelet-mimicking nanoassemblies to treat a number of conditions, including wounds, cancers, and bacterial infections. Compared to traditional polymer, lipsosome, and inorganic nanoparticles-based delivery systems, platelets and platelet-mimicking vehicles hold many advantages. Among these are their enhanced circulation time, their large volumes and surface areas for drug loading or conjugation, and their inherent ability to target some diseases. In this review, we will highlight the recent progress made in the development of disease-targeting platelets- and platelet-mimicking-vehicles as therapeutic platforms.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
364
|
Yu Z, Zhou P, Pan W, Li N, Tang B. A biomimetic nanoreactor for synergistic chemiexcited photodynamic therapy and starvation therapy against tumor metastasis. Nat Commun 2018; 9:5044. [PMID: 30487569 PMCID: PMC6262009 DOI: 10.1038/s41467-018-07197-8] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/17/2018] [Indexed: 12/23/2022] Open
Abstract
Photodynamic therapy (PDT) is ineffective against deeply seated metastatic tumors due to poor penetration of the excitation light. Herein, we developed a biomimetic nanoreactor (bio-NR) to achieve synergistic chemiexcited photodynamic-starvation therapy against tumor metastasis. Photosensitizers on the hollow mesoporous silica nanoparticles (HMSNs) are excited by chemical energy in situ of the deep metastatic tumor to generate singlet oxygen (1O2) for PDT, and glucose oxidase (GOx) catalyzes glucose into hydrogen peroxide (H2O2). Remarkably, this process not only blocks the nutrient supply for starvation therapy but also provides H2O2 to synergistically enhance PDT. Cancer cell membrane coating endows the nanoparticle with biological properties of homologous adhesion and immune escape. Thus, bio-NRs can effectively convert the glucose into 1O2 in metastatic tumors. The excellent therapeutic effects of bio-NRs in vitro and in vivo indicate their great potential for cancer metastasis therapy. Photodynamic therapy is usually ineffective against deeply seated metastatic tumors due to poor penetration of the excitation light. Here, the authors design a biomimetic nanoreactor which can convert nutriment glucose into toxic singlet oxygen via chemiluminescence resonance energy transfer with no light excitation and demonstrate its high efficacy in a mouse lung metastatic model.
Collapse
Affiliation(s)
- Zhengze Yu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, China
| | - Ping Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
365
|
Bose RJC, Uday Kumar S, Zeng Y, Afjei R, Robinson E, Lau K, Bermudez A, Habte F, Pitteri SJ, Sinclair R, Willmann JK, Massoud TF, Gambhir SS, Paulmurugan R. Tumor Cell-Derived Extracellular Vesicle-Coated Nanocarriers: An Efficient Theranostic Platform for the Cancer-Specific Delivery of Anti-miR-21 and Imaging Agents. ACS NANO 2018; 12:10817-10832. [PMID: 30346694 PMCID: PMC6684278 DOI: 10.1021/acsnano.8b02587] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
MicroRNAs are critical regulators of cancer initiation, progression, and dissemination. Extensive evidence suggests that the inhibition of over-expressed oncogenic miRNA function can be a robust strategy for anticancer therapy. However, in vivo targeted delivery of miRNA therapeutics to various types of cancers remains a major challenge. Inspired by their natural synthesis and cargo delivery capabilities, researchers have exploited tumor cell-derived extracellular vesicles (TEVs) for the cancer-targeted delivery of therapeutics and theranostics. Here, we investigate a TEV-based nanoplatform for multimodal miRNA delivery and phototherapy treatments as well as the magnetic resonance imaging of cancer. We demonstrated loading of anti-miR-21 that blocks the function of endogenous oncogenic miR-21 over-expressed in cancer cells into and subsequent delivery by TEVs derived from 4T1 cells. We also produced Cy5-anti-miR-21-loaded TEVs from two other cancer cell lines (HepG2 and SKBR3) and confirmed their robust homologous and heterologous transfection efficiency and intracellular Cy5-anti-miR-21 delivery. Additionally, TEV-mediated anti-miR-21 delivery attenuated doxorubicin (DOX) resistance in breast cancer cells with a 3-fold higher cell kill efficiency than in cells treated with DOX alone. We then investigated TEVs as a biomimetic source for the functionalization of gold-iron oxide nanoparticles (GIONs) and demonstrated nanotheranostic properties of TEV-GIONs in vitro. TEV-GIONs demonstrated excellent T2 contrast in in vitro magnetic resonance (MR) imaging and resulted in efficient photothermal effect in 4T1 cells. We also evaluated the biodistribution and theranostic property of anti-miR-21 loaded TEV-GIONs in vivo by labeling with indocyanine green near-infrared dye. We further validated the tumor specific accumulation of TEV-GIONs using MR imaging. Our findings demonstrate that the distribution pattern of the TEV-anti-miR-21-GIONs correlated well with the tumor-targeting capability as well as the activity and efficacy obtained in response to doxorubicin combination treatments. TEVs and TEV-GIONs are promising nanotheranostics for future applications in cancer molecular imaging and therapy.
Collapse
Affiliation(s)
| | | | - Yitian Zeng
- Department of Materials Science and Engineering , Stanford University , Stanford , California 94305-4034 , United States
| | | | | | | | | | | | | | - Robert Sinclair
- Department of Materials Science and Engineering , Stanford University , Stanford , California 94305-4034 , United States
| | | | | | - Sanjiv S Gambhir
- Department of Materials Science and Engineering , Stanford University , Stanford , California 94305-4034 , United States
| | | |
Collapse
|
366
|
Jiang Q, Liu Y, Guo R, Yao X, Sung S, Pang Z, Yang W. Erythrocyte-cancer hybrid membrane-camouflaged melanin nanoparticles for enhancing photothermal therapy efficacy in tumors. Biomaterials 2018; 192:292-308. [PMID: 30465973 DOI: 10.1016/j.biomaterials.2018.11.021] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/22/2018] [Accepted: 11/13/2018] [Indexed: 12/19/2022]
Abstract
Cell membrane coating has emerged as an intriguing biomimetic strategy to endow nanomaterials with functions and properties inherent to source cells for various biomedical applications. Hybrid membrane of different types of cells could be coated onto nanoparticle surface to achieve additional functions. Herein, we fused red blood cell (RBC) membrane together with MCF-7 cell membrane and fabricated an erythrocyte-cancer (RBC-M) hybrid membrane-camouflaged melanin nanoparticle (Melanin@RBC-M) platform for enhancing therapeutic efficacy of photothermal therapy (PTT). The fused RBC-M hybrid membrane vesicles retained both RBC and MCF-7 cell membrane proteins and the resultant Melanin@RBC-M exhibited prolonged blood circulation and homotypic targeting to source MCF-7 cells simultaneously. Interestingly, increasing MCF-7 membrane components in RBC-M significantly enhanced the homotypic targeting function of Melanin@RBC-M while increasing RBC membrane components in RBC-M effectively reduced the cellular uptake of Melanin@RBC-M by macrophages and improved their circulation time in the blood. After intravenous injection into MCF-7 tumor-bearing athymic nude mice, Melanin@RBC-M with 1:1 membrane protein weight ratio of RBC to MCF-7 exhibited significantly higher tumor accumulation and better PTT efficacy compared with other Melanin@RBC-M with different membrane protein weight ratios as well as pristine melanin nanoparticles, due to the optimal balance between prolonged blood circulation and homotypic targeting. In addition, in vitro photoacoustic results revealed that Melanin@RBC-M had a photoacoustic signal enhancement with the increase of nanoparticle size (64 → 148 nm) and the photoacoustic amplitudes increased linearly with nanoparticle concentration at the excitation wavelength ranged from 680 nm to 800 nm, which could be used for quantification of Melanin@RBC-M in vivo. Looking forward, coating hybrid membrane onto nanoparticles could add flexibility and controllability in enhancing nanoparticles functionality and offer new opportunities for biomedical applications.
Collapse
Affiliation(s)
- Qin Jiang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai 200433, PR China
| | - Yao Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Ranran Guo
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai 200433, PR China
| | - Xianxian Yao
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai 200433, PR China
| | - Seunghyun Sung
- Department of Chemistry, Hankuk University of Foreign Studies, Seoul Campus 107, Imun-ro, Dongdaemun-gu, Seoul 02450, Republic of Korea
| | - Zhiqing Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science, Fudan University, Shanghai 200433, PR China.
| |
Collapse
|
367
|
Qin SY, Zhang AQ, Zhang XZ. Recent Advances in Targeted Tumor Chemotherapy Based on Smart Nanomedicines. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802417. [PMID: 30247806 DOI: 10.1002/smll.201802417] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/03/2018] [Indexed: 05/22/2023]
Abstract
Efficacy and safety of chemotherapeutic drugs constitute two major criteria in tumor chemotherapy. Nanomedicines with tumor-targeted properties hold great promise for improving the efficacy and safety. To design targeted nanomedicines, the pathological characteristics of tumors are extensively and deeply excavated. Here, the rationale, principles, and advantages of exploiting these pathological characteristics to develop targeted nanoplatforms for tumor chemotherapy are discussed. Homotypic targeting with the ability of self-recognition to source tumors is reviewed individually. In the meanwhile, the limitations and perspective of these targeted nanomedicines are also discussed.
Collapse
Affiliation(s)
- Si-Yong Qin
- School of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan, 430074, China
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Ai-Qing Zhang
- School of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan, 430074, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
368
|
Jiang Y, Chekuri S, Fang RH, Zhang L. Engineering biological interactions on the nanoscale. Curr Opin Biotechnol 2018; 58:1-8. [PMID: 30390535 DOI: 10.1016/j.copbio.2018.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/14/2018] [Indexed: 01/28/2023]
Abstract
Nanoparticulate platforms have contributed significantly to the field of biomedical research, demonstrating advantages over traditional modalities in areas such as drug delivery, detoxification, and vaccination. When it comes to the design of nanoparticles, biomimetic strategies have become increasingly popular as a means of promoting effective interactions with biological systems. A recently developed cell membrane-coated nanoparticle platform can leverage the natural interactions that cells engage in with other cells, the extracellular matrix, and biomolecules in order to reduce undesirable nonspecific interactions, while increasing target-specific interactions. Here, we discuss the current state of these biomimetic nanoparticles and highlight how they can be used for various biomedical applications.
Collapse
Affiliation(s)
- Yao Jiang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Sanam Chekuri
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
369
|
He H, Guo C, Wang J, Korzun WJ, Wang XY, Ghosh S, Yang H. Leutusome: A Biomimetic Nanoplatform Integrating Plasma Membrane Components of Leukocytes and Tumor Cells for Remarkably Enhanced Solid Tumor Homing. NANO LETTERS 2018; 18:6164-6174. [PMID: 30207473 PMCID: PMC6292712 DOI: 10.1021/acs.nanolett.8b01892] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Cell membrane-camouflaged nanoparticles have appeared as a promising platform to develop active tumor targeting nanomedicines. To evade the immune surveillance, we designed a composite cell membrane-camouflaged biomimetic nanoplatform, namely, leutusome, which is made of liposomal nanoparticles incorporating plasma membrane components derived from both leukocytes (murine J774A.1 cells) and tumor cells (head and neck tumor cells HN12). Exogenous phospholipids were used as building blocks to fuse with two cell membranes to form liposomal nanoparticles. Liposomal nanoparticles made of exogenous phospholipids only or in combination with one type of cell membrane were fabricated and compared. The anticancer drug paclitaxel (PTX) was used to make drug-encapsulating liposomal nanoparticles. Leutusome resembling characteristic plasma membrane components of the two cell membranes were examined and confirmed in vitro. A xenograft mouse model of head and neck cancer was used to profile the blood clearance kinetics, biodistribution, and antitumor efficacy of the different liposomal nanoparticles. The results demonstrated that leutusome obtained prolonged blood circulation and was most efficient accumulating at the tumor site (79.1 ± 6.6% ID per gram of tumor). Similarly, leutusome composed of membrane fractions of B16 melanoma cells and leukocytes (J774A.1) showed prominent accumulation within the B16 tumor, suggesting the generalization of the approach. Furthermore, PTX-encapsulating leutusome was found to most potently inhibit tumor growth while not causing systemic adverse effects.
Collapse
Affiliation(s)
- Hongliang He
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23219, United States
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Chunqing Guo
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Jing Wang
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - William J. Korzun
- Department of Clinical Laboratory Sciences, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Xiang-Yang Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Shobha Ghosh
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Hu Yang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23219, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
370
|
Valcourt DM, Harris J, Riley RS, Dang M, Wang J, Day ES. Advances in targeted nanotherapeutics: From bioconjugation to biomimicry. NANO RESEARCH 2018; 11:4999-5016. [PMID: 31772723 PMCID: PMC6879063 DOI: 10.1007/s12274-018-2083-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 05/20/2023]
Abstract
Since the emergence of cancer nanomedicine, researchers have had intense interest in developing nanoparticles (NPs) that can specifically target diseased sites while avoiding healthy tissue to mitigate the off-target effects seen with conventional treatments like chemotherapy. Initial endeavors focused on the bioconjugation of targeting agents to NPs, and more recently, researchers have begun to develop biomimetic NP platforms that can avoid immune recognition to maximally accumulate in tumors. In this review, we describe the advantages and limitations of each of these targeting strategies. First, we review developments in bioconjugation strategies, where NPs are coated with biomolecules such as antibodies, aptamers, peptides, and small molecules to enable cell-specific binding. While bioconjugated NPs offer many exciting features and have improved pharmacokinetics and biodistribution relative to unmodified NPs, they are still recognized by the body as "foreign", resulting in their clearance by the mononuclear phagocytic system (MPS). To overcome this limitation, researchers have recently begun to investigate biomimetic approaches that can hide NPs from immune recognition and reduce clearance by the MPS. These biomimetic NPs fall into two distinct categories: synthetic NPs that present naturally occurring structures, and NPs that are completely disguised by natural structures. Overall, bioconjugated and biomimetic NPs have substantial potential to improve upon conventional treatments by reducing off-target effects through site-specific delivery, and they show great promise for future standards of care. Here, we provide a summary of each strategy, discuss considerations for their design moving forward, and highlight their potential clinical impact on cancer therapy.
Collapse
Affiliation(s)
- Danielle M Valcourt
- 161 Colburn Lab, Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Jenna Harris
- 201 DuPont Hall, Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA
| | - Rachel S Riley
- 161 Colburn Lab, Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Megan Dang
- 161 Colburn Lab, Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Jianxin Wang
- 161 Colburn Lab, Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Emily S Day
- 161 Colburn Lab, Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
- 201 DuPont Hall, Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA
- 4701 Ogletown Stanton Road, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA
| |
Collapse
|
371
|
Vijayan V, Uthaman S, Park IK. Cell Membrane-Camouflaged Nanoparticles: A Promising Biomimetic Strategy for Cancer Theragnostics. Polymers (Basel) 2018; 10:polym10090983. [PMID: 30960908 PMCID: PMC6404000 DOI: 10.3390/polym10090983] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/25/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
Biomimetic functionalization of nanoparticles through camouflaging with cellular membranes has emerged as a promising strategy for cancer theragnostics. Cellular membranes used for camouflaging nanoparticles are generally isolated from blood cells, immune cells, cancer cells, and stem cells. The camouflaging strategy of wrapping nanoparticles with cellular membranes allows for superior tumor targeting through self-recognition, homotypic targeting and prolonged systematic circulation, thereby aiding in effective tumor therapy. In this review, we emphasized the various types of cellular membrane-camouflaged nanoparticles, their mechanisms in targeted therapy and various biomimetic strategies for anti-cancer therapy.
Collapse
Affiliation(s)
- Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 61469, Korea.
| | - Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-Ro, Yuseong-Gu, Daejeon 34134, Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 61469, Korea.
| |
Collapse
|
372
|
Zhang Z, Qian H, Huang J, Sha H, Zhang H, Yu L, Liu B, Hua D, Qian X. Anti-EGFR-iRGD recombinant protein modified biomimetic nanoparticles loaded with gambogic acid to enhance targeting and antitumor ability in colorectal cancer treatment. Int J Nanomedicine 2018; 13:4961-4975. [PMID: 30214200 PMCID: PMC6124475 DOI: 10.2147/ijn.s170148] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Red blood cell membrane-coated nanoparticle (RBCm-NP) platform, which consist of natural RBCm and synthetic polymeric core, can extend circulation time in vivo with an improved biocompatibility and stability of this biomimetic nanocarrier. To achieve better bioavailability of antitumor drugs that were loaded in RBCm-NPs, the functionalization of coated RBCm with specific targeting ability is essential. Bispecific recombinant protein anti-EGFR-iRGD, containing both tumor penetrating peptide (internalizing RGD peptide) and EGFR single-domain antibody (sdAb), seems to be an optimal targeting ligand for RBCm-NPs in the treatment of multiple tumors, especially colorectal cancer with high EGFR expression. Materials and methods We modified the anti-EGFR-iRGD recombinant protein on the surface of RBCm-NPs by lipid insertion method to construct iE-RBCm-PLGA NPs and confirmed the presentation of active tumor-targeting ability in colorectal cancer models with high EGFR expression when compared with RBCm-PLGA NPs. In addition, potential anti-tumor drug gambogic acid (GA) was loaded into the NPs to endow the antitumor efficiency of iE-RBCm-GA/PLGA NPs. It was simultaneously evaluated whether GA can reach better biocompatibility benefiting from the improved antitumor efficiency of iE-RBCm-GA/PLGA NPs in colorectal cancer models. Results We successfully modified anti-EGFR-iRGD proteins on the surface of biomimetic NPs with integrated and stable "shell-core" structure. iE-RBCm-PLGA NPs showed its improved targeting ability in vitro (multicellular spheroids [MCS]) and in vivo (nude mice bearing tumors). Besides, no matter on short-term cell apoptosis at tumor site (terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling [TUNEL]) and long-term tumor inhibition, iE-RBCm-GA/PLGA NPs achieved better antitumor efficacy than free GA in spite of the similar effects of cytotoxicity and apoptosis to GA in vitro. Conclusion We expect that the bispecific biomimetic nanocarrier can extend the clinical application of many other potential antitumor drugs similar to GA and become a novel drug carrier in the colorectal cancer treatment.
Collapse
Affiliation(s)
- Zhen Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China, .,Department of Integrated Traditional Chinese Medicine and Western Medicine Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Hanqing Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Jie Huang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Huizi Sha
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Hang Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Lixia Yu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Baorui Liu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| | - Dong Hua
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China,
| | - Xiaoping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, People's Republic of China,
| |
Collapse
|
373
|
Li J, Zhen X, Lyu Y, Jiang Y, Huang J, Pu K. Cell Membrane Coated Semiconducting Polymer Nanoparticles for Enhanced Multimodal Cancer Phototheranostics. ACS NANO 2018; 12:8520-8530. [PMID: 30071159 DOI: 10.1021/acsnano.8b04066] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Phototheranostic nanoagents are promising for early diagnosis and precision therapy of cancer. However, their imaging ability and therapeutic efficacy are often limited due to the presence of delivery barriers in the tumor microenvironment. Herein, we report the development of organic multimodal phototheranostic nanoagents that can biomimetically target cancer-associated fibroblasts in the tumor microenvironment for enhanced multimodal imaging-guided cancer therapy. Such biomimetic nanocamouflages comprise a near-infrared (NIR) absorbing semiconducting polymer nanoparticle (SPN) coated with the cell membranes of activated fibroblasts. The homologous targeting mechanism allows the activated fibroblast cell membrane coated SPN (AF-SPN) to specifically target cancer-associated fibroblasts, leading to enhanced tumor accumulation relative to the uncoated and cancer cell membrane coated counterparts after systemic administration in living mice. As such, AF-SPN not only provides stronger NIR fluorescence and photoacoustic signals to detect tumors but also generates enhanced cytotoxic heat and singlet oxygen to exert combinational photothermal and photodynamic therapy, ultimately leading to an antitumor efficacy higher than that of the counterparts. This study introduces an organic phototheranostic system that biomimetically targets the component in the tumor microenvironment for enhanced multimodal cancer theranostics.
Collapse
Affiliation(s)
- Jingchao Li
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore 637457
| | - Xu Zhen
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore 637457
| | - Yan Lyu
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore 637457
| | - Yuyan Jiang
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore 637457
| | - Jiaguo Huang
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore 637457
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore 637457
| |
Collapse
|
374
|
Lang T, Yin Q, Li Y. Progress of Cell-Derived Biomimetic Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800053] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tianqun Lang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; 501 Haike Road Shanghai 201203 China
- School of Pharmacy; University of Chinese Academy of Sciences; Beijing 100049 China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; 501 Haike Road Shanghai 201203 China
- School of Pharmacy; University of Chinese Academy of Sciences; Beijing 100049 China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica; Chinese Academy of Sciences; 501 Haike Road Shanghai 201203 China
- School of Pharmacy; University of Chinese Academy of Sciences; Beijing 100049 China
| |
Collapse
|
375
|
He XY, Liu BY, Xu C, Zhuo RX, Cheng SX. A multi-functional macrophage and tumor targeting gene delivery system for the regulation of macrophage polarity and reversal of cancer immunoresistance. NANOSCALE 2018; 10:15578-15587. [PMID: 30090893 DOI: 10.1039/c8nr05294h] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To achieve effective tumor eradication using anti-tumor immunotherapies, a fusion peptide functionalized gene delivery system for macrophage and tumor targeting delivery of the plasmid DNA encoding the IL-12 gene (pDNA IL-12) was prepared for macrophage re-polarization as well as reversal of cancer immunosuppression. A fusion peptide containing the tuftsin sequence that can interact with Fc receptors and neuropilin-1, and hyaluronic acid (HA) that can interact with CD44 were introduced into the delivery system by self-assembly to form peptide/hyaluronic acid/protamine/CaCO3/DNA nanoparticles (PHNP) with both macrophage targeting and tumor targeting capabilities. PHNP provides an efficient immunoregulation on J774A.1 cells to shift the anti-inflammatory M2 phenotype to the anti-tumor M1 phenotype with enhanced secretion of pro-inflammatory cytokines and increased expression of M1 markers. Owing to the improved delivery efficiency caused by the fusion peptide and HA, the transfection mediated by multi-functional PHNP can up-regulate IL-12 as well as down-regulate IL-10 and IL-4 more effectively as compared with the nanoparticles without HA and/or peptide decoration. More importantly, the gene delivery system can also deliver pDNA IL-12 to targeted cancerous HeLa cells to realize the secretion of IL-12. PHNP not only enables tumorous cells to produce pDNA IL-12, but also down-regulates CD47 and up-regulate CD80 and HLA-1 in the malignant cells, indicating that the gene delivery system can effectively reverse tumor induced immunosuppression.
Collapse
Affiliation(s)
- Xiao-Yan He
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, People's Republic of China.
| | | | | | | | | |
Collapse
|
376
|
Wang JL, Du XJ, Yang JX, Shen S, Li HJ, Luo YL, Iqbal S, Xu CF, Ye XD, Cao J, Wang J. The effect of surface poly(ethylene glycol) length on in vivo drug delivery behaviors of polymeric nanoparticles. Biomaterials 2018; 182:104-113. [PMID: 30114562 DOI: 10.1016/j.biomaterials.2018.08.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 12/29/2022]
Abstract
Engineering nanoparticles of reasonable surface poly(ethylene glycol) (PEG) length is important for designing efficient drug delivery systems. Eliminating the disturbance by other nanoproperties, such as size, PEG density, etc., is crucial for systemically investigating the impact of surface PEG length on the biological behavior of nanoparticles. In the present study, nanoparticles with different surface PEG length but similar other nanoproperties were prepared by using poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) copolymers of different molecular weights and incorporating different contents of PCL3500 homopolymer. The molecular weight of PEG block in PEG-PCL was between 3400 and 8000 Da, the sizes of nanoparticles were around 100 nm, the terminal PEG density was controlled at 0.4 PEG/nm2 (or the frontal PEG density was controlled at 0.16 PEG/nm2). Using these nanoproperties well-designed nanoparticles, we demonstrated PEG length-dependent changes in the biological behaviors of nanoparticles and exhibited nonmonotonic improvements as the PEG molecular weight increased from 3400 to 8000 Da. Moreover, under the experimental conditions, we found nanoparticles with a surface PEG length of 13.8 nm (MW = 5000 Da) significantly decreased the absorption with serum protein and interaction with macrophages, which led to prolonged blood circulation time, enhanced tumor accumulation and improved antitumor efficacy. The present study will help to establish a relatively precise relationship between surface PEG length and the in vivo behavior of nanoparticles.
Collapse
Affiliation(s)
- Ji-Long Wang
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China
| | - Xiao-Jiao Du
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China.
| | - Jin-Xian Yang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Song Shen
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China
| | - Hong-Jun Li
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China
| | - Ying-Li Luo
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Shoaib Iqbal
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Cong-Fei Xu
- Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
| | - Xiao-Dong Ye
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui, 230026, PR China
| | - Jie Cao
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, PR China.
| | - Jun Wang
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, PR China; Institutes for Life Sciences, School of Biomedical Science and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
377
|
Liu WL, Liu T, Zou MZ, Yu WY, Li CX, He ZY, Zhang MK, Liu MD, Li ZH, Feng J, Zhang XZ. Aggressive Man-Made Red Blood Cells for Hypoxia-Resistant Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802006. [PMID: 30015997 DOI: 10.1002/adma.201802006] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/23/2018] [Indexed: 05/06/2023]
Abstract
Extreme hypoxia of tumors represents the most notable barrier against the advance of tumor treatments. Inspired by the biological nature of red blood cells (RBCs) as the primary oxygen supplier in mammals, an aggressive man-made RBC (AmmRBC) is created to combat the hypoxia-mediated resistance of tumors to photodynamic therapy (PDT). Specifically, the complex formed between hemoglobin and enzyme-mimicking polydopamine, and polydopamine-carried photosensitizer is encapsulated inside the biovesicle that is engineered from the recombined RBC membranes. The mean corpuscular hemoglobin of AmmRBCs reaches about tenfold as high as that of natural RBCs. Owing to the same origin of outer membranes, AmmRBCs share excellent biocompatibility with parent RBCs. The introduced polydopamine plays the role of the antioxidative enzymes existing inside RBCs to effectively prevent the oxygen-carrying hemoglobin from the oxidation damage during the circulation. This biomimetic engineering can accumulate in tumors, permit in situ efficient oxygen supply, and impose strong PDT efficacy toward the extremely hypoxic tumor with complete tumor elimination. The man-made pseudo-RBC shows potentials as a universal oxygen-self-supplied platform to sensitize hypoxia-limited tumor treatment means, including but not limited to PDT. Meanwhile, this study offers ideas to the production of artificial substitutes of packed RBCs for clinical blood transfusion.
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
| | - Tao 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
| | - 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
| | - Zu-Yang He
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Ming-Kang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Miao-Deng Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Zi-Hao Li
- 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
| |
Collapse
|
378
|
Liang H, Huang K, Su T, Li Z, Hu S, Dinh PU, Wrona EA, Shao C, Qiao L, Vandergriff AC, Hensley MT, Cores J, Allen T, Zhang H, Zeng Q, Xing J, Freytes DO, Shen D, Yu Z, Cheng K. Mesenchymal Stem Cell/Red Blood Cell-Inspired Nanoparticle Therapy in Mice with Carbon Tetrachloride-Induced Acute Liver Failure. ACS NANO 2018; 12:6536-6544. [PMID: 29943967 PMCID: PMC6373867 DOI: 10.1021/acsnano.8b00553] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Acute liver failure is a critical condition characterized by global hepatocyte death and often time needs a liver transplantation. Such treatment is largely limited by donor organ shortage. Stem cell therapy offers a promising option to patients with acute liver failure. Yet, therapeutic efficacy and feasibility are hindered by delivery route and storage instability of live cell products. We fabricated a nanoparticle that carries the beneficial regenerative factors from mesenchymal stem cells and further coated it with the membranes of red blood cells to increase blood stability. Unlike uncoated nanoparticles, these particles promote liver cell proliferation in vitro and have lower internalization by macrophage cells. After intravenous delivery, these artificial stem cell analogs are able to remain in the liver and mitigate carbon tetrachloride-induced liver failure in a mouse model, as gauged by histology and liver function test. Our technology provides an innovative and off-the-shelf strategy to treat liver failure.
Collapse
Affiliation(s)
- Hongxia Liang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Teng Su
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Analytical Chemistry Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, Hebei 071002, China
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Phuong-Uyen Dinh
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Emily A. Wrona
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Chen Shao
- Department of Pathology, China-Japan Friendship Hospital, Peking 100029, China
| | - Li Qiao
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Adam C. Vandergriff
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - M. Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Tyler Allen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Hongyu Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Qinglei Zeng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jiyuan Xing
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| | - Deliang Shen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zujiang Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ke Cheng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering and Comparative Medicine Institute, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina 27599 and 27607, United States
| |
Collapse
|
379
|
Zhai Y, Ran W, Su J, Lang T, Meng J, Wang G, Zhang P, Li Y. Traceable Bioinspired Nanoparticle for the Treatment of Metastatic Breast Cancer via NIR-Trigged Intracellular Delivery of Methylene Blue and Cisplatin. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802378. [PMID: 29989211 DOI: 10.1002/adma.201802378] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 06/04/2018] [Indexed: 05/18/2023]
Abstract
Cytotoxic T lymphocyte (CTL) eliminates abnormal cells through target recognition-triggered intracellular toxin delivery. Chimeric antigen receptor T-cell improves cancer cell recognition of CTL, but its effectiveness and safety in solid tumor treatment are still hampered by poor tumor infiltration, suppressive tumor microenvironment, and severe on-target off-tumor toxicity. Given the functionality and challenges of CTL in cancer therapy, herein, a CTL-inspired nanovesicle (MPV) with a cell membrane-derived shell and a methylene blue (MB) and cisplatin (Pt) loaded gelatin nanogel core is created. The MPV generates contrast for tumor photoacoustic imaging, and produces hyperthermia upon laser irradiation, enabling photothermal imaging and deep tumor penetration. Meanwhile, it releases MB and Pt, and then delivers them into the cytosol of cancer cells, which process can be visualized by imaging the recovery of MB-derived fluorescence. The localized hyperthermia, photodynamic therapy, and chemotherapy together kill 4T1 breast cancer cells effectively, resulting in primary tumor regression and 97% inhibition of pulmonary metastasis, without significant toxicity to the animals. Taken together, the MPV shows tumor-specific and stimuli-triggered intracellular toxin delivery with advantages in traceable accumulation and activation, high tumor penetration, and triple combination therapy, and thus can be an effective nanomedicine for combating metastatic breast cancer.
Collapse
Affiliation(s)
- Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Wei Ran
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Jinghan Su
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Tianqun Lang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Jia Meng
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Guanru Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai, 264005, China
| |
Collapse
|
380
|
Ranganath SH. Bioengineered cellular and cell membrane-derived vehicles for actively targeted drug delivery: So near and yet so far. Adv Drug Deliv Rev 2018; 132:57-80. [PMID: 29935987 DOI: 10.1016/j.addr.2018.06.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 06/18/2018] [Indexed: 12/16/2022]
Abstract
Cellular carriers for drug delivery are attractive alternatives to synthetic nanoparticles owing to their innate homing/targeting abilities. Here, we review molecular interactions involved in the homing of Mesenchymal stem cells (MSCs) and other cell types to understand the process of designing and engineering highly efficient, actively targeting cellular vehicles. In addition, we comprehensively discuss various genetic and non-genetic strategies and propose futuristic approaches of engineering MSC homing using micro/nanotechnology and high throughput small molecule screening. Most of the targeting abilities of a cell come from its plasma membrane, thus, efforts to harness cell membranes as drug delivery vehicles are gaining importance and are highlighted here. We also recognize and report the lack of detailed characterization of cell membranes in terms of safety, structural integrity, targeting functionality, and drug transport. Finally, we provide insights on future development of bioengineered cellular and cell membrane-derived vesicles for successful clinical translation.
Collapse
Affiliation(s)
- Sudhir H Ranganath
- Bio-INvENT Lab, Department of Chemical Engineering, Siddaganga Institute of Technology, B.H. Road, Tumakuru, 572103, Karnataka, India.
| |
Collapse
|
381
|
Fang RH, Kroll AV, Gao W, Zhang L. Cell Membrane Coating Nanotechnology. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706759. [PMID: 29582476 PMCID: PMC5984176 DOI: 10.1002/adma.201706759] [Citation(s) in RCA: 1122] [Impact Index Per Article: 160.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/12/2017] [Indexed: 05/03/2023]
Abstract
Nanoparticle-based therapeutic, prevention, and detection modalities have the potential to greatly impact how diseases are diagnosed and managed in the clinic. With the wide range of nanomaterials available, the rational design of nanocarriers on an application-specific basis has become increasingly commonplace. Here, a comprehensive overview is provided on an emerging platform: cell-membrane-coating nanotechnology. As a fundamental unit of biology, cells carry out a wide range of functions, including the remarkable ability to interface and interact with their surrounding environment. Instead of attempting to replicate such functions via synthetic techniques, researchers are now directly leveraging naturally derived cell membranes as a means of bestowing nanoparticles with enhanced biointerfacing capabilities. This top-down technique is facile, highly generalizable, and has the potential to greatly augment existing nanocarriers. Further, the introduction of a natural membrane substrate onto nanoparticles surfaces has enabled additional applications beyond those traditionally associated with nanomedicine. Despite its relative youth, there exists an impressive body of literature on cell membrane coating, which is covered here in detail. Overall, there is still significant room for development, as researchers continue to refine existing workflows while finding new and exciting applications that can take advantage of this developing technology.
Collapse
Affiliation(s)
- Ronnie H. Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Ashley V. Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Weiwei Gao
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
382
|
Shao D, Li M, Wang Z, Zheng X, Lao YH, Chang Z, Zhang F, Lu M, Yue J, Hu H, Yan H, Chen L, Dong WF, Leong KW. Bioinspired Diselenide-Bridged Mesoporous Silica Nanoparticles for Dual-Responsive Protein Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801198. [PMID: 29808576 DOI: 10.1002/adma.201801198] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/06/2018] [Indexed: 05/20/2023]
Abstract
Controlled delivery of protein therapeutics remains a challenge. Here, the inclusion of diselenide-bond-containing organosilica moieties into the framework of silica to fabricate biodegradable mesoporous silica nanoparticles (MSNs) with oxidative and redox dual-responsiveness is reported. These diselenide-bridged MSNs can encapsulate cytotoxic RNase A into the 8-10 nm internal pores via electrostatic interaction and release the payload via a matrix-degradation controlled mechanism upon exposure to oxidative or redox conditions. After surface cloaking with cancer-cell-derived membrane fragments, these bioinspired RNase A-loaded MSNs exhibit homologous targeting and immune-invasion characteristics inherited from the source cancer cells. The efficient in vitro and in vivo anti-cancer performance, which includes increased blood circulation time and enhanced tumor accumulation along with low toxicity, suggests that these cell-membrane-coated, dual-responsive degradable MSNs represent a promising platform for the delivery of bio-macromolecules such as protein and nucleic acid therapeutics.
Collapse
Affiliation(s)
- Dan Shao
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical, Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Mingqiang Li
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Zheng Wang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical, Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Xiao Zheng
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Zhimin Chang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical, Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Fan Zhang
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Mengmeng Lu
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Juan Yue
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical, Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Huize Yan
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Li Chen
- Department of Pharmacology, Nanomedicine Engineering Laboratory of Jilin Province, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Wen-Fei Dong
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical, Engineering and Technology, Chinese Academy of Sciences, Suzhou, 215163, China
| | - Kam W Leong
- Department of Systems Biology, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
383
|
Zhu JY, Zhang MK, Ding XG, Qiu WX, Yu WY, Feng J, Zhang XZ. Virus-Inspired Nanogenes Free from Man-Made Materials for Host-Specific Transfection and Bio-Aided MR Imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1707459. [PMID: 29675900 DOI: 10.1002/adma.201707459] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/29/2018] [Indexed: 06/08/2023]
Abstract
Many viruses have a lipid envelope derived from the host cell membrane that contributes much to the host specificity and the cellular invasion. This study puts forward a virus-inspired technology that allows targeted genetic delivery free from man-made materials. Genetic therapeutics, metal ions, and biologically derived cell membranes are nanointegrated. Vulnerable genetic therapeutics contained in the formed "nanogene" can be well protected from unwanted attacks by blood components and enzymes. The surface envelope composed of cancer cell membrane fragments enables host-specific targeting of the nanogene to the source cancer cells and homologous tumors while effectively inhibiting recognition by macrophages. High transfection efficiency highlights the potential of this technology for practical applications. Another unique merit of this technology arises from the facile combination of special biofunction of metal ions with genetic therapy. Typically, Gd(III)-involved nanogene generates a much higher T1 relaxation rate than the clinically used Gd magnetic resonance imaging agent and harvests the enhanced MRI contrast at tumors. This virus-inspired technology points out a distinctive new avenue for the disease-specific transport of genetic therapeutics and other biomacromolecules.
Collapse
Affiliation(s)
- Jing-Yi Zhu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Ming-Kang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Guang Ding
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore, 117585, Singapore
| | - Wen-Xiu Qiu
- 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
| | - 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
| |
Collapse
|
384
|
A cancer vaccine-mediated postoperative immunotherapy for recurrent and metastatic tumors. Nat Commun 2018; 9:1532. [PMID: 29670088 PMCID: PMC5906566 DOI: 10.1038/s41467-018-03915-4] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 03/22/2018] [Indexed: 12/27/2022] Open
Abstract
Vaccines to induce effective and sustained antitumor immunity have great potential for postoperative cancer therapy. However, a robust cancer vaccine simultaneously eliciting tumor-specific immunity and abolishing immune resistance continues to be a challenge. Here we present a personalized cancer vaccine (PVAX) for postsurgical immunotherapy. PVAX is developed by encapsulating JQ1 (a BRD4 inhibitor) and indocyanine green (ICG) co-loaded tumor cells with a hydrogel matrix. Activation of PVAX by 808 nm NIR laser irradiation significantly inhibits the tumor relapse by promoting the maturation of dendritic cells and eliciting tumor infiltration of cytotoxic T lymphocytes. A mechanical study reveals that NIR light-triggered antigen release and JQ1-mediated PD-L1 checkpoint blockade cumulatively contribute to the satisfied therapeutic effect. Furthermore, PVAX prepared from the autologous tumor cells induces patient-specific memory immune response to prevent tumor recurrence and metastasis. The PVAX model might provide novel insights for postoperative immunotherapy. Cancer vaccines represent a promising personalized therapeutic approach to treating cancer. Here, the authors report the efficacy in a metastatic model of a cancer vaccine-mediated postoperative immunotherapy, based on the coencapsulation of the JQ1 and a photosensitizer ICG together with inactivated tumor cells into a hydrogel matrix.
Collapse
|
385
|
Jing L, Qu H, Wu D, Zhu C, Yang Y, Jin X, Zheng J, Shi X, Yan X, Wang Y. Platelet-camouflaged nanococktail: Simultaneous inhibition of drug-resistant tumor growth and metastasis via a cancer cells and tumor vasculature dual-targeting strategy. Am J Cancer Res 2018; 8:2683-2695. [PMID: 29774068 PMCID: PMC5957002 DOI: 10.7150/thno.23654] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/20/2018] [Indexed: 11/24/2022] Open
Abstract
Multidrug resistance (MDR) poses a great challenge to cancer therapy. It is difficult to inhibit the growth of MDR cancer due to its chemoresistance. Furthermore, MDR cancers are more likely to metastasize, causing a high mortality among cancer patients. In this study, a nanomedicine RGD-NPVs@MNPs/DOX was developed by encapsulating melanin nanoparticles (MNPs) and doxorubicin (DOX) inside RGD peptide (c(RGDyC))-modified nanoscale platelet vesicles (RGD-NPVs) to efficiently inhibit the growth and metastasis of drug-resistant tumors via a cancer cells and tumor vasculature dual-targeting strategy. Methods: The in vitro immune evasion potential and the targeting performance of RGD-NPVs@MNPs/DOX were examined using RAW264.7, HUVECs, MDA-MB-231 and MDA-MB-231/ADR cells lines. We also evaluated the pharmacokinetic behavior and the in vivo therapeutic performance of RGD-NPVs@MNPs/DOX using a MDA-MB-231/ADR tumor-bearing nude mouse model. Results: By taking advantage of the self-recognizing property of the platelet membrane and the conjugated RGD peptides, RGD-NPVs@MNPs/DOX was found to evade immune clearance and target the αvβ3 integrin on tumor vasculature and resistant breast tumor cells. Under irradiation with a NIR laser, RGD-NPVs@MNPs/DOX produced a multipronged effect, including reversal of cancer MDR, efficient killing of resistant cells by chemo-photothermal therapy, elimination of tumor vasculature for blocking metastasis, and long-lasting inhibition of the expressions of VEGF, MMP2 and MMP9 within the tumor. Conclusion: This versatile nanomedicine of RGD-NPVs@MNPs/DOX integrating unique biomimetic properties, excellent targeting performance, and comprehensive therapeutic strategies in one formulation might bring opportunities to MDR cancer therapy.
Collapse
|
386
|
Zhu J, Zhang M, Zheng D, Hong S, Feng J, Zhang XZ. A Universal Approach to Render Nanomedicine with Biological Identity Derived from Cell Membranes. Biomacromolecules 2018; 19:2043-2052. [PMID: 29584410 DOI: 10.1021/acs.biomac.8b00242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Biomimetic nanoengineering built through integrating the specific cell membrane with artificially synthetic nanomedicines represents one of the most promising directions for the actualization of personalized therapy. For addressing the technical hurdle against the development of this biomimetic technology, the present report describes the in-depth exploration and optimization over each critical preparation step, including establishment of a nanoparticle-stabilized dispersion system, cargo loading, membrane coating, and product isolation. Magnetic iron oxide nanoparticles loaded with DOX is used as a typical model for the coating with cancer cell membranes, providing compact DNP@CCCM nanostructure well-characterized by various techniques. Furthermore, the feasibility of this optimized approach in constructing biomimetic membrane-coated nanomedicines has been validated on the basis of the remarkably improved biofunctions, such as the targetability, magnetic property, hemolysis risk, macrophage evasion, in vitro cytotoxicity, in vivo circulation duration, and in vivo principal component analysis postinjection. We hope this study regarding technique optimization will prompt the advancement of biomembrane-camouflaged nanoparticles as a newly emerging biomimetic technology.
Collapse
Affiliation(s)
- Jingyi Zhu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , P.R. China.,Department of Chemical and Biomolecular Engineering , National University of Singapore , Singapore 117585 , Singapore
| | - Mingkang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , P.R. China
| | - Diwei Zheng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , P.R. China
| | - Sheng Hong
- 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
| |
Collapse
|
387
|
Xuan M, Shao J, Zhao J, Li Q, Dai L, Li J. Magnetic Mesoporous Silica Nanoparticles Cloaked by Red Blood Cell Membranes: Applications in Cancer Therapy. Angew Chem Int Ed Engl 2018; 57:6049-6053. [DOI: 10.1002/anie.201712996] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/04/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
| | - Qi Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| | - Luru Dai
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| |
Collapse
|
388
|
Xuan M, Shao J, Zhao J, Li Q, Dai L, Li J. Magnetic Mesoporous Silica Nanoparticles Cloaked by Red Blood Cell Membranes: Applications in Cancer Therapy. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201712996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
| | - Qi Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| | - Luru Dai
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology Beiyitiao 11 Beijing 100190 China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS) CAS Key Lab for Colloid, Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Beiyijie 2 Beijing 100190 China
- University of Chinese Academy of Sciences China
| |
Collapse
|
389
|
Li YJ, Yang CX, Yan XP. Biomimetic Persistent Luminescent Nanoplatform for Autofluorescence-Free Metastasis Tracking and Chemophotodynamic Therapy. Anal Chem 2018; 90:4188-4195. [PMID: 29504391 DOI: 10.1021/acs.analchem.8b00311] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Metastasis is the main cause of death in people with cancer. Early diagnosis and targeted therapy for metastasis is crucial for the survival of the cancer patients. However, metastasis is hard to trace for its small size, dispersed distribution and unvascularized anatomy. Here we report a biomimetic persistent luminescent nanoplatform for noninvasive high-sensitive diagnosis and 808 nm laser controlled photodynamics assisted chemotherapy of metastasis. The nanoplatform is composed of a photosensitizer functionalized persistent luminescent nanoparticle core, a doxorubicin loaded hollow silica interlayer and a cancer cell membrane shell for effective metastasis theranostic. The cancer cell membrane shell prevents drug leakage and endows the nanoplatform with targeting ability to metastasis. The reactivatable persistent luminescence of persistent luminescent nanoparticles not only enables long-term in vivo metastasis tracking, but also provides internal light source for singlet oxygen generation to kill cancer cells and further break the membrane shell for drug release. This work provides a promising strategy to develop persistent luminescence imaging guided theranostic nanoplatforms for early metastasis.
Collapse
Affiliation(s)
- Yu-Jie Li
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Cheng-Xiong Yang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China
| | - Xiu-Ping Yan
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing , Nankai University , Tianjin 300071 , China.,State Key Laboratory of Food Science and Technology (Jiangnan University), Institute of Analytical Food Safety, School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China.,Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300071 , China
| |
Collapse
|
390
|
Zhang N, Li M, Sun X, Jia H, Liu W. NIR-responsive cancer cytomembrane-cloaked carrier-free nanosystems for highly efficient and self-targeted tumor drug delivery. Biomaterials 2018; 159:25-36. [DOI: 10.1016/j.biomaterials.2018.01.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/08/2017] [Accepted: 01/03/2018] [Indexed: 12/31/2022]
|
391
|
Pei Q, Hu X, Zheng X, Liu S, Li Y, Jing X, Xie Z. Light-Activatable Red Blood Cell Membrane-Camouflaged Dimeric Prodrug Nanoparticles for Synergistic Photodynamic/Chemotherapy. ACS NANO 2018; 12:1630-1641. [PMID: 29346736 DOI: 10.1021/acsnano.7b08219] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Biomimetic approach offers numerous opportunities to design therapeutic platforms with enhanced antitumor performance and biocompatibility. Herein we report red blood cell membrane-camouflaged nanoparticles (RBC(M(TPC-PTX))) for synergistic chemo- and photodynamic therapy (PDT). Specifically, the inner core is mainly constructed by reactive oxygen species (ROS)-responsive PTX dimer (PTX2-TK) and photosensitizer 5,10,15,20-tetraphenylchlorin (TPC). In vitro experiments show that the prepared RBC(M(TPC-PTX)) is readily taken up into endosomes. Under appropriate light irradiation, the TPC can generate ROS, not only for PDT but also for triggering PTX2-TK cleavage and on-demand PTX release for chemotherapy. In vivo results show that the coating of RBC membrane prolongs blood circulation and improves tumor accumulation. The combination of chemo- and photodynamic therapy enhances anticancer therapeutic activity, and light-triggered drug release reduces systematic toxicity. All these characteristics render the described technology extremely promising for cancer treatment.
Collapse
Affiliation(s)
- Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China , Hefei, Anhui 230026, P. R. China
| | - Xiuli Hu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Xiaohua Zheng
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
- University of Science and Technology of China , Hefei, Anhui 230026, P. R. China
| | - Shi Liu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Yawei Li
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , 5625 Renmin Street, Changchun, Jilin 130022, P. R. China
| |
Collapse
|
392
|
Bose RJ, Paulmurugan R, Moon J, Lee SH, Park H. Cell membrane-coated nanocarriers: the emerging targeted delivery system for cancer theranostics. Drug Discov Today 2018; 23:891-899. [PMID: 29426004 DOI: 10.1016/j.drudis.2018.02.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/06/2018] [Accepted: 02/01/2018] [Indexed: 01/12/2023]
Abstract
Cancer is a leading cause of death worldwide. The use of nanocarriers (NCs) has generated significant interest to improve cancer therapy by targeted delivery. However, conventional NCs in general lack specificity and have poor biodistribution, resulting in low efficacy in cancer therapy. To circumvent this problem, there has been an increasing focus on cancer cell membrane-coated NCs (CCMCNCs), which can deliver therapeutics directly to tumor cells. CCMCNCs comprise active cancer cell surface adhesive molecules combined with other functional proteins, and offer extended blood circulation with robust cell-specific targeting, ensuring enhanced intratumoral penetration and higher tumor-specific accumulation of NCs. In this review, we discuss the preparation, homologous targeting mechanisms, and application of CCMCNCs in targeted cancer therapy.
Collapse
Affiliation(s)
- Rajendran Jc Bose
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - James Moon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Soo-Hong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
393
|
Zhang P, Zhang L, Qin Z, Hua S, Guo Z, Chu C, Lin H, Zhang Y, Li W, Zhang X, Chen X, Liu G. Genetically Engineered Liposome-like Nanovesicles as Active Targeted Transport Platform. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1705350. [PMID: 29280210 DOI: 10.1002/adma.201705350] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/15/2017] [Indexed: 05/26/2023]
Abstract
Ligand-targeted delivery of drug molecules to various types of tumor cells remains a major challenge in precision medicine. Inspired by the secretion process and natural cargo delivery functions of natural exosomes, biomimetic synthetic strategies are exploited to prepare biofunctionalized liposome-like nanovesicles (BLNs) that can artificially display a wide variety of targeting protein/peptide ligands and directly encapsulate medical agents for enhanced drug delivery. Here, as a proof of concept, genetically engineered BLNs, which display human epidermal growth factor (hEGF) or anti-HER2 Affibody as targeting moieties, are developed to, respectively, target two types of tumor cells. Notably, in comparison to synthetic liposomes covalently coupled with hEGF, it is demonstrated in this work that biosynthetically displayed hEGF ligands on BLNs possess higher biological activities and targeting capabilities. Additionally, treatments with doxorubicin-loaded BLNs displaying Affibody ligands exhibit much better antitumor therapeutic outcomes than clinically approved liposomal doxorubicin (Doxil) in HER2-overexpressing BT474 tumor xenograft models. These data suggest that BLN is suitable as a potent surrogate for conventional proteoliposomes or immunoliposomes as a result of excellent targeting capacities and facile production of BLNs.
Collapse
Affiliation(s)
- Pengfei Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Long Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Department of Hepatobiliary Pancreas and Vessel Surgery, Chenggong Hospital of Xiamen University, Xiamen, 361005, China
| | - Zainen Qin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Collaborative Innovation Center of Guangxi Biological Medicine and the, Medical and Scientific Research Center, Guangxi Medical University, Nanning, 530021, China
| | - Suhang Hua
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chengchao Chu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Huirong Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Wengang Li
- Department of Hepatobiliary Pancreas and Vessel Surgery, Chenggong Hospital of Xiamen University, Xiamen, 361005, China
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
394
|
Li Y, Xu Y, Fleischer CC, Huang J, Lin R, Yang L, Mao H. Impact of Anti-Biofouling Surface Coatings on the Properties of Nanomaterials and Their Biomedical Applications. J Mater Chem B 2018; 6:9-24. [PMID: 29479429 PMCID: PMC5821433 DOI: 10.1039/c7tb01695f] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Understanding and subsequently controlling non-specific interactions between engineered nanomaterials and biological environment have become increasingly important for further developing and advancing nanotechnology for biomedical applications. Such non-specific interactions, also known as the biofouling effect, mainly associate with the adsorption of biomolecules (such as proteins, DNAs, RNAs, and peptides) onto the surface of nanomaterials and the adhesion or uptake of nanomaterials by various cells. By altering the surface properties of nanomaterials the biofouling effect can lead to in situ changes of physicochemical properties, pharmacokinetics, functions, and toxicity of nanomaterials. This review provides discussions on the current understanding of the biofouling effect, the factors that affect the non-specific interactions associated with biofouling, and the impact of the biofouling effect on the performances and functions of nanomaterials. An overview of the development and applications of various anti-biofouling coating materials to preserve and improve the properties and functions of engineered nanomaterials for intended biomedical applications is also provided.
Collapse
Affiliation(s)
- Yuancheng Li
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yaolin Xu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Candace C Fleischer
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Run Lin
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
395
|
Li SY, Xie BR, Cheng H, Li CX, Zhang MK, Qiu WX, Liu WL, Wang XS, Zhang XZ. A biomimetic theranostic O 2 -meter for cancer targeted photodynamic therapy and phosphorescence imaging. Biomaterials 2018; 151:1-12. [DOI: 10.1016/j.biomaterials.2017.10.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/24/2017] [Accepted: 10/09/2017] [Indexed: 12/16/2022]
|
396
|
Ngandeu Neubi GM, Opoku-Damoah Y, Gu X, Han Y, Zhou J, Ding Y. Bio-inspired drug delivery systems: an emerging platform for targeted cancer therapy. Biomater Sci 2018; 6:958-973. [DOI: 10.1039/c8bm00175h] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bio-inspired platforms directly derived from biological sources are becoming a rapidly emerging field in the development of future anticancer therapeutics. The various platforms discussed are bacteria-based, virus-inspired, cell-derived, nanostructured lipid nanoparticles, and biomacromolecular drug delivery systems.
Collapse
Affiliation(s)
- Gella Maelys Ngandeu Neubi
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yaw Opoku-Damoah
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Xiaochen Gu
- Faculty of Pharmacy
- University of Manitoba
- Winnipeg
- Canada R3E 0T5
| | - Yue Han
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yang Ding
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
397
|
Zhang J, Zhang D, Hu X, Liu R, Li Z, Luan Y. Rational design of a new cytarabine-based prodrug for highly efficient oral delivery of cytarabine. RSC Adv 2018; 8:13103-13111. [PMID: 35542498 PMCID: PMC9079750 DOI: 10.1039/c8ra01225c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/27/2018] [Indexed: 11/25/2022] Open
Abstract
Because of the drawbacks of cytarabine (Ara-C) such as poor lipid solubility, deamination inactivation and low oral bioavailability limiting its application by oral administration, herein we propose a novel amphiphilic low molecular weight cytarabine prodrug (PA-Ara) by conjugating palmitic acid (PA) to Ara-C, making it possible to avoid the deamination inactivation by protecting the active 4-amino, as well as improving lipid solubility. Thanks to the rational design, the oil/water partition coefficient (P) of PA-Ara was improved tremendously compared with Ara-C, and the PA-Ara conjugation was stable enough in artificial digestive juice, ensuring that most molecules could be absorbed in the form of the prodrug. Results from an MTT assay conducted to measure the cytotoxicity of Ara-C and PA-Ara to HL60 (acute myeloblastic leukemia cell line) and K562 cells (chronic granulocytic leukemia cell line) showed that PA-Ara had significantly stronger antiproliferation activities than Ara-C. Significantly, we firstly compared the bioavailability of the oral fatty acid chain modified cytarabine prodrug preparation with injection and the relative bioavailability was up to 61.77% for our PA-Ara, which was much superior to that of oral Ara-C solution (3.23%). Overall, these findings make it clear that the PA-Ara suspension has the potential to be a promising new cytarabine oral preparation for leukemia therapy. To overcome the drawbacks of cytarabine such as bad liposolubility and low bioavailability, we rationally designed a new cytarabine-based prodrug for oral cytarabine delivery, realizing significantly enhanced bioavailability for cancer therapy.![]()
Collapse
Affiliation(s)
- Jing Zhang
- School of Pharmaceutical Science
- Shandong University
- Jinan
- China
| | - Di Zhang
- School of Pharmaceutical Science
- Shandong University
- Jinan
- China
| | - Xu Hu
- School of Pharmaceutical Science
- Shandong University
- Jinan
- China
| | - Ruiling Liu
- School of Pharmaceutical Science
- Shandong University
- Jinan
- China
| | - Zhonghao Li
- Key Laboratry of Colloid & Interface Chemistry
- Shandong University
- Ministry of Education
- China
| | - Yuxia Luan
- School of Pharmaceutical Science
- Shandong University
- Jinan
- China
| |
Collapse
|
398
|
Liu Y, Wang X, Ouyang B, Liu X, Du Y, Cai X, Guo H, Pang Z, Yang W, Shen S. Erythrocyte–platelet hybrid membranes coating polypyrrol nanoparticles for enhanced delivery and photothermal therapy. J Mater Chem B 2018; 6:7033-7041. [PMID: 32254586 DOI: 10.1039/c8tb02143k] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A dual-membrane coating technique was developed to camouflage polypyrrole nanoparticles with red blood cells and platelet membranes for enhanced delivery and photothermal tumor therapy.
Collapse
|
399
|
Cell Membrane Coated Nanoparticles: An Emerging Biomimetic Nanoplatform for Targeted Bioimaging and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:45-59. [PMID: 30471025 DOI: 10.1007/978-981-13-0445-3_3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biomimetic nanoplatform being a recent and emerging strategy plays an important role in a wide variety of applications. The different types of membranes used for coating include membranes from red blood cells, platelets, leucocytes, neutrophils, cancer cells, stem cells, etc. The as obtained membrane vesicles are fused onto the core nanoparticles through extrusion, sonication, electroporation. Biomimetic nanoparticles attain special functions which include ligand recognition and targeting, long blood circulation, immune escaping, tumor targeting depending on the core-shell interactions. The membrane coated nanoparticles indeed mimic the source cells and improves the therapeutic efficacy of drugs other cargos through specific delivery and enhanced accumulation in the tumor.
Collapse
|
400
|
Jia Y, Sheng Z, Hu D, Yan F, Zhu M, Gao G, Wang P, Liu X, Wang X, Zheng H. Highly penetrative liposome nanomedicine generated by a biomimetic strategy for enhanced cancer chemotherapy. Biomater Sci 2018; 6:1546-1555. [DOI: 10.1039/c8bm00256h] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Biomimetic liposome nanomedicine with deep tumor penetration and specific homotypic targeting ability enhanced cancer chemotherapy.
Collapse
|