51
|
Shi J, Ma Z, Pan H, Liu Y, Chu Y, Wang J, Chen L. Biofilm-encapsulated nano drug delivery system for the treatment of colon cancer. J Microencapsul 2020; 37:481-491. [DOI: 10.1080/02652048.2020.1797914] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Jinyan Shi
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Zhiwei Ma
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Hao Pan
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Yang Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Yuqi Chu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Jinglei Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| | - Lijiang Chen
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, China
| |
Collapse
|
52
|
Zhou J, Kroll AV, Holay M, Fang RH, Zhang L. Biomimetic Nanotechnology toward Personalized Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901255. [PMID: 31206841 PMCID: PMC6918015 DOI: 10.1002/adma.201901255] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/07/2019] [Indexed: 04/14/2023]
Abstract
While traditional approaches for disease management in the era of modern medicine have saved countless lives and enhanced patient well-being, it is clear that there is significant room to improve upon the current status quo. For infectious diseases, the steady rise of antibiotic resistance has resulted in super pathogens that do not respond to most approved drugs. In the field of cancer treatment, the idea of a cure-all silver bullet has long been abandoned. As a result of the challenges facing current treatment and prevention paradigms in the clinic, there is an increasing push for personalized therapeutics, where plans for medical care are established on a patient-by-patient basis. Along these lines, vaccines, both against bacteria and tumors, are a clinical modality that could benefit significantly from personalization. Effective vaccination strategies could help to address many challenging disease conditions, but current vaccines are limited by factors such as a lack of potency and antigenic breadth. Recently, researchers have turned toward the use of biomimetic nanotechnology as a means of addressing these hurdles. Recent progress in the development of biomimetic nanovaccines for antibacterial and anticancer applications is discussed, with an emphasis on their potential for personalized medicine.
Collapse
Affiliation(s)
- Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ashley V Kroll
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
53
|
Xu CH, Ye PJ, Zhou YC, He DX, Wei H, Yu CY. Cell membrane-camouflaged nanoparticles as drug carriers for cancer therapy. Acta Biomater 2020; 105:1-14. [PMID: 32001369 DOI: 10.1016/j.actbio.2020.01.036] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/18/2019] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
The translocation of natural cell membranes to the surface of synthetic nanoparticles, which allows man-made vectors to share merits and functionalities created by nature, has been a hot subject of research in the past decade. The resulting biomimetic nanoparticles not only retain the physicochemical properties of nanomaterials, but also inherit the advantageous functions of source cells. Combined with the preponderances of both synthetic and natural platforms, the optimized biomimetic systems can maximize the drug delivery efficiency. In this review, we first summarize the preparation strategies of the biomimetic systems from the perspective of the correlation between the properties of nanoparticles and cell membranes. Six types of cell membrane-camouflaged nanoparticles are further introduced with an emphasis on their properties and performance. Finally, a concluding remark regarding the primary challenges and opportunities associated with these nanoparticles is presented. STATEMENT OF SIGNIFICANCE: Translocation of natural cell membranes to the surface of synthetic nanoparticles has been repeatedly highlighted in the past decade to endow man-made vectors with merits and functionalities created by nature; therefore, the resulting biomimetic systems not only retain the physicochemical properties of nanomaterials but also inherit the biological functions of source cells for efficient drug delivery. To provide a timely review on this hot and rapidly developing subject of research, this paper summarized recent progress on the cell membrane-camouflaged nanoparticles as drug carriers for cancer therapy, and focused primarily on six different types of cell membrane-coated nanoparticles with an emphasis on the preparation strategies from the perspective of the correlation between the properties of nanoparticles and cell membrane.
Collapse
Affiliation(s)
- Cheng-Hui Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Peng-Ju Ye
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Yang-Chun Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Dong-Xiu He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China; Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| |
Collapse
|
54
|
Zhang Y, Ma N, Luo C, Zhu J, Bao C. Photosensitizer-loaded cell membrane biomimetic nanoparticles for enhanced tumor synergetic targeted therapy. RSC Adv 2020; 10:9378-9386. [PMID: 35497215 PMCID: PMC9050061 DOI: 10.1039/c9ra08926h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/23/2020] [Indexed: 11/21/2022] Open
Abstract
Photodynamic therapy (PDT) has the advantages of low toxicity and specificity, but photosensitizers usually fail to accumulate efficiently at the tumor site. In this study, a new multifunctional nano-drug delivery system was exploited by a biomimetic strategy to improve the PDT effects. The self-assembled methoxy poly(ethylene glycol)-poly(lactide-co-glycolide) (mPEG-PLGA) nanoparticles encapsulated with the photosensitizer chlorin e6 (Ce6) by microfluidics were employed as the nano-core, followed by coating red blood cell (RBC) membranes as the biomimetic agent to prolong the circulation time in vivo. In order to boost the therapeutic effect, doxorubicin (Dox) was preloaded into RBC nanovesicles. The cell membrane surface was modified with folic acid (FA) to further enhance the tumor targeting efficiency. The prepared biomimetic nanoparticles with a homogeneous size (70 nm) can trigger sufficient reactive oxygen species (ROS), leading to significant tumor ablation without side effects. In addition, the system had high tumor targeting efficiency, with an increase of 25% compared with no FA-modified nanoparticles. Therefore, this biomimetic multifunctional nanodrug delivery system possesses a prolonged circulation time and higher tumor targeting efficiency and can exert better tumor cytotoxicity for improved PDT due to homophilic targeting in vivo.
Collapse
Affiliation(s)
- Yunjiao Zhang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Nan Ma
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Congcong Luo
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Jiaquan Zhu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| | - Chunrong Bao
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092 P. R. China
| |
Collapse
|
55
|
Wang S, Yin Y, Song W, Zhang Q, Yang Z, Dong Z, Xu Y, Cai S, Wang K, Yang W, Wang X, Pang Z, Feng L. Red-blood-cell-membrane-enveloped magnetic nanoclusters as a biomimetic theranostic nanoplatform for bimodal imaging-guided cancer photothermal therapy. J Mater Chem B 2020; 8:803-812. [PMID: 31904076 DOI: 10.1039/c9tb01829h] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of red blood cell (RBC) membrane coatings has recently been found to be a biomimetic strategy to confer inner core nanomaterials with improved pharmacokinetic profiles by utilizing the intrinsic long blood circulation time of RBCs. Here, we envelope superparamagnetic nanoclusters (MNCs) with RBC membrane ghosts to obtain MNC@RBCs with significantly improved physiological stability compared to that of bare MNCs. After being loaded with near-infrared (NIR) cypate molecules, the as-prepared Cyp-MNC@RBCs show remarkably increased NIR absorbance and resultant efficient photothermal conversion efficacy. By tracking the NIR fluorescence of cypate in an in vivo fluorescence imaging system, we uncover that such Cyp-MNC@RBCs upon intravenous injection show significantly improved tumor-homing capacity as compared to bare cypate-loaded MNCs. A similar result is further evidenced by recording the T2-weighted magnetic resonance imaging (MRI) signal of MNCs. Furthermore, upon exposure to 808 nm laser irradiation, the tumors grown on the mice with the intravenous injection of Cyp-MNC@RBCs show a higher temperature increase than the tumors grown on the mice injected with plain MNC@RBCs and thus are significantly suppressed via photothermal ablation. This study presents the preparation of biomimetic Cyp-MNC@RBCs with greatly improved tumor-homing capacity as multifunctional nanotheranostic agents for fluorescence and MRI bimodal imaging-guided cancer photothermal therapy.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wang Song
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhijuan Yang
- Jiangsu Key Laboratory for Carbon-based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, P. R. China.
| | - Ziliang Dong
- Jiangsu Key Laboratory for Carbon-based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, P. R. China.
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Kuang Wang
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Wuli Yang
- Department of Macromolecular Science, Fudan University, Shanghai 200433, China
| | - Xuejun Wang
- Shanghai University of Traditional Chinese Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China.
| | - Liangzhu Feng
- Jiangsu Key Laboratory for Carbon-based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, P. R. China.
| |
Collapse
|
56
|
Liu X, Jansman MMT, Hosta-Rigau L. Haemoglobin-loaded metal organic framework-based nanoparticles camouflaged with a red blood cell membrane as potential oxygen delivery systems. Biomater Sci 2020; 8:5859-5873. [DOI: 10.1039/d0bm01118e] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metal organic frameworks are used to protect hemoglobin from denaturation thus preserving its excellent oxygen-binding and releasing properties. Decorating with cell membranes minimizes protein adsorption holding potential for long circulation.
Collapse
Affiliation(s)
- Xiaoli Liu
- DTU Health Tech
- Centre for Nanomedicine and Theranostics
- Technical University of Denmark
- 2800 Kgs. Lyngby
- Denmark
| | - Michelle M. T. Jansman
- DTU Health Tech
- Centre for Nanomedicine and Theranostics
- Technical University of Denmark
- 2800 Kgs. Lyngby
- Denmark
| | - Leticia Hosta-Rigau
- DTU Health Tech
- Centre for Nanomedicine and Theranostics
- Technical University of Denmark
- 2800 Kgs. Lyngby
- Denmark
| |
Collapse
|
57
|
Park JH, Dehaini D, Zhou J, Holay M, Fang RH, Zhang L. Biomimetic nanoparticle technology for cardiovascular disease detection and treatment. NANOSCALE HORIZONS 2020; 5:25-42. [PMID: 32133150 PMCID: PMC7055493 DOI: 10.1039/c9nh00291j] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Cardiovascular disease (CVD), which encompasses a number of conditions that can affect the heart and blood vessels, presents a major challenge for modern-day healthcare. Nearly one in three people has some form of CVD, with many suffering from multiple or intertwined conditions that can ultimately lead to traumatic events such as a heart attack or stroke. While the knowledge obtained in the past century regarding the cardiovascular system has paved the way for the development of life-prolonging drugs and treatment modalities, CVD remains one of the leading causes of death in developed countries. More recently, researchers have explored the application of nanotechnology to improve upon current clinical paradigms for the management of CVD. Nanoscale delivery systems have many advantages, including the ability to target diseased sites, improve drug bioavailability, and carry various functional payloads. In this review, we cover the different ways in which nanoparticle technology can be applied towards CVD diagnostics and treatments. The development of novel biomimetic platforms with enhanced functionalities is discussed in detail.
Collapse
Affiliation(s)
| | | | - Jiarong Zhou
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
58
|
Erythrocyte Membrane-Coated Arsenic Trioxide-Loaded Sodium Alginate Nanoparticles for Tumor Therapy. Pharmaceutics 2019; 12:pharmaceutics12010021. [PMID: 31878155 PMCID: PMC7022614 DOI: 10.3390/pharmaceutics12010021] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 11/23/2022] Open
Abstract
Arsenic trioxide (ATO) has a significant effect on the treatment of acute promyelocytic leukemia (APL) and advanced primary liver cancer, but it still faces severe side effects. Considering these problems, red blood cell membrane-camouflaged ATO-loaded sodium alginate nanoparticles (RBCM-SA-ATO-NPs, RSANs) were developed to relieve the toxicity of ATO while maintaining its efficacy. ATO-loaded sodium alginate nanoparticles (SA-ATO-NPs, SANs) were prepared by the ion crosslinking method, and then RBCM was extruded onto the surface to obtain RSANs. The average particle size of RSANs was found to be 163.2 nm with a complete shell-core bilayer structure, and the average encapsulation efficiency was 14.31%. Compared with SANs, RAW 264.7 macrophages reduced the phagocytosis of RSANs by 51%, and the in vitro cumulative release rate of RSANs was 95% at 84 h, which revealed a prominent sustained release. Furthermore, it demonstrated that RSANs had lower cytotoxicity as compared to normal 293 cells and exhibited anti-tumor effects on both NB4 cells and 7721 cells. In vivo studies further showed that ATO could cause mild lesions of main organs while RSANs could reduce the toxicity and improve the anti-tumor effects. In brief, the developed RSANs system provides a promising alternative for ATO treatment safely and effectively.
Collapse
|
59
|
Resveratrol-loaded PLGA nanoparticles functionalized with red blood cell membranes as a biomimetic delivery system for prolonged circulation time. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
60
|
Zhuang J, Holay M, Park JH, Fang RH, Zhang J, Zhang L. Nanoparticle Delivery of Immunostimulatory Agents for Cancer Immunotherapy. Theranostics 2019; 9:7826-7848. [PMID: 31695803 PMCID: PMC6831474 DOI: 10.7150/thno.37216] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023] Open
Abstract
Immunostimulatory agents, including adjuvants, cytokines, and monoclonal antibodies, hold great potential for the treatment of cancer. However, their direct administration often results in suboptimal pharmacokinetics, vulnerability to biodegradation, and compromised targeting. More recently, encapsulation into biocompatible nanoparticulate carriers has become an emerging strategy for improving the delivery of these immunotherapeutic agents. Such approaches can address many of the challenges facing current treatment modalities by endowing additional protection and significantly elevating the bioavailability of the encapsulated payloads. To further improve the delivery efficiency and subsequent immune responses associated with current nanoscale approaches, biomimetic modifications and materials have been employed to create delivery platforms with enhanced functionalities. By leveraging nature-inspired design principles, these biomimetic nanodelivery vehicles have the potential to alter the current clinical landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Jia Zhuang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Maya Holay
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Joon Ho Park
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H. Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jie Zhang
- Cello Therapeutics, Inc., San Diego, CA 92121, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
61
|
Abstract
Many diseases and conditions affect a relatively localized area of the body. They can be treated either by direct deposition of drug in the target area, or by giving the drug systemically. Here we review nanoparticle-based approaches to achieving both. We highlight advantages and disadvantages that nanoscale solutions have for locally administered therapies, with emphasis on the former. We discuss strategies to enable systemically delivered nanoparticles to deliver their payloads at specific locations in the body, including triggering (local and remote) and targeting.
Collapse
Affiliation(s)
- Tianjiao Ji
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
62
|
Maity A, Pal U, Chakraborty B, Sengupta C, Sau A, Chakraborty S, Basu S. Preferential photochemical interaction of Ru (III) doped carbon nano dots with bovine serum albumin over human serum albumin. Int J Biol Macromol 2019; 137:483-494. [DOI: 10.1016/j.ijbiomac.2019.06.126] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022]
|
63
|
Hanley T, Yin R, Mac JT, Tan W, Anvari B. Functionalized erythrocyte-derived optical nanoparticles to target ephrin-B2 ligands. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-9. [PMID: 31429216 PMCID: PMC6983482 DOI: 10.1117/1.jbo.24.8.085002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/29/2019] [Indexed: 06/10/2023]
Abstract
Over- or under-expression of erythropoietin-production human hepatocellular receptors (Eph) and their ligands are associated with various diseases. Therefore, these molecular biomarkers can potentially be used as binding targets for the delivery of therapeutic and/or imaging agents to cells characterized by such irregular expressions. We have engineered nanoparticles derived from erythrocytes and doped with the near-infrared (NIR) FDA-approved dye, indocyanine green. We refer to these nanoparticles as NIR erythrocyte-derived transducers (NETs). We functionalized the NETs with the ligand-binding domain of a particular Eph receptor, EphB1, to target the genetically modified human dermal microvascular endothelial cells (hDMVECs) with coexpression of EphB1 receptor and its ligand ephrin-B2. This cell model mimics the pathological phenotypes of lesional endothelial cells (ECs) in port wine stains (PWSs). Our quantitative fluorescence imaging results demonstrate that such functionalized NETs bind to the ephrin-B2 ligands on these hDMVECs in a dose-dependent manner that varies sigmoidally with the number density of the particles. These nanoparticles may potentially serve as agents to target PWS lesional ECs and other diseases characterized with over-expression of Eph receptors or their associated ligands to mediate phototherapy.
Collapse
Affiliation(s)
- Taylor Hanley
- University of California, Riverside, Department of Bioengineering, Riverside, California, United States
| | - Rong Yin
- University of South Carolina School of Medicine, Department of Cell Biology and Anatomy, Columbia, South Carolina, United States
| | - Jenny T. Mac
- University of California, Riverside, Department of Biochemistry, Riverside, California, United States
| | - Wenbin Tan
- University of South Carolina School of Medicine, Department of Cell Biology and Anatomy, Columbia, South Carolina, United States
| | - Bahman Anvari
- University of California, Riverside, Department of Bioengineering, Riverside, California, United States
| |
Collapse
|
64
|
Sun L, Li Q, Hou M, Gao Y, Yang R, Zhang L, Xu Z, Kang Y, Xue P. Light-activatable Chlorin e6 (Ce6)-imbedded erythrocyte membrane vesicles camouflaged Prussian blue nanoparticles for synergistic photothermal and photodynamic therapies of cancer. Biomater Sci 2019; 6:2881-2895. [PMID: 30192355 DOI: 10.1039/c8bm00812d] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multiple therapeutic modalities, such as photodynamic (PDT) and photothermal (PTT) therapies, have been jointly applied to produce a synergistic effect for tumor eradication based on the hyperthermia and generation of reactive oxygen species (ROS) mediated by photoactive agents. Effective delivery of highly efficient photosensitizers and photothermal agents is the key for combination of PDT/PTT. Herein, we propose a strategy to functionalize Prussian blue (PB) nanoparticles (NPs) with Chlorin e6 (Ce6)-imbedded erythrocyte membrane vesicles. This nanoplatform can address the major issues of these two capable photoactive agents, such as limited biocompatibility, lack of functional chemical groups, and poor bioavailability due to rapid blood clearance or self-aggregation. Specifically, PB NPs were packaged within Ce6-imbedded erythrocyte membrane vesicles, named as PB@RBC/Ce6 NPs, to take advantage of both biological functions of natural erythrocyte membranes and the unique physicochemical properties of synthetic nanoagents. Compared to bare PB NPs or free Ce6, PB@RBC/Ce6 NPs exhibited considerably enhanced cellular uptake and accumulation in tumoral tissues. Moreover, the PB@RBC/Ce6 NP-mediated PDT/PTT combination therapies produced a notable effect in boosting the necrosis and late apoptosis of tumor cells in vitro, and further showed a synergistic therapeutic effect against an orthotopic tumor model in vivo.
Collapse
Affiliation(s)
- Lihong Sun
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Rodríguez-Carlos A, Martinez-Gutierrez F, Torres-Juarez F, Rivas-Santiago B. Antimicrobial Peptides-based Nanostructured Delivery Systems: An Approach for Leishmaniasis Treatment. Curr Pharm Des 2019; 25:1593-1603. [PMID: 31264542 DOI: 10.2174/1381612825666190628152842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 06/19/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Leishmaniasis is a major health problem mainly in tropical and subtropical areas worldwide, although in the last decades it has been treated with the use of conventional drugs such as amphotericin, the emergence of multidrug-resistant strains has raised a warning signal to the public health systems thus a new call for the creation of new leishmanicidal drugs is needed. METHODS The goal of this review was to explore the potential use of antimicrobial peptides-based nanostructured delivery systems as an approach for leishmaniasis treatment. RESULTS Within these new potential drugs, human host defense peptides (HDP) can be included given their remarkable antimicrobial activity and their outstanding immunomodulatory functions for the therapy of leishmaniasis. CONCLUSION Though several approaches have been done using these peptides, new ways for delivering HDPs need to be analyzed, such is the case for nanotechnology.
Collapse
Affiliation(s)
- Adrian Rodríguez-Carlos
- Medical Research Unit- Zacatecas-IMSS, Zacatecas, Mexico.,División de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí. Mexico
| | - Fidel Martinez-Gutierrez
- Microbiology Laboratory, Facultad de Ciencias Quimicas, Universidad Autonoma de San Luis Potosi, Alvaro Obregon 64, Centro 78300, San Luis, S.L.P, Mexico
| | | | | |
Collapse
|
66
|
Chiu CY, Chung TW, Chen SY, Ma YH. Effects of PEGylation on capture of dextran-coated magnetic nanoparticles in microcirculation. Int J Nanomedicine 2019; 14:4767-4780. [PMID: 31308657 PMCID: PMC6613455 DOI: 10.2147/ijn.s204844] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/23/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Magnetic nanoparticles (MNPs) can be localized against hemodynamic forces in blood vessels with the application of an external magnetic field. In addition, PEGylation of nanoparticles may increase the half-life of nanocomposites in circulation. In this work, we examined the effect of PEGylation on the magnetic capture of MNPs in vivo. METHODS Laser speckle contrast imaging and capillaroscopy were used to assess the magnetic capture of dextran-coated MNPs and red blood cell (RBC) flow in cremaster microvessels of anesthetized rats. Magnetic capture of MNPs in serum flow was visualized with an in vitro circulating system. The effect of PEGylation on MNP-endothelial cell interaction was studied in cultured cells using an iron assay. RESULTS In microcirculation through cremaster muscle, magnet-induced retention of 250 nm MNPs was associated with a variable reduction in RBC flow, suggesting a dynamic coupling of hemodynamic and magnetic forces. After magnet removal, faster restoration of flow was observed in PEG(+) than PEG(-) group, which may be attributed to a reduced interaction with vascular endothelium. However, PEGylation appears to be required for magnetic capture of 50 nm MNPs in microvessels, which was associated with increased hydrodynamic diameter to 130±6 nm in serum, but independent of the ς-potential. CONCLUSION These results suggest that PEGylation may enhance magnetic capture of smaller MNPs and dispersion of larger MNPs after magnet removal, which may potentially affect the targeting, pharmacokinetics and therapeutic efficacy.
Collapse
Affiliation(s)
- Chien-Yu Chiu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Guishan, Taoyuan City33302, Taiwan, ROC
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Guishan, Taoyuan City33302, Taiwan, ROC
| | - Tze-Wen Chung
- Department of Biomedical Engineering, National Yang-Ming University, Beitou, Taipei City11221, Taiwan, ROC
- Center for Advanced Pharmaceutical Research and Drug Delivery, National Yang-Ming University, Beitou, Taipei City11221, Taiwan, ROC
| | - Si-Yi Chen
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Guishan, Taoyuan City33302, Taiwan, ROC
| | - Yunn-Hwa Ma
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Guishan, Taoyuan City33302, Taiwan, ROC
- Department of Neurology, Chang Gung Memorial Hospital, Guishan, Taoyuan City33305, Taiwan, ROC
| |
Collapse
|
67
|
Xia Q, Zhang Y, Li Z, Hou X, Feng N. Red blood cell membrane-camouflaged nanoparticles: a novel drug delivery system for antitumor application. Acta Pharm Sin B 2019; 9:675-689. [PMID: 31384529 PMCID: PMC6663920 DOI: 10.1016/j.apsb.2019.01.011] [Citation(s) in RCA: 341] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/30/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022] Open
Abstract
Erythrocytes (red blood cells, RBCs) are the most abundant circulating cells in the blood and have been widely used in drug delivery systems (DDS) because of their features of biocompatibility, biodegradability, and long circulating half-life. Accordingly, a "camouflage" comprised of erythrocyte membranes renders nanoparticles as a platform that combines the advantages of native erythrocyte membranes with those of nanomaterials. Following injection into the blood of animal models, the coated nanoparticles imitate RBCs and interact with the surroundings to achieve long-term circulation. In this review, the biomimetic platform of erythrocyte membrane-coated nano-cores is described with regard to various aspects, with particular focus placed on the coating mechanism, preparation methods, verification methods, and the latest anti-tumor applications. Finally, further functional modifications of the erythrocyte membranes and attempts to fuse the surface properties of multiple cell membranes are discussed, providing a foundation to stimulate extensive research into multifunctional nano-biomimetic systems.
Collapse
Key Words
- ABC, accelerated blood clearance
- APCs, antigen presenting cells
- Antitumor
- AuNCs, gold nanocages
- AuNPs, gold nanoparticles
- Biomimetic nanoparticles
- C8bp, C8 binding protein
- CR1, complement receptor 1
- DAF, decay accelerating factor
- DDS, drug delivery systems
- DLS, dynamic light scattering
- Dox, doxorubicin
- Drug delivery
- ECM, extracellular matrix
- EPR, enhanced permeability and retention
- ETA, endothelin A
- EpCam, epithelial cell adhesion molecule
- FA, folic acid
- GA, gambogic acid
- H&E, hematoxylin and eosin
- HRP, homologous restriction protein
- MCP, membrane cofactor protein
- MNCs, magnetic nanoclusters
- MNs, magnetic nanoparticles
- MPS, mononuclear phagocyte system
- MRI, magnetic resonance imaging
- MSNs, mesoporous silica nanoparticles
- Membrane
- NIR, near-infrared radiation
- Nanoparticles
- PAI, photoacoustic imaging
- PBS, phosphate buffered saline
- PCL, poly(caprolactone)
- PDT, photodynamic therapy
- PEG, polyethylene glycol
- PFCs, perfluorocarbons
- PLA, poly(lactide acid)
- PLGA, poly(d,l-lactide-co-glycolide)
- PPy, polypyrrole
- PS, photosensitizers
- PTT, photothermal therapy
- PTX, paclitaxel
- RBCM-NPs, RBCM-coated nanoparticles
- RBCMs, RBC membranes
- RBCs, red blood cells
- RES, reticuloendothelial system
- ROS, reactive oxygen species
- RVs, RBCM-derived vesicles
- Red blood cells
- SEM, scanning electron microscopy
- SIRPα, signal-regulatory protein alpha
- TEM, transmission electron microscopy
- TEMPO, 2,2,6,6-tetramethylpiperidin-1-yl oxyl
- TPP, triphenylphosphonium
- UCNPs, upconversion nanoparticles
- UV, ultraviolet
- rHuPH20, recombinant hyaluronidase, PH20
Collapse
Affiliation(s)
| | | | | | | | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
68
|
Yu W, He X, Yang Z, Yang X, Xiao W, Liu R, Xie R, Qin L, Gao H. Sequentially responsive biomimetic nanoparticles with optimal size in combination with checkpoint blockade for cascade synergetic treatment of breast cancer and lung metastasis. Biomaterials 2019; 217:119309. [PMID: 31271855 DOI: 10.1016/j.biomaterials.2019.119309] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/15/2019] [Accepted: 06/25/2019] [Indexed: 12/30/2022]
Abstract
Recently, photodynamic therapy (PDT) emerges as a promising way to initiate immune response and being used in combination with chemotherapy. However, the antitumor effect is restricted due to the poor tumor penetration and retention, premature drug release and immunosuppressive environment of tumor sites. And as the size of nanoparticles plays a key role in drug delivery, series of hyaluronidase-responsive size-reducible biomimetic nanoparticles (mCAuNCs@HA) with different initial sizes are synthesized, and the optimal size of 150 nm is screened out because of the best blood circulation, tumor penetration and retention. Then the photosensitizer pheophorbide A and ROS-responsive paclitaxel dimer prodrug (PXTK) are co-loaded to facilitate on-demand drug release. The hydrolysis byproduct cinnamaldehyde in turn stimulates the ROS production by mitochondria, which compensates for the ROS consumed in the hydrolysis process. Anti-PD-L1 peptide (dPPA) is furthered loaded to alleviate the immunosuppressive environment of tumor and enhance the function of cytotoxic T lymphocytes activated by PDT-induced immunogenic cell death. The combination therapy activates CD4+, CD8+ T cells and NK cells and enhances secretion of cytokines (TNF-α and IL-12) with tumor inhibition rate increased to 84.2% and no metastasis is observed, providing a viable combination therapy for better anti-tumor and anti-metastasis efficacy.
Collapse
Affiliation(s)
- Wenqi Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Xueqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Zhihang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Xiaotong Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Wei Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Rui Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Rou Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
69
|
Que X, Su J, Guo P, Kamal Z, Xu E, Liu S, Chen J, Qiu M. Study on preparation, characterization and multidrug resistance reversal of red blood cell membrane-camouflaged tetrandrine-loaded PLGA nanoparticles. Drug Deliv 2019; 26:199-207. [PMID: 30835586 PMCID: PMC6407593 DOI: 10.1080/10717544.2019.1573861] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The multidrug resistance in tumor (MDR) is a major barrier to efficient cancer therapy. Modern pharmacological studies have proven that tetrandrine (TET) has great potential in reversing MDR. However, it has a series of medication problems in clinic such as poor water solubility, low oral bioavailability and short half-life in vivo. Aiming at the above problems, red blood cell membrane-camouflaged TET-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (RPTNs) had been developed. The RPTNs had spherical shell-core double layer structure with average particle size of 164.1 ± 1.65 nm and encapsulation efficiency of 84.1% ± 0.41%. Compared with TET-PLGA nanoparticles (PTNs), the RPTNs reduced RAW 264.7 macrophages’ swallowing by 32% due to its retention of natural membrane proteins. The cumulative drug release of RPTNs was 81.88% within 120 h. And pharmacokinetic study showed that the blood half-life of RPTNs was 19.38 h, which was 2.95 times of free drug. When RPTNs of 2 μg/mL TET were administered in combination with adriamycin (ADR), significant MDR reversal effect was observed in drug-resistant cells MCF-7/ADR. In a word, the RPTNs hold potential to improve its efficacy and broaden its clinical application.
Collapse
Affiliation(s)
- Xiao Que
- a School of Pharmacy , Shanghai Jiao Tong University , Shanghai , China
| | - Jing Su
- a School of Pharmacy , Shanghai Jiao Tong University , Shanghai , China
| | - Pengcheng Guo
- a School of Pharmacy , Shanghai Jiao Tong University , Shanghai , China
| | - Zul Kamal
- b Department of Pharmacy , Shaheed Benazir Bhutto University , Sheringal Dir (Upper) , Pakistan
| | - Enge Xu
- a School of Pharmacy , Shanghai Jiao Tong University , Shanghai , China
| | - Siyu Liu
- a School of Pharmacy , Shanghai Jiao Tong University , Shanghai , China
| | | | - Mingfeng Qiu
- a School of Pharmacy , Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
70
|
Xuan M, Shao J, Li J. Cell membrane-covered nanoparticles as biomaterials. Natl Sci Rev 2019; 6:551-561. [PMID: 34691904 PMCID: PMC8291551 DOI: 10.1093/nsr/nwz037] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/12/2019] [Accepted: 02/27/2019] [Indexed: 12/19/2022] Open
Abstract
Surface engineering of synthetic carriers is an essential and important strategy for drug delivery in vivo. However, exogenous properties make synthetic nanosystems invaders that easily trigger the passive immune clearance mechanism, increasing the retention effect caused by the reticuloendothelial systems and bioadhesion, finally leading to low therapeutic efficacy and toxic effects. Recently, a cell membrane cloaking technique has been reported as a novel interfacing approach from the biological/immunological perspective, and has proved useful for improving the performance of synthetic nanocarriers in vivo. After cell membrane cloaking, nanoparticles not only acquire the physiochemical properties of natural cell membranes but also inherit unique biological functions due to the presence of membrane-anchored proteins, antigens, and immunological moieties. The derived biological properties and functions, such as immunosuppressive capability, long circulation time, and targeted recognition integrated in synthetic nanosystems, have enhanced their potential in biomedicine in the future. Here, we review the cell membrane-covered nanosystems, highlight their novelty, introduce relevant biomedical applications, and describe the future prospects for the use of this novel biomimetic system constructed from a combination of cell membranes and synthetic nanomaterials.
Collapse
Affiliation(s)
- Mingjun Xuan
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Jingxin Shao
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing 100190, China
| |
Collapse
|
71
|
Mu Q, Wang H, Gu X, Stephen ZR, Yen C, Chang FC, Dayringer CJ, Zhang M. Biconcave Carbon Nanodisks for Enhanced Drug Accumulation and Chemo-Photothermal Tumor Therapy. Adv Healthc Mater 2019; 8:e1801505. [PMID: 30856295 PMCID: PMC6483846 DOI: 10.1002/adhm.201801505] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/18/2019] [Indexed: 12/11/2022]
Abstract
It is considered a significant challenge to construct nanocarriers that have high drug loading capacity and can overcome physiological barriers to deliver efficacious amounts of drugs to solid tumors. Here, the development of a safe, biconcave carbon nanodisk to address this challenge for treating breast cancer is reported. The nanodisk demonstrates fluorescent imaging capability, an exceedingly high loading capacity (947.8 mg g-1 , 94.78 wt%) for doxorubicin (DOX), and pH-responsive drug release. It exhibits a higher uptake rate by tumor cells and greater accumulation in tumors in a mouse model than its carbon nanosphere counterpart. In addition, the nanodisk absorbs and transforms near-infrared (NIR) light to heat, which enables simultaneous NIR-responsive drug release for chemotherapy and generation of thermal energy for tumor cell destruction. Notably, this NIR-activated dual therapy demonstrates a near complete suppression of tumor growth in a mouse model of triple-negative breast cancer when DOX-loaded nanodisks are administered systemically.
Collapse
Affiliation(s)
- Qingxin Mu
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Hui Wang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
- The Anhui Key Laboratory of Condensed Matter Physics at Extreme Conditions, High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
| | - Xinyu Gu
- Department of Biochemistry, University of Washington Seattle, Washington, DC, 98195, USA
| | - Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Charles Yen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Christopher J Dayringer
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington, DC, 98195, USA
| |
Collapse
|
72
|
Perera AS, Coppens MO. Re-designing materials for biomedical applications: from biomimicry to nature-inspired chemical engineering. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2019; 377:20180268. [PMID: 30967073 PMCID: PMC6335285 DOI: 10.1098/rsta.2018.0268] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/30/2018] [Indexed: 05/24/2023]
Abstract
Gathering inspiration from nature for the design of new materials, products and processes is a topic gaining rapid interest among scientists and engineers. In this review, we introduce the concept of nature-inspired chemical engineering (NICE). We critically examine how this approach offers advantages over straightforward biomimicry and distinguishes itself from bio-integrated design, as a systematic methodology to present innovative solutions to challenging problems. The scope of application of the nature-inspired approach is demonstrated via examples from the field of biomedicine, where much of the inspiration is still more narrowly focused on imitation or bio-integration. We conclude with an outlook on prospective future applications, offered by the more systematic and mechanistically based NICE approach, complemented by rapid progress in manufacturing, computation and robotics. This article is part of the theme issue 'Bioinspired materials and surfaces for green science and technology'.
Collapse
Affiliation(s)
- Ayomi S. Perera
- Centre for Nature Inspired Engineering, Department of Chemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
- Department of Chemical and Pharmaceutical Sciences, Kingston University London, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Marc-Olivier Coppens
- Centre for Nature Inspired Engineering, Department of Chemical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| |
Collapse
|
73
|
Wan X, Zhang S, Wang F, Fan W, Wu C, Mao K, Wang H, Hu Z, Yang YG, Sun T. Red blood cell-derived nanovesicles for safe and efficient macrophage-targeted drug delivery in vivo. Biomater Sci 2019; 7:187-195. [PMID: 30421747 DOI: 10.1039/c8bm01258j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RBC-derived nanovesicles are effective hydrophilic drug carriers and can effectively deliver drugs into macrophages both in vitro and in vivo.
Collapse
|
74
|
Kroll AV, Jiang Y, Zhou J, Holay M, Fang RH, Zhang L. Biomimetic Nanoparticle Vaccines for Cancer Therapy. ADVANCED BIOSYSTEMS 2019; 3:e1800219. [PMID: 31728404 PMCID: PMC6855307 DOI: 10.1002/adbi.201800219] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Indexed: 12/25/2022]
Abstract
It is currently understood that, in order for a tumor to successfully grow, it must evolve means of evading immune surveillance. In the past several decades, researchers have leveraged increases in our knowledge of tumor immunology to develop therapies capable of augmenting endogenous immunity and eliciting strong antitumor responses. In particular, the goal of anticancer vaccination is to train the immune system to properly utilize its own resources in the fight against cancer. Although attractive in principle, there are currently only limited examples of anticancer vaccines that have been successfully translated to the clinic. Recently, there has been a significant push towards the use of nanotechnology for designing vaccine candidates that exhibit enhanced potency and specificity. In this progress report, we discuss recent developments in the field of anticancer nanovaccines. By taking advantage of the flexibility offered by nanomedicine to purposefully program immune responses, this new generation of vaccines has the potential to address many of the hurdles facing traditional platforms. A specific emphasis is placed on the emergence of cell membrane-coated nanoparticles, a novel biomimetic platform that can be used to generate personalized nanovaccines that elicit strong, multi-antigenic antitumor responses.
Collapse
Affiliation(s)
- Ashley V Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yao Jiang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jiarong Zhou
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maya Holay
- 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
|
75
|
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: 1.3] [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
|
76
|
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: 65] [Impact Index Per Article: 10.8] [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
|
77
|
Vankayala R, Hwang KC. Near-Infrared-Light-Activatable Nanomaterial-Mediated Phototheranostic Nanomedicines: An Emerging Paradigm for Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706320. [PMID: 29577458 DOI: 10.1002/adma.201706320] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/11/2017] [Indexed: 05/22/2023]
Abstract
Cancer is one of the most deadly diseases threatening the lives of humans. Although many treatment methods have been developed to tackle cancer, each modality of cancer treatment has its own limitations and drawbacks. The development of minimally invasive treatment modalities for cancers remains a great challenge. Near-infrared (NIR) light-activated nanomaterial-mediated phototherapies, including photothermal and photodynamic therapies, provide an alternative means for spatially and temporally controlled minimally invasive treatments of cancers. Nanomaterials can serve as nanocargoes for the delivery of chemo-drugs, diagnostic contrast reagents, and organic photosensitizers, and can be used to directly generate heat or reactive oxygen species for the treatment of tumors without the need for organic photosensitizers with NIR-light irradiation. Here, current progress in NIR-light-activated nanomaterial-mediated photothermal therapy and photodynamic therapy is summarized. Furthermore, the effects of size, shape, and surface functionalities of nanomaterials on intracellular uptake, macrophage clearance, biodistribution, cytotoxicities, and biomedical efficacies are discussed. The use of various types of nanomaterials, such as gold nanoparticles, carbon nanotubes, graphene, and many other inorganic nanostructures, in combination with diagnostic and therapeutic modalities for solid tumors, is briefly reviewed.
Collapse
Affiliation(s)
- Raviraj Vankayala
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| | - Kuo Chu Hwang
- Department of Chemistry, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| |
Collapse
|
78
|
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: 1004] [Impact Index Per Article: 167.3] [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
|
79
|
Affiliation(s)
- Xiao Han
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
80
|
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: 10.8] [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
|
81
|
Angsantikul P, Fang RH, Zhang L. Toxoid Vaccination against Bacterial Infection Using Cell Membrane-Coated Nanoparticles. Bioconjug Chem 2017; 29:604-612. [PMID: 29241006 DOI: 10.1021/acs.bioconjchem.7b00692] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As nanoparticles exhibit unique properties attractive for vaccine development, they have been progressively implemented as antigen delivery platforms and immune potentiators. Recently, cell membrane-coated nanoparticles have provided a novel approach for intercepting and neutralizing bacterial toxins by leveraging their natural affinity to cellular membranes. Such toxin-nanoparticle assemblies, termed nanotoxoids, allow rapid loading of different types of toxins and have been investigated for their ability to effectively confer protection against bacterial infection. This topical review will cover the current progress in antibacterial vaccine nanoformulations and highlight the nanotoxoid platform as a novel class of nanoparticulate vaccine. We aim to provide insights into the potential of nanotoxoids as a platform that is facile to implement and can be broadly applied to help address the rising threat of super pathogens.
Collapse
Affiliation(s)
- Pavimol Angsantikul
- Department of NanoEngineering and Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
82
|
Ettelt V, Ekat K, Kämmerer PW, Kreikemeyer B, Epple M, Veith M. Streptavidin-coated surfaces suppress bacterial colonization by inhibiting non-specific protein adsorption. J Biomed Mater Res A 2017; 106:758-768. [DOI: 10.1002/jbm.a.36276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Volker Ettelt
- Laboratory of Biophysics, Faculty of Applied Natural Sciences; Westphalian University of Applied Sciences; Recklinghausen D-45665 Germany
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), Faculty of Chemistry; University of Duisburg-Essen; Essen D-45141 Germany
| | - Katharina Ekat
- Department Cell Biology; University Medical Center Rostock; Rostock D-18057 Germany
- Department of Operative Dentistry and Periodontology; University Medical Center Rostock; Rostock D-18057 Germany
| | - Peer W. Kämmerer
- Department of Oral, Maxillofacial and Plastic Surgery; University Medical Center Rostock; Rostock D-18057 Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, University Medical Center Rostock; Rostock D-18057 Germany
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), Faculty of Chemistry; University of Duisburg-Essen; Essen D-45141 Germany
| | - Michael Veith
- Laboratory of Biophysics, Faculty of Applied Natural Sciences; Westphalian University of Applied Sciences; Recklinghausen D-45665 Germany
| |
Collapse
|
83
|
Fan W, Yung B, Huang P, Chen X. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem Rev 2017; 117:13566-13638. [DOI: 10.1021/acs.chemrev.7b00258] [Citation(s) in RCA: 1059] [Impact Index Per Article: 151.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wenpei Fan
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
- Key
Laboratory of Optoelectronic Devices and Systems of Ministry of Education
and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Bryant Yung
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peng Huang
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyuan Chen
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
84
|
Parodi A, Molinaro R, Sushnitha M, Evangelopoulos M, Martinez JO, Arrighetti N, Corbo C, Tasciotti E. Bio-inspired engineering of cell- and virus-like nanoparticles for drug delivery. Biomaterials 2017; 147:155-168. [PMID: 28946131 DOI: 10.1016/j.biomaterials.2017.09.020] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/13/2017] [Accepted: 09/17/2017] [Indexed: 12/25/2022]
Abstract
The engineering of future generations of nanodelivery systems aims at the creation of multifunctional vectors endowed with improved circulation, enhanced targeting and responsiveness to the biological environment. Moving past purely bio-inert systems, researchers have begun to create nanoparticles capable of proactively interacting with the biology of the body. Nature offers a wide-range of sources of inspiration for the synthesis of more effective drug delivery platforms. Because the nano-bio-interface is the key driver of nanoparticle behavior and function, the modification of nanoparticles' surfaces allows the transfer of biological properties to synthetic carriers by imparting them with a biological identity. Modulation of these surface characteristics governs nanoparticle interactions with the biological barriers they encounter. Building off these observations, we provide here an overview of virus- and cell-derived biomimetic delivery systems that combine the intrinsic hallmarks of biological membranes with the delivery capabilities of synthetic carriers. We describe the features and properties of biomimetic delivery systems, recapitulating the distinctive traits and functions of viruses, exosomes, platelets, red and white blood cells. By mimicking these biological entities, we will learn how to more efficiently interact with the human body and refine our ability to negotiate with the biological barriers that impair the therapeutic efficacy of nanoparticles.
Collapse
Affiliation(s)
- Alessandro Parodi
- Department of Pharmacology, University of Illinois, Chicago College of Medicine, Chicago, IL, USA
| | - Roberto Molinaro
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manuela Sushnitha
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA; Department of Bioengineering, Rice University, Houston, TX, USA
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA
| | - Jonathan O Martinez
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA
| | - Noemi Arrighetti
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA; Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale per Lo Studio e La Cura Dei Tumori, Milan, Italy
| | - Claudia Corbo
- Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, MA, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute (HMRI), Houston, TX, USA; Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
85
|
Harisa GI, Badran MM, Alanazi FK, Attia SM. An overview of nanosomes delivery mechanisms: trafficking, orders, barriers and cellular effects. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:669-679. [DOI: 10.1080/21691401.2017.1354301] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Gamaleldin I. Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Biochemistry, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mohamed M. Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Fars K. Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
86
|
Paluri SLA, Ryan JD, Lam NH, Nepal D, Sizemore IE. Analytical-Based Methodologies for Examining the In Vitro Absorption, Distribution, Metabolism, and Elimination (ADME) of Silver Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1603093. [PMID: 28440049 DOI: 10.1002/smll.201603093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/31/2017] [Indexed: 06/07/2023]
Abstract
The clinical applications of silver nanoparticles (AgNPs) remain limited due to the lack of well-established methodologies for studying their nanokinetics. Hereby, the primary goal is to adapt a suite of analytical-based methodologies for examining the in vitro absorption, distribution, metabolism, and elimination of AgNPs. Vero 76 and HEK 293 cells are exposed to ≈10-nm spherical AgNPs+ and AgNPs- at relevant concentrations (0-300 µg mL-1 ) and times (4-48 h). Absorption: Inductively coupled plasma optical emission spectroscopy (ICP-OES) demonstrates that the two AgNP formulations are not bioequivalent. For example, different bioavailabilities (Cmaximum < 20.7 ± 4% and 6.82 ± 0.4%), absorption times (Tmaximum > 48 and ≈24 h), and absorption rate laws (first- and zeroth-order at 300 µg mL-1 ) are determined in Vero 76 for AgNPs+ and AgNPs- , respectively. Distribution: Raman and CytoViva hyperspectral imaging show different cellular localizations for AgNPs+ and AgNPs- . Metabolism: Cloud point extraction (CPE)-tangential flow filtration (TFF) reveal that ≤ 11% ± 4% of the administered, sublethal AgNPs release Ag+ and contribute to the observed cytotoxicity. Elimination: ICP-OES-CPE suggests that AgNPs are cleared via exocytosis.
Collapse
Affiliation(s)
- Sesha L A Paluri
- Department of Chemistry, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435-0001, USA
- Biomedical Sciences Ph.D. program, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435-0001, USA
| | - John D Ryan
- Department of Chemistry, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435-0001, USA
| | - Nhi H Lam
- Department of Chemistry, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435-0001, USA
| | - Dhriti Nepal
- Air Force Research Laboratory, Wright Patterson Air Force Base, 2941 Hobson Way, OH, 45433, USA
| | - Ioana E Sizemore
- Department of Chemistry, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435-0001, USA
- Biomedical Sciences Ph.D. program, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH, 45435-0001, USA
| |
Collapse
|
87
|
Fang RH, Jiang Y, Fang JC, Zhang L. Cell membrane-derived nanomaterials for biomedical applications. Biomaterials 2017; 128:69-83. [PMID: 28292726 PMCID: PMC5417338 DOI: 10.1016/j.biomaterials.2017.02.041] [Citation(s) in RCA: 305] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 02/06/2023]
Abstract
The continued evolution of biomedical nanotechnology has enabled clinicians to better detect, prevent, manage, and treat human disease. In order to further push the limits of nanoparticle performance and functionality, there has recently been a paradigm shift towards biomimetic design strategies. By taking inspiration from nature, the goal is to create next-generation nanoparticle platforms that can more effectively navigate and interact with the incredibly complex biological systems that exist within the body. Of great interest are cellular membranes, which play essential roles in biointerfacing, self-identification, signal transduction, and compartmentalization. In this review, we explore the major ways in which researchers have directly leveraged cell membrane-derived biomaterials for the fabrication of novel nanotherapeutics and nanodiagnostics. Such emerging technologies have the potential to significantly advance the field of nanomedicine, helping to improve upon traditional modalities while also enabling novel applications.
Collapse
Affiliation(s)
- Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yao Jiang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jean C 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
|
88
|
Zhang Z, Qian H, Yang M, Li R, Hu J, Li L, Yu L, Liu B, Qian X. Gambogic acid-loaded biomimetic nanoparticles in colorectal cancer treatment. Int J Nanomedicine 2017; 12:1593-1605. [PMID: 28280328 PMCID: PMC5339001 DOI: 10.2147/ijn.s127256] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Gambogic acid (GA) is expected to be a potential new antitumor drug, but its poor aqueous solubility and inevitable side effects limit its clinical application. Despite these inhe rent defects, various nanocarriers can be used to promote the solubility and tumor targeting of GA, improving antitumor efficiency. In addition, a cell membrane-coated nanoparticle platform that was reported recently, unites the customizability and flexibility of a synthetic copolymer, as well as the functionality and complexity of natural membrane, and is a new synthetic biomimetic nanocarrier with improved stability and biocompatibility. Here, we combined poly(lactic-co-glycolic acid) (PLGA) with red blood-cell membrane (RBCm), and evaluated whether GA-loaded RBCm nanoparticles can retain and improve the antitumor efficacy of GA with relatively lower toxicity in colorectal cancer treatment compared with free GA. We also confirmed the stability, biocompatibility, passive targeting, and few side effects of RBCm-GA/PLGA nanoparticles. We expect to provide a new drug carrier in the treatment of colorectal cancer, which has strong clinical application prospects. In addition, the potential antitumor drug GA and other similar drugs could achieve broader clinical applications via this biomimetic nanocarrier.
Collapse
Affiliation(s)
- Zhen Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine
| | - Hanqing Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Mi Yang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Rutian Li
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Jing Hu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine
| | - Li Li
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine
| | - Lixia Yu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Baorui Liu
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine; Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| | - Xiaoping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Traditional Chinese Medicine; Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute, Nanjing University, Nanjing, China
| |
Collapse
|
89
|
Mononuclear phagocytes as a target, not a barrier, for drug delivery. J Control Release 2017; 259:53-61. [PMID: 28108325 DOI: 10.1016/j.jconrel.2017.01.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/06/2017] [Accepted: 01/16/2017] [Indexed: 02/07/2023]
Abstract
Mononuclear phagocytes have been generally recognized as a barrier to drug delivery. Recently, a new understanding of mononuclear phagocytes (MPS) ontogeny has surfaced and their functions in disease have been unveiled, demonstrating the need for re-evaluation of perspectives on mononuclear phagocytes in drug delivery. In this review, we described mononuclear phagocyte biology and focus on their accumulation mechanisms in disease sites with explanations of monocyte heterogeneity. In the 'MPS as a barrier' section, we summarized recent studies on mechanisms to avoid phagocytosis based on two different biological principles: protein adsorption and self-recognition. In the 'MPS as a target' section, more detailed descriptions were given on mononuclear phagocyte-targeted drug delivery systems and their applications to various diseases. Collectively, we emphasize in this review that mononuclear phagocytes are potent targets for future drug delivery systems. Mononuclear phagocyte-targeted delivery systems should be created with an understanding of mononuclear phagocyte ontogeny and pathology. Each specific subset of phagocytes should be targeted differently by location and function for improved disease-drug delivery while avoiding RES clearance such as Kupffer cells and splenic macrophages.
Collapse
|
90
|
Combination therapy with doxorubicin-loaded galactosylated poly(ethyleneglycol)-lithocholic acid to suppress the tumor growth in an orthotopic mouse model of liver cancer. Biomaterials 2016; 116:130-144. [PMID: 27914985 DOI: 10.1016/j.biomaterials.2016.11.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/12/2016] [Accepted: 11/24/2016] [Indexed: 12/26/2022]
Abstract
Despite advances in technology, neither conventional anti-cancer drugs nor current nanoparticle (NP) drugs have gained substantial success in cancer treatment. While conventional chemotherapy drugs have several limitations such as low potency, poor in vivo stability and limited bioavailability, non-specific targeting of NP drugs diminishes their potency at actual target sites. In addition, the development of drug resistance to anti-cancer drugs is another challenging problem. To overcome these limitations, we aimed to develop a polymer-drug conjugate, which functions as an active NP drug and drug carrier both, to deliver a chemotherapeutic drug for combination therapy. Accordingly, we made targeting NP carrier of lithocholic acid-poly(ethylene glycol)-lactobionic acid (LPL) loading doxorubicin (Dox) to produce Dox/LPL NPs. The cellular uptake of Dox/LPL NPs was relatively higher in human liver cancer cell line (SK-HEP-1) due to galactose ligand-asialoglycoprotein receptor interaction. Consequently, the cellular uptake of Dox/LPL NPs led to massive cell death of SK-HEP-1 cells by two different mechanisms, particularly apoptotic activity by LPL and mitotic catastrophe by Dox. Most importantly, Dox/LPL NPs, when administered to orthotopic xenograft model of liver cancer, greatly reduced proliferation, invasion, migration, and angiogenesis of liver tumor in vivo. Thus, this study exemplifies the superiority of combination therapy over individual NP drug or conventional small molecule drug for cancer therapy. Overall, we present a promising approach of combinatorial therapy to inhibit the hepatic tumor growth and metastasis in the orthotopic xenograft model mice, thus representing an effective weapon for cancer treatment.
Collapse
|
91
|
Dehaini D, Fang RH, Zhang L. Biomimetic strategies for targeted nanoparticle delivery. Bioeng Transl Med 2016; 1:30-46. [PMID: 29313005 PMCID: PMC5689512 DOI: 10.1002/btm2.10004] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 01/02/2023] Open
Abstract
Nanoparticle‐based drug delivery and imaging platforms have become increasingly popular over the past several decades. Among different design parameters that can affect their performance, the incorporation of targeting functionality onto nanoparticle surfaces has been a widely studied subject. Targeted formulations have the ability to improve efficacy and function by positively modulating tissue localization. Many methods exist for creating targeted nanoformulations, including the use of custom biomolecules such as antibodies or aptamers. More recently, a great amount of focus has been placed on biomimetic targeting strategies that leverage targeting interactions found directly in nature. Such strategies, which have been painstakingly selected over time by the process of evolution to maximize functionality, oftentimes enable scientists to forgo the specialized discovery processes associated with many traditional ligands and help to accelerate development of novel nanoparticle formulations. In this review, we categorize and discuss in‐depth recent works in this growing field of bioinspired research.
Collapse
Affiliation(s)
- Diana Dehaini
- Dept. of NanoEngineering and Moores Cancer Center University of California San Diego, La Jolla CA 92093
| | - Ronnie H Fang
- Dept. of NanoEngineering and Moores Cancer Center University of California San Diego, La Jolla CA 92093
| | - Liangfang Zhang
- Dept. of NanoEngineering and Moores Cancer Center University of California San Diego, La Jolla CA 92093
| |
Collapse
|
92
|
Xu P, Wang R, Wang X, Ouyang J. Recent advancements in erythrocytes, platelets, and albumin as delivery systems. Onco Targets Ther 2016; 9:2873-84. [PMID: 27274282 PMCID: PMC4876107 DOI: 10.2147/ott.s104691] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In the past few years, nanomaterial-based drug delivery systems have been applied to enhance the efficacy of therapeutics and to alleviate negative effects through the controlled delivery of targeting and releasing agents. However, few drug carriers can achieve high targeting efficacy, even when targeting modalities and surface markers are introduced. Immunological problems have also limited their wide applications. Biological drug delivery systems, such as erythrocytes, platelets, and albumin, have been extensively investigated because of their unique properties. In this review, erythrocytes, platelets, and albumin are described as efficient drug delivery systems. Their properties, applications, advantages, and limitations in disease treatment are explained. This review confirms that these systems can be used to facilitate a specific, biocompatible, and smart drug delivery.
Collapse
Affiliation(s)
- Peipei Xu
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Ruju Wang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Medical School, Southeast University, Nanjing, People's Republic of China
| | - Xiaohui Wang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Jian Ouyang
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| |
Collapse
|
93
|
Luk BT, Fang RH, Hu CMJ, Copp JA, Thamphiwatana S, Dehaini D, Gao W, Zhang K, Li S, Zhang L. Safe and Immunocompatible Nanocarriers Cloaked in RBC Membranes for Drug Delivery to Treat Solid Tumors. Am J Cancer Res 2016; 6:1004-11. [PMID: 27217833 PMCID: PMC4876624 DOI: 10.7150/thno.14471] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 12/11/2015] [Indexed: 12/23/2022] Open
Abstract
The therapeutic potential of nanoparticle-based drug carriers depends largely on their ability to evade the host immune system while delivering their cargo safely to the site of action. Of particular interest are simple strategies for the functionalization of nanoparticle surfaces that are both inherently safe and can also bestow immunoevasive properties, allowing for extended blood circulation times. Here, we evaluated a recently reported cell membrane-coated nanoparticle platform as a drug delivery vehicle for the treatment of a murine model of lymphoma. These biomimetic nanoparticles, consisting of a biodegradable polymeric material cloaked with natural red blood cell membrane, were shown to efficiently deliver a model chemotherapeutic, doxorubicin, to solid tumor sites for significantly increased tumor growth inhibition compared with conventional free drug treatment. Importantly, the nanoparticles also showed excellent immunocompatibility as well as an advantageous safety profile compared with the free drug, making them attractive for potential translation. This study demonstrates the promise of using a biomembrane-coating approach as the basis for the design of functional, safe, and immunocompatible nanocarriers for cancer drug delivery.
Collapse
|
94
|
Abstract
The immune system is an incredibly complex biological network that plays a significant role in almost all disease pathogenesis. With an increased understanding of how this vital system operates, there has been a great emphasis on leveraging, manipulating, and/or supplementing endogenous immunity to better prevent or treat different disease states. More recently, the advent of nanotechnology has ushered in a plethora of new nanoparticle-based platforms that can be used to improve existing immunomodulation modalities. As the ability to engineer at the nanoscale becomes increasingly sophisticated, nanoparticles can be finely tuned to effect the desired immune responses, leading to exciting new avenues for addressing pressing issues in public health. In this review, we give an overview of the different areas in which nanoparticle technology has been applied toward modulating the immune system and highlight the recent advances within each.
Collapse
Affiliation(s)
- Ronnie H Fang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093;
| | - Liangfang Zhang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
95
|
Zhao Y, He G, Guo W, Bao L, Yi M, Gong Y, Zhang S. Self-assembled micelles prepared from amphiphilic copolymers bearing cell outer membrane phosphorylcholine zwitterions for a potential anti-phagocytic clearance carrier. Polym Chem 2016. [DOI: 10.1039/c6py00845c] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A versatile strategy using amphiphilic copolymers to prepare micelles with cell membrane mimetic phosphorylcholine shell and PCL core showing potential anti-phagocytic clearance properties was reported.
Collapse
Affiliation(s)
- Yuping Zhao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| | - Guiqiang He
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| | - Weihong Guo
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| | - Lili Bao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| | - Meijun Yi
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| | - Yongkuan Gong
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| | - Shiping Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education
- College of Chemistry and Materials Science
- Northwest University
- Xi'an 710127
- PR China
| |
Collapse
|
96
|
Pereira P, Correia A, Gama FM. In Vivo Imaging of Glycol Chitosan-Based Nanogel Biodistribution. Macromol Biosci 2015; 16:432-40. [DOI: 10.1002/mabi.201500267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 11/11/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Paula Pereira
- Centre of Biological Engineering; University of Minho; Campus de Gualtar; 4710-057 Braga Portugal
| | - Alexandra Correia
- I3S - Instituto de Investigação e Inovação em Saúde; Universidade do Porto and IBMC - Instituto de Biologia Molecular e Celular Rua do Campo Alegre; 4099-003 Porto Portugal
| | - Francisco M. Gama
- Centre of Biological Engineering; University of Minho; Campus de Gualtar; 4710-057 Braga Portugal
| |
Collapse
|
97
|
Rao L, Bu LL, Xu JH, Cai B, Yu GT, Yu X, He Z, Huang Q, Li A, Guo SS, Zhang WF, Liu W, Sun ZJ, Wang H, Wang TH, Zhao XZ. Red Blood Cell Membrane as a Biomimetic Nanocoating for Prolonged Circulation Time and Reduced Accelerated Blood Clearance. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:6225-36. [PMID: 26488923 DOI: 10.1002/smll.201502388] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/06/2015] [Indexed: 05/18/2023]
Abstract
For decades, poly(ethylene glycol) (PEG) has been widely incorporated into nanoparticles for evading immune clearance and improving the systematic circulation time. However, recent studies have reported a phenomenon known as "accelerated blood clearance (ABC)" where a second dose of PEGylated nanomaterials is rapidly cleared when given several days after the first dose. Herein, we demonstrate that natural red blood cell (RBC) membrane is a superior alternative to PEG. Biomimetic RBC membrane-coated Fe(3)O(4) nanoparticles (Fe(3)O(4) @RBC NPs) rely on CD47, which is a "don't eat me" marker on the RBC surface, to escape immune clearance through interactions with the signal regulatory protein-alpha (SIRP-α) receptor. Fe(3)O(4) @RBC NPs exhibit extended circulation time and show little change between the first and second doses, with no ABC suffered. In addition, the administration of Fe(3)O(4) @RBC NPs does not elicit immune responses on neither the cellular level (myeloid-derived suppressor cells (MDSCs)) nor the humoral level (immunoglobulin M and G (IgM and IgG)). Finally, the in vivo toxicity of these cell membrane-camouflaged nanoparticles is systematically investigated by blood biochemistry, hematology testing, and histology analysis. These findings are significant advancements toward solving the long-existing clinical challenges of developing biomaterials that are able to resist both immune response and rapid clearance.
Collapse
Affiliation(s)
- Lang Rao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| | - Lin-Lin Bu
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Key Laboratory of Oral Biomedicine of Ministry of Education, Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jun-Hua Xu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Bo Cai
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| | - Guang-Tao Yu
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Key Laboratory of Oral Biomedicine of Ministry of Education, Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Xiaolei Yu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| | - Zhaobo He
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| | - Qinqin Huang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| | - Andrew Li
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, 21218, MA, USA
| | - Shi-Shang Guo
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| | - Wen-Feng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Key Laboratory of Oral Biomedicine of Ministry of Education, Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Wei Liu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, 21218, MA, USA
| | - Zhi-Jun Sun
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Key Laboratory of Oral Biomedicine of Ministry of Education, Department of Oral Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Hao Wang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Tza-Huei Wang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, 21218, MA, USA
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
98
|
Beck-Broichsitter M, Nicolas J, Couvreur P. Design attributes of long-circulating polymeric drug delivery vehicles. Eur J Pharm Biopharm 2015; 97:304-17. [PMID: 25857838 DOI: 10.1016/j.ejpb.2015.03.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/11/2015] [Accepted: 03/23/2015] [Indexed: 02/03/2023]
Abstract
Following systemic administration polymeric drug delivery vehicles allow for a controlled and targeted release of the encapsulated medication at the desired site of action. For an elevated and organ specific accumulation of their cargo, nanocarriers need to avoid opsonization, activation of the complement system and uptake by macrophages of the mononuclear phagocyte system. In this respect, camouflaged vehicles revealed a delayed elimination from systemic circulation and an improved target organ deposition. For instance, a steric shielding of the carrier surface by poly(ethylene glycol) substantially decreased interactions with the biological environment. However, recent studies disclosed possible deficits of this approach, where most notably, poly(ethylene glycol)-modified drug delivery vehicles caused significant immune responses. At present, identification of novel potential carrier coating strategies facilitating negligible immune reactions is an emerging field of interest in drug delivery research. Moreover, physical carrier properties including geometry and elasticity seem to be very promising design attributes to surpass numerous biological barriers, in order to improve the efficacy of the delivered medication.
Collapse
Affiliation(s)
- Moritz Beck-Broichsitter
- Institut Galien UMR CNRS 8612, Faculté de Pharmacie, Université Paris-Sud XI, Châtenay-Malabry, France
| | - Julien Nicolas
- Institut Galien UMR CNRS 8612, Faculté de Pharmacie, Université Paris-Sud XI, Châtenay-Malabry, France
| | - Patrick Couvreur
- Institut Galien UMR CNRS 8612, Faculté de Pharmacie, Université Paris-Sud XI, Châtenay-Malabry, France.
| |
Collapse
|
99
|
Zhao J, Chai YD, Zhang J, Huang PF, Nakashima K, Gong YK. Long circulating micelles of an amphiphilic random copolymer bearing cell outer membrane phosphorylcholine zwitterions. Acta Biomater 2015; 16:94-102. [PMID: 25637066 DOI: 10.1016/j.actbio.2015.01.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 12/22/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022]
Abstract
Polymeric micelles with cell outer membrane mimetic structure were prepared in water from amphiphilic random copolymers bearing both the hydrophilic phosphorylcholine zwitterions and hydrophobic octadecyl side chains of cell outer membrane. The polymeric micelles showed sizes ranging from 80 nm to 120 nm in hydrodynamic diameter and zeta-potentials from -6.4 mV to -2.4 mV by dynamic light scattering measurements. The micelles loaded with 6-coumarin as a fluorescence probe were stable to investigate their blood circulation and biodistribution. The in vitro phagocytosis results using murine peritoneal macrophages showed 10-fold reduction compared with a reference micelle. The in vivo blood circulation half-life of the polymeric micelles following intravenous administration in New Zealand Rabbits was increased from 0.55 h to 90.5h. More interestingly, tissue distribution results showed that the concentration of the micelles in the kidney is 4-fold higher than that in the liver and other organs 48 h after administration. The results of this work show great promise for designing more effective stealth drug carriers that can minimize reticuloendothelial system clearance and circulate for long time to reach target by using simple cell membrane mimetic random copolymer micelles.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, PR China; School of Environmental and Chemical Engineering, Xi'an Polytechnic University, Xi'an 710048, Shaanxi, PR China
| | - Yu-Dong Chai
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, PR China
| | - Jing Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, PR China
| | - Peng-Fei Huang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, PR China
| | - Kenichi Nakashima
- Department of Chemistry, Faculty of Science and Engineering, Saga University, 1 Honjo-machi, Saga 840-8502, Japan.
| | - Yong-Kuan Gong
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, Shaanxi, PR China.
| |
Collapse
|
100
|
Harisa GI, Badran MM, AlQahtani SA, Alanazi FK, Attia SM. Pravastatin chitosan nanogels-loaded erythrocytes as a new delivery strategy for targeting liver cancer. Saudi Pharm J 2015; 24:74-81. [PMID: 26903771 PMCID: PMC4720020 DOI: 10.1016/j.jsps.2015.03.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/16/2015] [Indexed: 01/07/2023] Open
Abstract
Chitosan nanogels (CNG) are developed as one of the most promising carriers for cancer targeting. However, these carriers are rapidly eliminated from circulation by reticuloendothelial system (RES), which limits their application. Therefore, erythrocytes (ER) loaded CNG as multifunctional carrier may overcome the massive elimination of nanocarriers by RES. In this study, erythrocytes loaded pravastatin-chitosan nanogels (PR-CNG-ER) were utilized as a novel drug carrier to target liver cancer. Thus, PR-CNG formula was developed in nanosize, with good entrapment efficiency, drug loading and sustained release over 48 h. Then, PR-CNG loaded into ER were prepared by hypotonic preswelling technique. The resulting PR-CNG-ER showed 36.85% of entrapment efficiency, 66.82% of cell recovery and release consistent to that of hemoglobin over 48 h. Moreover, PR-CNG-ER exhibited negative zeta potential, increasing of hemolysis percent, marked phosphatidylserine exposure and stomatocytes shape compared to control unloaded erythrocytes. PR-CNG-ER reduced cells viability of HepG2 cells line by 28% compared to unloaded erythrocytes (UER). These results concluded that PR-CNG-ER are promising drug carriers to target liver cancer.
Collapse
Affiliation(s)
- Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Biochemistry, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohamed M Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Pharmaceutics, College of Pharmacy, Al-Azhar University Cairo, Egypt
| | - Saeed A AlQahtani
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|