101
|
Hernández-Camarero P, Amezcua-Hernández V, Jiménez G, García MA, Marchal JA, Perán M. Clinical failure of nanoparticles in cancer: mimicking nature's solutions. Nanomedicine (Lond) 2020; 15:2311-2324. [PMID: 32969312 DOI: 10.2217/nnm-2020-0234] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanotechnology has become a promising approach in the treatment of cancer. However, most intravenously injected nanoparticles (NPs) do not effectively reach the tumor mass due to the biological barriers in the body. In an attempt to unify clinical criteria and basic research, we have collected the latest studies and described novel alternatives such as the use of NPs covered with cell membranes to increase NP delivery efficiency. Furthermore, we focus on the prospect of using the cell's natural messengers, exosomes, as vehicles to transport anti-cancer agents and we discuss the technical complications involved. Finally, we propose novel approaches to produce engineered exosomes which may overcome such technical limitations in order to achieve a proper anti-cancer nanotherapy.
Collapse
Affiliation(s)
- Pablo Hernández-Camarero
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas SN, Jaén E-23071, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain
| | - Víctor Amezcua-Hernández
- Hospital Universitario Virgen de las Nieves, Department Medical Oncology, Av. de las Fuerzas Armadas, 2, E-18014 Granada, Spain
| | - Gema Jiménez
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain.,Instituto de Investigación Biosanitaria IBS.GRANADA, Granada, E-18071, Spain
| | - María A García
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain.,Instituto de Investigación Biosanitaria IBS.GRANADA, Granada, E-18071, Spain.,Department of Biochemistry & Molecular Biology & Immunology, University of Granada, Granada, Spain
| | - Juan A Marchal
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain.,Instituto de Investigación Biosanitaria IBS.GRANADA, Granada, E-18071, Spain.,Department of Human Anatomy & Embryology, Faculty of Medicine, University of Granada, Avda. de la Investigación 11, Granada E-18016, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas SN, Jaén E-23071, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain
| |
Collapse
|
102
|
Chen Z, Xu L, Gao X, Wang C, Li R, Xu J, Zhang M, Panichayupakaranant P, Chen H. A multifunctional CeO 2@SiO 2-PEG nanoparticle carrier for delivery of food derived proanthocyanidin and curcumin as effective antioxidant, neuroprotective and anticancer agent. Food Res Int 2020; 137:109674. [PMID: 33233251 DOI: 10.1016/j.foodres.2020.109674] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/04/2020] [Accepted: 09/06/2020] [Indexed: 02/05/2023]
Abstract
The nanoparticle systems could effectively overcome the drug delivery challenges of food bioactive compounds. In this study, a novel and effective multifunctional PEG modified CeO2@SiO2 nanoparticle (CSP-NPs) system was successfully fabricated. Food derived proanthocyanidin (PAC) and curcumin (Cur) were loaded onto CSP-NPs and formed as PAC-NPs and Cur-NPs. Fourier transform Infrared spectra, X-ray diffraction, scanning electron microscopy, transmission electron microscopy, and dynamic light scattering were used to characterize the prepared NPs. CSP-NPs, PAC-NPs and Cur-NPs displayed spherical shape with about 35-45 nm size. The bioactivity analysis revealed that CSP-NPs system could effectively deliver PAC and Cur to exhibit strong antioxidant activity, potent neuroprotective effect against Aβ1-42-mediated toxicity in PC-12 cells (recovered cell viability from 57.5% to 81.0% at the dose of 25 μg/mL) and effective antiproliferative effects on HepG2 and Hela cells. Besides, all prepared nanoparticles (0-100 µg/ml) used in this study showed no significant toxicity on cell models of antioxidative and neuroprotective activities, excepting for cancer cells, suggesting that these nanoparticles had the potential of being utilized in drug delivery. Therefore, CSP-NPs might be a promising delivery system for hydrophilic molecule proanthocyanidin and hydrophobic molecule curcumin against the oxidative damage, neurodegenerative diseases and cancer, which could facilitate the application of food derived nutrients in functional foods industry.
Collapse
Affiliation(s)
- Zhongqin Chen
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Leilei Xu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Xudong Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Chunli Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Ruilin Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Jun Xu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Min Zhang
- Tianjin Agricultural University, Tianjin 300384, PR China; State Key Laboratory of Nutrition and Safety, Tianjin University of Science & Technology, Tianjin 300457, PR China
| | - Pharkphoom Panichayupakaranant
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Haixia Chen
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| |
Collapse
|
103
|
Yuan J, Yin WY, Wang Y, Chen J, Zhang ZM, Tang YX, Pei SY, Tan LX, Hu XW, Fan XG, Li N. Cargo-laden erythrocyte ghosts target liver mediated by macrophages. Transfus Apher Sci 2020; 60:102930. [PMID: 32933846 DOI: 10.1016/j.transci.2020.102930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/12/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
Liver-targeted cargo delivery possesses great potential for the treatment of liver disease. It is urgent to find an efficient and biocompatible liver targeted delivery system. This study focused on the liver targeting properties of erythrocyte ghosts and its possible mechanism. Herein, we optimized conditions to fabricate human and mouse erythrocyte ghosts with sufficient room capable of incorporating various model substances. Erythrocyte ghosts are biocompatible cargo carriers because it is derived from autologous red blood cells (RBCs), and the cell size, zeta potential, and biconcave-disk shape of the ghosts were consistent with those of RBCs. An in vivo imaging system and positron emission tomography/computed tomography imaging showed that the ghosts were captured mainly in the liver by intravenous injection of fluorescence or 18F-fluorodeoxyglucose (FDG)-labelled ghosts into mice. In contrast, the main concentration of naked octreotide was trapped in the lungs while naked 18F-FDG was trapped in the heart. However, the concentration of cargo-loaded ghosts decreased significantly in the liver in macrophage-depleted mice. Accordingly, in vitro experiments showed that higher phosphatidylserine exposure was observed in the ghosts (38.9 %) compared to normal erythrocytes (0.69 %), and the phagocytic activity of the macrophage RAW 264.7. on the ghosts was significantly higher than that of normal erythrocytes (p < 0.001). Together they indicate that erythrocyte ghosts show liver targeting properties, and possibly owing to macrophage phagocytosis. This promising and effective therapeutic delivery system may provide therapeutic benefits for liver disease.
Collapse
Affiliation(s)
- Jiao Yuan
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Yu Yin
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Laboratory of Ethnopharmacology Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Chen
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Min Zhang
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Xiang Tang
- Department of PET Centre, Xiangya Hospital, Central South University, Changsha, China
| | - Si-Ya Pei
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Lin-Xia Tan
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Xing-Wang Hu
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Gong Fan
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
104
|
Liu J, Wu Y, Fu C, Li B, Li L, Zhang R, Xu T, Xu ZP. Charge Reversion Simultaneously Enhances Tumor Accumulation and Cell Uptake of Layered Double Hydroxide Nanohybrids for Effective Imaging and Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002115. [PMID: 32608187 DOI: 10.1002/smll.202002115] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/18/2020] [Indexed: 05/19/2023]
Abstract
Nanotheranostics have been actively sought in precision nanomedicine in recent years. However, insufficient tumor accumulation and limited cell uptake often impede the nanotheranostic efficacy. Herein, pH-sensitive charge-reversible polymer-coated layered double hydroxide (LDH) nanohybrids are devised to possess long circulation in blood but reserve surface charges in the weakly acidic tumor tissue to re-expose therapeutic LDH nanoparticles for enhanced tumor accumulation and cell uptake. In vitro experimental data demonstrate that charge-reversible nanohybrids mitigate the cell uptake in physiological conditions (pH 7.4), but remarkably facilitate internalization by tumor cells after charge reversion in the weakly acidic environment (pH 6.8). More significantly, about 6.0% of injected charge-reversible nanohybrids accumulate in the tumor tissue at 24 h post injection, far higher than the average accumulation (0.7%) reported elsewhere for nanoparticles. This high tumor accumulation clearly shows the tumor tissues in T1 -weighted magnetic resonance imaging. As a consequence, >95% inhibition of tumor growth in the B16F0-bearing mouse model is achieved via only one treatment combining RNAi and photothermal therapy under very mild irradiation (808 nm laser, 0.3 W cm-2 for 180 s). The current research thus demonstrates a new strategy to functionalize nanoparticles and simultaneously enhance their tumor accumulation and cell internalization for effective cancer theranostics.
Collapse
Affiliation(s)
- Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Yilun Wu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Changkui Fu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Bei Li
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Tiefeng Xu
- The First Affiliated Hospital of Hainan Medical University, Cancer Institute of Hainan Medical University, Haikou, Hainan, 570102, China
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD, 4072, Australia
| |
Collapse
|
105
|
Synthesis of Poly(Dimethylmalic Acid) Homo- and Copolymers to Produce Biodegradable Nanoparticles for Drug Delivery: Cell Uptake and Biocompatibility Evaluation in Human Heparg Hepatoma Cells. Polymers (Basel) 2020; 12:polym12081705. [PMID: 32751402 PMCID: PMC7464256 DOI: 10.3390/polym12081705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Hydrophobic and amphiphilic derivatives of the biocompatible and biodegradable poly(dimethylmalic acid) (PdiMeMLA), varying by the nature of the lateral chains and the length of each block, respectively, have been synthesized by anionic ring-opening polymerization (aROP) of the corresponding monomers using an initiator/base system, which allowed for very good control over the (co)polymers' characteristics (molar masses, dispersity, nature of end-chains). Hydrophobic and core-shell nanoparticles (NPs) were then prepared by nanoprecipitation of hydrophobic homopolymers and amphiphilic block copolymers, respectively. Negatively charged NPs, showing hydrodynamic diameters (Dh) between 50 and 130 nm and narrow size distributions (0.08 < PDI < 0.22) depending on the (co)polymers nature, were obtained and characterized by dynamic light scattering (DLS), zetametry, and transmission electron microscopy (TEM). Finally, the cytotoxicity and cellular uptake of the obtained NPs were evaluated in vitro using the hepatoma HepaRG cell line. Our results showed that both cytotoxicity and cellular uptake were influenced by the nature of the (co)polymer constituting the NPs.
Collapse
|
106
|
Kumar S, Sharma B. Leveraging Electrostatic Interactions for Drug Delivery to the Joint. Bioelectricity 2020; 2:82-100. [PMID: 32856016 DOI: 10.1089/bioe.2020.0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Arthritis is a debilitating joint disease with a high economic burden and prevalence. There are many challenges delivering therapeutics to the joint, including low bioavailability when administered systemically and low joint retention after intra-articular injection. Therefore, drug delivery systems such as nanoparticles, liposomes, dendrimers, and carrier proteins have been utilized to overcome some of these limitations. To enhance joint tissue localization and retention, there are opportunities to leverage electrostatic interactions between drug carriers and various tissues and cells. These opportunities, as they pertain to specific joint tissues, are explored in this review. Further, the impact that electrostatic interactions has on various drug delivery parameters, such as the formation of a protein corona, the uptake and cytotoxicity, and the biodistribution of the drug delivery systems, is discussed. Lastly, this review summarizes key findings from studies that have investigated the use of electrostatic interactions to increase targeting of specific joint tissues and limitations in preclinical investigations are identified. As more novel targets are discovered in treating arthritis, there will be a continued need to localize therapeutics to specific tissues for greater therapeutic outcomes and hence attention must be paid in designing the drug delivery systems.
Collapse
Affiliation(s)
- Shreedevi Kumar
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
107
|
Koo DJ, Choi J, Ahn M, Ahn BH, Min DH, Kim SY. Large-Scale 3D Optical Mapping and Quantitative Analysis of Nanoparticle Distribution in Tumor Vascular Microenvironment. Bioconjug Chem 2020; 31:1784-1794. [DOI: 10.1021/acs.bioconjchem.0c00263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Dong-Jun Koo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, South Korea
- Program in Neuroscience, Seoul National University, Seoul 08826, South Korea
| | - Jinahn Choi
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Minchul Ahn
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 08826, South Korea
| | - Benjamin H. Ahn
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, South Korea
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 08826, South Korea
| | - Sung-Yon Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, South Korea
- Program in Neuroscience, Seoul National University, Seoul 08826, South Korea
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
108
|
Pircalabioru GG, Chifiriuc MC. Nanoparticulate drug-delivery systems for fighting microbial biofilms: from bench to bedside. Future Microbiol 2020; 15:679-698. [PMID: 32495694 DOI: 10.2217/fmb-2019-0251] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Biofilms are highly tolerant to antimicrobial agents and adverse environmental conditions being important reservoirs for chronic and hard-to-treat infections. Nanomaterials exhibit microbiostatic/microbicidal/antipathogenic properties and can be also used for the delivery of antibiofilm agents. However, few of the many promising leads offered by nanotechnology reach clinical studies and eventually, become available to clinicians. The aim of this paper was to review the progress and challenges in the development of nanotechnology-based antibiofilm drug-delivery systems. The main identified challenges are: most papers report only in vitro studies of the activity of different nanoformulations; lack of standardization in the methodological approaches; insufficient collaboration between material science specialists and clinicians; paucity of in vivo studies to test efficiency and safety.
Collapse
Affiliation(s)
- Gratiela G Pircalabioru
- University of Bucharest, Faculty of Biology, Research Institute of The University of Bucharest (ICUB), Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- University of Bucharest, Faculty of Biology, Research Institute of The University of Bucharest (ICUB), Bucharest, Romania
| |
Collapse
|
109
|
Zahin N, Anwar R, Tewari D, Kabir MT, Sajid A, Mathew B, Uddin MS, Aleya L, Abdel-Daim MM. Nanoparticles and its biomedical applications in health and diseases: special focus on drug delivery. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:19151-19168. [PMID: 31079299 DOI: 10.1007/s11356-019-05211-0] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/16/2019] [Indexed: 05/21/2023]
Abstract
Nanotechnology is an emerging technology that deals with nanosized particles possessing crucial research roles and application. Disciplines like chemistry, biology, physics, engineering, materials science, and health sciences provide an accumulated knowledge of nanotechnology. Nonetheless, it has vast submissions precisely in biology, electronics, and medicine. Aimed at drug delivery system, nanoparticles are based on the mechanism of entrapment of the drugs or biomolecules into the interior structure of the particles; another mechanism could be that the drugs or the biomolecules can be absorbed onto the exterior surfaces of the particles. Currently, nanoparticles (NPs) are used in the delivery of drugs, proteins, genes, vaccines, polypeptides, nucleic acids, etc. In recent years, various applications of the drug delivery system via NPs have encountered an enormous position sector like pharmaceutical, medical, biological, and others. Considering the impact of NPs in drug delivery systems, this review focuses on the detailed profile of NPs, its impact on biology and medicine, and their commercialization prospects.
Collapse
Affiliation(s)
- Nuzhat Zahin
- Department of Pharmacy, BRAC University, Dhaka, Bangladesh
| | | | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | | | - Amin Sajid
- Department of Pharmacy, BRAC University, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | | |
Collapse
|
110
|
Gisbert-Garzarán M, Vallet-Regí M. Influence of the Surface Functionalization on the Fate and Performance of Mesoporous Silica Nanoparticles. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E916. [PMID: 32397449 PMCID: PMC7279540 DOI: 10.3390/nano10050916] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Mesoporous silica nanoparticles have been broadly applied as drug delivery systems owing to their exquisite features, such as excellent textural properties or biocompatibility. However, there are various biological barriers that prevent their proper translation into the clinic, including: (1) lack of selectivity toward tumor tissues, (2) lack of selectivity for tumoral cells and (3) endosomal sequestration of the particles upon internalization. In addition, their open porous structure may lead to premature drug release, consequently affecting healthy tissues and decreasing the efficacy of the treatment. First, this review will provide a comprehensive and systematic overview of the different approximations that have been implemented into mesoporous silica nanoparticles to overcome each of such biological barriers. Afterward, the potential premature and non-specific drug release from these mesoporous nanocarriers will be addressed by introducing the concept of stimuli-responsive gatekeepers, which endow the particles with on-demand and localized drug delivery.
Collapse
Affiliation(s)
- Miguel Gisbert-Garzarán
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
111
|
Cheng YH, He C, Riviere JE, Monteiro-Riviere NA, Lin Z. Meta-Analysis of Nanoparticle Delivery to Tumors Using a Physiologically Based Pharmacokinetic Modeling and Simulation Approach. ACS NANO 2020; 14:3075-3095. [PMID: 32078303 PMCID: PMC7098057 DOI: 10.1021/acsnano.9b08142] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/20/2020] [Indexed: 05/18/2023]
Abstract
Numerous studies have engineered nanoparticles with different physicochemical properties to enhance the delivery efficiency to solid tumors, yet the mean and median delivery efficiencies are only 1.48% and 0.70% of the injected dose (%ID), respectively, according to a study using a nonphysiologically based modeling approach based on published data from 2005 to 2015. In this study, we used physiologically based pharmacokinetic (PBPK) models to analyze 376 data sets covering a wide range of nanomedicines published from 2005 to 2018 and found mean and median delivery efficiencies at the last sampling time point of 2.23% and 0.76%ID, respectively. Also, the mean and median delivery efficiencies were 2.24% and 0.76%ID at 24 h and were decreased to 1.23% and 0.35%ID at 168 h, respectively, after intravenous administration. While these delivery efficiencies appear to be higher than previous findings, they are still quite low and represent a critical barrier in the clinical translation of nanomedicines. We explored the potential causes of this poor delivery efficiency using the more mechanistic PBPK perspective applied to a subset of gold nanoparticles and found that low delivery efficiency was associated with low distribution and permeability coefficients at the tumor site (P < 0.01). We also demonstrate how PBPK modeling and simulation can be used as an effective tool to investigate tumor delivery efficiency of nanomedicines.
Collapse
Affiliation(s)
- Yi-Hsien Cheng
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Chunla He
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Jim E. Riviere
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- 1Data
Consortium, Kansas State University, Manhattan, Kansas 66506, United States
| | - Nancy A. Monteiro-Riviere
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zhoumeng Lin
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
112
|
Kapadia CH, Ioele SA, Day ES. Layer-by-layer assembled PLGA nanoparticles carrying miR-34a cargo inhibit the proliferation and cell cycle progression of triple-negative breast cancer cells. J Biomed Mater Res A 2020; 108:601-613. [PMID: 31742868 PMCID: PMC7103458 DOI: 10.1002/jbm.a.36840] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/06/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for 15-25% of diagnosed breast cancers, and its lack of a clinically defined therapeutic target has caused patients to suffer from earlier relapse and higher mortality rates than patients with other breast cancer subtypes. MicroRNAs (miRNAs) are small non-coding RNAs that regulate the expression of multiple genes through RNA interference to maintain normal tissue function. The tumor suppressor miR-34a is downregulated in TNBC, and its loss-of-expression correlates with worse disease outcomes. Therefore, delivering miR-34a mimics into TNBC cells is a promising strategy to combat disease progression. To achieve this goal, we synthesized layer-by-layer assembled nanoparticles (LbL NPs) comprised of spherical poly(lactic-co-glycolic acid) cores surrounded by alternating layers of poly-L-lysine (PLL) and miR-34a. TNBC cells internalized these LbL NPs to a greater extent than polyplexes comprised of PLL and miRNA, and confocal microscopy showed that LbL NPs delivered a substantial fraction of miR-34a cargo into the cytosol. This yielded robust suppression of the miR-34a target genes CCND-1, Notch-1, Bcl-2, Survivin, and MDR-1, which reduced TNBC cell proliferation and induced cell cycle arrest. These data validate that miR-34a delivery can impair TNBC cell function and support continued investigation of this platform for treatment of TNBC.
Collapse
Affiliation(s)
- Chintan H. Kapadia
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - Stephen A. Ioele
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - Emily S. Day
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware
- Helen F. Graham Cancer Center and Research Institute, Newark, Delaware
| |
Collapse
|
113
|
Hu J, Gao G, He M, Yin Q, Gao X, Xu H, Sun T. Optimal route of gold nanoclusters administration in mice targeting Parkinson’s disease. Nanomedicine (Lond) 2020; 15:563-580. [DOI: 10.2217/nnm-2019-0268] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: To explore the optimal route of gold nanoclusters (AuNCs) administration in mice targeting Parkinson’s disease. Materials & methods: Assessing the pharmacokinetic and bioavailability of AuNCs in mice administrated following intravenous, intraperitoneal, gavage and intranasal injection. Investigating the biodistribution of AuNCs in mice by atomic absorption spectrometry and transmission electron microscope. Toxicity assessments of AuNCs were carried out both in cells and in mice. Results: Administration of AuNCs via intraperitoneal injection showed the greatest bioavailability and the longest residence in brain. AuNCs could penetrate blood–brain barrier and be excreted mainly through kidney. No obvious toxicity of AuNCs found in cells and in mice. Conclusion: The optimal route of AuNCs administration in mice targeting Parkinson’s disease is intraperitoneal administration.
Collapse
Affiliation(s)
- Jinqi Hu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis & Processing, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Meng He
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Qiang Yin
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Xiaobing Gao
- General Hospital of Central Theater Command, No. 627 Wuluo Road, Wuhan, 430070, PR China
| | - Haixing Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
- State Key Laboratory of Advanced Technology for Materials Synthesis & Processing, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan, 430070, PR China
| |
Collapse
|
114
|
Liu TI, Lu TY, Chang SH, Shen MY, Chiu HC. Dual stimuli-guided lipid-based delivery system of cancer combination therapy. J Control Release 2020; 318:16-24. [DOI: 10.1016/j.jconrel.2019.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/25/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022]
|
115
|
Tang J, Zhang R, Guo M, Zhou H, Zhao Y, Liu Y, Wu Y, Chen C. Gd-metallofullerenol drug delivery system mediated macrophage polarization enhances the efficiency of chemotherapy. J Control Release 2020; 320:293-303. [PMID: 32004584 DOI: 10.1016/j.jconrel.2020.01.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/13/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Treatment of solid tumors by chemotherapy is usually failed in clinical because of its low effectiveness and side effects. Stimulation of immune system in vivo to fight cancer has been proved to be a pleasant complementary to systemic chemotherapy. Herein, we have developed a combination cancer therapy strategy by using polymer nanoparticles to deliver Gd-metallofullerenol and doxorubicin simultaneously. The Gd-metallofullerenol provoked the Th1 immune response by regulating the M1 macrophage polarization and the doxorubicin realized direct tumor cells killing by its cytotoxic effect. Also, the Gd-metallofullerenol as part of component in delivery system enhances the encapsulation efficiency of doxorubicin in polymer cargo for potential passive tumor target. The biocompatible and reliable method by combining nanoparticle-induced immune modulation and chemotherapy triggers systemic antitumor immune responses for the synergistic inhibition of tumor growth in vivo. The integration of Gd-metallofullerenol and doxorubicin with potentially complementary functions in one nanoplatform may provide new opportunities to improve cancer treatments.
Collapse
Affiliation(s)
- Jinglong Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; School of Public Health, Qingdao University, Qingdao 226021, China
| | - Ruirui Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Beijing Key Laboratory of Ionic Liquids Clean Process, Key Laboratory of Green Process and Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
116
|
Gisbert-Garzarán M, Manzano M, Vallet-Regí M. Mesoporous Silica Nanoparticles for the Treatment of Complex Bone Diseases: Bone Cancer, Bone Infection and Osteoporosis. Pharmaceutics 2020; 12:E83. [PMID: 31968690 PMCID: PMC7022913 DOI: 10.3390/pharmaceutics12010083] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/13/2020] [Accepted: 01/19/2020] [Indexed: 12/13/2022] Open
Abstract
Bone diseases, such as bone cancer, bone infection and osteoporosis, constitute a major issue for modern societies as a consequence of their progressive ageing. Even though these pathologies can be currently treated in the clinic, some of those treatments present drawbacks that may lead to severe complications. For instance, chemotherapy lacks great tumor tissue selectivity, affecting healthy and diseased tissues. In addition, the inappropriate use of antimicrobials is leading to the appearance of drug-resistant bacteria and persistent biofilms, rendering current antibiotics useless. Furthermore, current antiosteoporotic treatments present many side effects as a consequence of their poor bioavailability and the need to use higher doses. In view of the existing evidence, the encapsulation and selective delivery to the diseased tissues of the different therapeutic compounds seem highly convenient. In this sense, silica-based mesoporous nanoparticles offer great loading capacity within their pores, the possibility of modifying the surface to target the particles to the malignant areas and great biocompatibility. This manuscript is intended to be a comprehensive review of the available literature on complex bone diseases treated with silica-based mesoporous nanoparticles-the further development of which and eventual translation into the clinic could bring significant benefits for our future society.
Collapse
Affiliation(s)
- Miguel Gisbert-Garzarán
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Miguel Manzano
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
117
|
Mursaleen L, Somavarapu S, Zariwala MG. Deferoxamine and Curcumin Loaded Nanocarriers Protect Against Rotenone-Induced Neurotoxicity. JOURNAL OF PARKINSONS DISEASE 2020; 10:99-111. [DOI: 10.3233/jpd-191754] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Leah Mursaleen
- School of Life Sciences, University of Westminster, London, UK
- Department of Pharmaceutics, UCL School of Pharmacy, London, UK
- The Cure Parkinson’s Trust, London, UK
| | | | | |
Collapse
|
118
|
Alfranca G, Beola L, Liu Y, Gutiérrez L, Zhang A, Artiga A, Cui D, de la Fuente JM. In vivo comparison of the biodistribution and long-term fate of colloids – gold nanoprisms and nanorods – with minimum surface modification. Nanomedicine (Lond) 2019; 14:3035-3055. [DOI: 10.2217/nnm-2019-0253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To study the difference in biodistribution of gold nanoprisms (NPr) and nanorods (NR), PEGylated to ensure colloidal stability. Materials & methods: Surface changes were studied for nanoparticles in different media, while the biodistribution was quantified and imaged in vivo. Results: Upon interaction with the mouse serum, NR showed more abrupt changes in surface properties than NPr. In the in vivo tests, while NPr accumulated similarly in the spleen and liver, NR showed much higher gold presence in the spleen than in liver; together with some accumulation in kidneys, which was nonexistent in NPr. NPr were cleared from the tissues 2 months after administration, while NR were more persistent. Conclusion: The results suggest that the differential biodistribution is caused by size-/shape-dependent interactions with the serum.
Collapse
Affiliation(s)
- Gabriel Alfranca
- Department of Instrument Science & Engineering, School of Electronic Information & Electrical Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis & Treatment Instrument, Institute of Nano Biomedicine & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC/Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Lilianne Beola
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC/Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Yanlei Liu
- Department of Instrument Science & Engineering, School of Electronic Information & Electrical Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis & Treatment Instrument, Institute of Nano Biomedicine & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
| | - Lucía Gutiérrez
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC/Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 50018 Madrid, Spain
- Department of Analytical Chemistry, Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Amin Zhang
- Department of Instrument Science & Engineering, School of Electronic Information & Electrical Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis & Treatment Instrument, Institute of Nano Biomedicine & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
| | - Alvaro Artiga
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC/Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 50018 Madrid, Spain
| | - Daxiang Cui
- Department of Instrument Science & Engineering, School of Electronic Information & Electrical Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis & Treatment Instrument, Institute of Nano Biomedicine & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
| | - Jesús M de la Fuente
- Department of Instrument Science & Engineering, School of Electronic Information & Electrical Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis & Treatment Instrument, Institute of Nano Biomedicine & Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd, Shanghai 200240, China
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC/Universidad de Zaragoza, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 50018 Madrid, Spain
| |
Collapse
|
119
|
Shreffler JW, Pullan JE, Dailey KM, Mallik S, Brooks AE. Overcoming Hurdles in Nanoparticle Clinical Translation: The Influence of Experimental Design and Surface Modification. Int J Mol Sci 2019; 20:E6056. [PMID: 31801303 PMCID: PMC6928924 DOI: 10.3390/ijms20236056] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/23/2019] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles are becoming an increasingly popular tool for biomedical imaging and drug delivery. While the prevalence of nanoparticle drug-delivery systems reported in the literature increases yearly, relatively little translation from the bench to the bedside has occurred. It is crucial for the scientific community to recognize this shortcoming and re-evaluate standard practices in the field, to increase clinical translatability. Currently, nanoparticle drug-delivery systems are designed to increase circulation, target disease states, enhance retention in diseased tissues, and provide targeted payload release. To manage these demands, the surface of the particle is often modified with a variety of chemical and biological moieties, including PEG, tumor targeting peptides, and environmentally responsive linkers. Regardless of the surface modifications, the nano-bio interface, which is mediated by opsonization and the protein corona, often remains problematic. While fabrication and assessment techniques for nanoparticles have seen continued advances, a thorough evaluation of the particle's interaction with the immune system has lagged behind, seemingly taking a backseat to particle characterization. This review explores current limitations in the evaluation of surface-modified nanoparticle biocompatibility and in vivo model selection, suggesting a promising standardized pathway to clinical translation.
Collapse
Affiliation(s)
| | | | | | | | - Amanda E. Brooks
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (J.W.S.); (J.E.P.); (K.M.D.); (S.M.)
| |
Collapse
|
120
|
Lee SWL, Paoletti C, Campisi M, Osaki T, Adriani G, Kamm RD, Mattu C, Chiono V. MicroRNA delivery through nanoparticles. J Control Release 2019; 313:80-95. [PMID: 31622695 PMCID: PMC6900258 DOI: 10.1016/j.jconrel.2019.10.007] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are attracting a growing interest in the scientific community due to their central role in the etiology of major diseases. On the other hand, nanoparticle carriers offer unprecedented opportunities for cell specific controlled delivery of miRNAs for therapeutic purposes. This review critically discusses the use of nanoparticles for the delivery of miRNA-based therapeutics in the treatment of cancer and neurodegenerative disorders and for tissue regeneration. A fresh perspective is presented on the design and characterization of nanocarriers to accelerate translation from basic research to clinical application of miRNA-nanoparticles. Main challenges in the engineering of miRNA-loaded nanoparticles are discussed, and key application examples are highlighted to underline their therapeutic potential for effective and personalized medicine.
Collapse
Affiliation(s)
- Sharon Wei Ling Lee
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy; Singapore-MIT Alliance for Research & Technology (SMART), BioSystems and Micromechanics (BioSyM), Singapore, Singapore(3); Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore(3); Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore, Singapore(3)
| | - Camilla Paoletti
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy
| | - Marco Campisi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy
| | - Tatsuya Osaki
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, Room NE47-321, Cambridge, MA, 02139, USA; Institute of Industrial Science, The University of Tokyo, Meguro-ku, Tokyo 153-8505, Japan(3)
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore, Singapore(3); Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Roger D Kamm
- Singapore-MIT Alliance for Research & Technology (SMART), BioSystems and Micromechanics (BioSyM), Singapore, Singapore(3); Department of Mechanical Engineering, Massachusetts Institute of Technology, 500 Technology Square, Room NE47-321, Cambridge, MA, 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 500 Technology Square, Room NE47-321, Cambridge, MA, 02139, USA
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy.
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
121
|
Chin DD, Wang J, Mel de Fontenay M, Plotkin A, Magee GA, Chung EJ. Hydroxyapatite-binding micelles for the detection of vascular calcification in atherosclerosis. J Mater Chem B 2019; 7:6449-6457. [PMID: 31553027 PMCID: PMC6812598 DOI: 10.1039/c9tb01918a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a chronic disease characterized by the formation of calcified, arterial plaques. Microcalcifications (5 μm to 100 μm), mainly composed of hydroxyapatite (HA, Ca5(PO4)3(OH)), develop in the fibrous caps of atherosclerotic plaques and can trigger plaque rupture due to the loss of compliance and elasticity. Ultimately, plaque rupture can cause arterial occlusion and embolization and result in ischemic events such as strokes and myocardial infarctions. Unfortunately, current imaging technologies used to detect calcifications are limited by low signal-to-noise ratio or use invasive procedures that pose risk of arterial dissection. To mitigate these drawbacks, in our study, we developed a novel, fluorescently-labeled peptide amphiphile micelle (PAM) that uses a 12 amino acid HA-binding peptide (HABP) [SVSVGMKPSPRP] to target and detect atherosclerotic calcification (HA PAM). Our results show HA PAMs can successfully target HA microcrystals with a strong binding affinity (KD = 6.26 ± 1.2 μM) in vitro. In addition, HA PAMs detected HA mineralization (HA PAM vs. non-targeting micelle, p≤ 0.001; HA PAM vs. scrambled HABP PAM, p≤ 0.01) formed by calcifying mouse aortic vascular smooth muscle cells (MOVAS). Moreover, HA PAMs successfully detected calcifications in atherosclerotic mouse models as well as in patient-derived arteries. Our studies show that HA PAMs show promise as calcium-targeting nanoparticles for the detection of calcifications in atherosclerosis.
Collapse
Affiliation(s)
- Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Margot Mel de Fontenay
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Anastasia Plotkin
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gregory A Magee
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA. and Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA and Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
122
|
Mosaiab T, Farr DC, Kiefel MJ, Houston TA. Carbohydrate-based nanocarriers and their application to target macrophages and deliver antimicrobial agents. Adv Drug Deliv Rev 2019; 151-152:94-129. [PMID: 31513827 DOI: 10.1016/j.addr.2019.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
Many deadly infections are produced by microorganisms capable of sustained survival in macrophages. This reduces exposure to chemadrotherapy, prevents immune detection, and is akin to criminals hiding in police stations. Therefore, the use of glyco-nanoparticles (GNPs) as carriers of therapeutic agents is a burgeoning field. Such an approach can enhance the penetration of drugs into macrophages with specific carbohydrate targeting molecules on the nanocarrier to interact with macrophage lectins. Carbohydrates are natural biological molecules and the key constituents in a large variety of biological events such as cellular communication, infection, inflammation, enzyme trafficking, cellular migration, cancer metastasis and immune functions. The prominent characteristics of carbohydrates including biodegradability, biocompatibility, hydrophilicity and the highly specific interaction of targeting cell-surface receptors support their potential application to drug delivery systems (DDS). This review presents the 21st century development of carbohydrate-based nanocarriers for drug targeting of therapeutic agents for diseases localized in macrophages. The significance of natural carbohydrate-derived nanoparticles (GNPs) as anti-microbial drug carriers is highlighted in several areas of treatment including tuberculosis, salmonellosis, leishmaniasis, candidiasis, and HIV/AIDS.
Collapse
Affiliation(s)
- Tamim Mosaiab
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Dylan C Farr
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Milton J Kiefel
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia.
| | - Todd A Houston
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia.
| |
Collapse
|
123
|
Synergistic combination treatment to break cross talk between cancer cells and bone cells to inhibit progression of bone metastasis. Biomaterials 2019; 227:119558. [PMID: 31654872 DOI: 10.1016/j.biomaterials.2019.119558] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/05/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
Advanced-stage cancers often metastasize to bone, and is the major cause of cancer-related morbidity and mortality. Due to poor biodistribution of intravenously administered anticancer drugs within the bone, chemotherapy is not optimally effective in treating bone metastasis. Additionally, overexpression of receptor activator of nuclear factor κB ligand (RANKL) in the bone microenvironment drives the vicious, destructive cycle of progression of bone metastasis and bone resorption. We hypothesized that the combination treatment - with docetaxel (TXT), an anticancer drug encapsulated in sustained release biodegradable nanoparticles (TXT-NPs) that are designed to localize in bone marrow, and denosumab monoclonal antibody (DNmb), which binds to RANKL - could be more effective than either treatment alone. We tested our hypothesis in intraosseous prostate cancer (PC-3) cell-induced osteolytic mouse model of bone metastasis with treatments given intravenously. The results demonstrated better efficacy with TXT-NPs than with TXT-CrEL or saline control in inhibiting progression of metastasis and improving survival. TXT-NPs showed ~3-fold higher drug levels in metastasized bone tissue at 1 wk post-administration than TXT-CrEL, thus explaining their efficacy. However, the combination treatment (TXT-NPs + DNmb) given simultaneously was significantly more effective in inhibiting metastatic progression; it caused early tumor regression and improved survival, and caused no body weight loss or tumor relapse, even when the treatment was discontinued, whereas TXT-NPs or DNmb alone treatments showed tumor relapse after an initial regression. Micro-CT analysis of the bone from the combination treatment showed no bone loss and normal bone mineral content, bone density, and bone volume fraction, whereas TXT-NPs or DNmb alone treatments showed bone loss. Confirming the above results, histochemical analysis of the bone from the combination treatment demonstrated normal bone morphology, and osteoblast and osteoclast cell activities. In conclusion, TXT-NPs and DNmb in combination, because of their complementary roles in breaking the cross talk between cancer cells and bone cells, was significantly effective in treating bone metastasis.
Collapse
|
124
|
Feng S, Zhou J, Li Z, Appelman HD, Zhao L, Zhu J, Wang TD. Sorafenib encapsulated in nanocarrier functionalized with glypican-3 specific peptide for targeted therapy of hepatocellular carcinoma. Colloids Surf B Biointerfaces 2019; 184:110498. [PMID: 31536939 DOI: 10.1016/j.colsurfb.2019.110498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/23/2019] [Accepted: 09/08/2019] [Indexed: 12/18/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer in the world with increasing incidence. Chemotherapy is required for HCC patients after receiving surgical resection. Serious off-target induced side effects and systemic toxicity limit the clinical utility of drugs. Targeting therapeutic nanomedicine is an innovative strategy for enhancing drug delivery efficiency and reducing side effects. Here, we successfully formulated nanocarriers to encapsulate sorafenib, an FDA approved drug for treatment of HCC. Sorafenib is encapsulated with an entrapment efficiency >80% over 20 days. The effective aqueous solubility is improved over 1900-fold. The release ratio in vitro is characterized by a half-life of T1/2 = 22.7 h. The peak target-to-background ratio for nanocarrier uptake by tumor occurs at 24 h post-injection, and is significantly greater for the target peptide versus controls. Ex vivo biodistribution confirms the in vivo results. Tumor regression is significantly greater for the target peptide versus controls after 21 days of therapy. No acute toxicity is found by blood chemistry or necropsy. In summary, a peptide specific for GPC3 has been identified, and used to modify the surface of a nanocarrier that encapsulates sorafenib with high entrapment efficiency. Regression of HCC xenograft tumors showed promise for targeted drug delivery.
Collapse
Affiliation(s)
- Shuo Feng
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Juan Zhou
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Henry D Appelman
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China.
| | - Thomas D Wang
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
125
|
Bailly AL, Correard F, Popov A, Tselikov G, Chaspoul F, Appay R, Al-Kattan A, Kabashin AV, Braguer D, Esteve MA. In vivo evaluation of safety, biodistribution and pharmacokinetics of laser-synthesized gold nanoparticles. Sci Rep 2019; 9:12890. [PMID: 31501470 PMCID: PMC6734012 DOI: 10.1038/s41598-019-48748-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 08/08/2019] [Indexed: 12/19/2022] Open
Abstract
Capable of generating plasmonic and other effects, gold nanostructures can offer a variety of diagnostic and therapy functionalities for biomedical applications, but conventional chemically-synthesized Au nanomaterials cannot always match stringent requirements for toxicity levels and surface conditioning. Laser-synthesized Au nanoparticles (AuNP) present a viable alternative to chemical counterparts and can offer exceptional purity (no trace of contaminants) and unusual surface chemistry making possible direct conjugation with biocompatible polymers (dextran, polyethylene glycol). This work presents the first pharmacokinetics, biodistribution and safety study of laser-ablated dextran-coated AuNP (AuNPd) under intravenous administration in small animal model. Our data show that AuNPd are rapidly eliminated from the blood circulation and accumulated preferentially in liver and spleen, without inducing liver or kidney toxicity, as confirmed by the plasmatic ALAT and ASAT activities, and creatininemia values. Despite certain residual accumulation in tissues, we did not detect any sign of histological damage or inflammation in tissues, while IL-6 level confirmed the absence of any chronic inflammation. The safety of AuNPd was confirmed by healthy behavior of animals and the absence of acute and chronic toxicities in liver, spleen and kidneys. Our results demonstrate that laser-synthesized AuNP are safe for biological systems, which promises their successful biomedical applications.
Collapse
Affiliation(s)
- Anne-Laure Bailly
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Florian Correard
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
- APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Anton Popov
- Aix Marseille Univ, CNRS, LP3, Campus de Luminy, Case 917, 13288, Marseille, France
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), Bio-nanophotonics Lab., 115409, Moscow, Russia
| | - Gleb Tselikov
- Aix Marseille Univ, CNRS, LP3, Campus de Luminy, Case 917, 13288, Marseille, France
| | - Florence Chaspoul
- Aix Marseille Univ, Avignon Université, CNRS, IRD, IMBE, Marseille, France
| | - Romain Appay
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
- APHM, Hôpital de la Timone, Service d'Anatomie Pathologique et de Neuropathologie, Marseille, France
| | - Ahmed Al-Kattan
- Aix Marseille Univ, CNRS, LP3, Campus de Luminy, Case 917, 13288, Marseille, France
| | - Andrei V Kabashin
- Aix Marseille Univ, CNRS, LP3, Campus de Luminy, Case 917, 13288, Marseille, France.
- MEPhI, Institute of Engineering Physics for Biomedicine (PhysBio), Bio-nanophotonics Lab., 115409, Moscow, Russia.
| | - Diane Braguer
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
- APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Marie-Anne Esteve
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
- APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France.
| |
Collapse
|
126
|
Xu X, Ballauff M. Interaction of Lysozyme with a Dendritic Polyelectrolyte: Quantitative Analysis of the Free Energy of Binding and Comparison to Molecular Dynamics Simulations. J Phys Chem B 2019; 123:8222-8231. [DOI: 10.1021/acs.jpcb.9b07448] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Xiao Xu
- School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei, 210094 Nanjing, P. R. China
| | - Matthias Ballauff
- Soft Matter and Functional Materials, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, 14109 Berlin, Germany
- Institut für Physik, Humboldt-Universität zu Berlin, 12489 Berlin, Germany
| |
Collapse
|
127
|
Li X, Zhen M, Zhou C, Deng R, Yu T, Wu Y, Shu C, Wang C, Bai C. Gadofullerene Nanoparticles Reverse Dysfunctions of Pancreas and Improve Hepatic Insulin Resistance for Type 2 Diabetes Mellitus Treatment. ACS NANO 2019; 13:8597-8608. [PMID: 31314991 DOI: 10.1021/acsnano.9b02050] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Type 2 diabetes mellitus (T2DM) has been one of the most prevalent metabolic disorders. Nonetheless, the commonly used anti-T2DM drugs failed to substant to treat T2DM when anti-T2DM was withdrawn. Here we put forward a superior and sustainable anti-diabetic strategy using intraperitoneal administration of amino-acid-functionalized gadofullerene nanoparticles (GFNPs) in db/db diabetic mice. Highly accumulated in the pancreas and liver, GFNPs could prominently decrease hyperglycemia, along with permanently maintaining normal blood sugar levels in T2DM mice and even stopping administration. Importantly, GFNPs reversed the pancreas islets dysfunctions by reducing oxidative stress and inflammation responses and fundamentally normalized the insulin secretory function of the pancreas islets. Mechanistically, GFNPs improved hepatic insulin resistance by regulating glucose and lipid metabolism through the activation of IRS2/PI3K/AKT signal pathways, resulting in inhibiting gluconeogenesis and increasing glycogenesis in the liver. Additionally, GFNPs relieved hepatic steatosis in the liver, ultimately maintaining systemic glucose and lipid metabolic homeostasis without obvious toxicity. Together, GFNPs reverse the dysfunctions of the pancreas and improve hepatic insulin resistance, providing a promising approach for T2DM treatment.
Collapse
Affiliation(s)
- Xue Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Mingming Zhen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Chen Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Ruijun Deng
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Tong Yu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Yingjie Wu
- Institute of Genome Engineered Animal Models for Human Disease , Dalian Medical University , Dalian 116044 , China
| | - Chunying Shu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Chunli Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
128
|
Fathy MM, Fahmy HM, Balah AMM, Mohamed FF, Elshemey WM. Magnetic nanoparticles-loaded liposomes as a novel treatment agent for iron deficiency anemia: In vivo study. Life Sci 2019; 234:116787. [PMID: 31445028 DOI: 10.1016/j.lfs.2019.116787] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/17/2019] [Accepted: 08/20/2019] [Indexed: 12/19/2022]
Abstract
Iron deficiency anemia (IDA) is a major worldwide public health problem. This is due to its prevalence among infants, children, adolescents, pregnant and reproductive age women. Ferrous sulfate (FeSO4) is the first line therapy for iron IDA. Unfortunately, it is reported that FeSO4 suffers from low absorption rate in the body and itself exhibits severe side effects. Herein, iron oxide magnetic nanoparticles-loaded liposomes (LMNPs) are prepared, characterized and evaluated as a treatment regimen for IDA in Wistar rats (as an animal model). Iron oxide magnetic nanoparticles (MNPs) are prepared and loaded into liposomes using the thin film hydration method. The size of the prepared formulations is in the range 10-100 nm, thus it can avoid the reticular endothelial system (RES), and increased their blood circulation time. For in vivo assessment, thirty-five Wistar rats are divided into 5 groups (n = 7): negative control group, positive control group, and three groups treated with different iron formulations (FeSO4, MNPs and LMNPs). Anemia is induced in the anemic groups by the bleeding method and then treatment started with different iron compounds administrated orally for 13 days. Hematological parameters are followed up during the treatment period. Results indicate that, in the LMNPs group, the hematological parameters turn to normal values and the histopathological structures of the liver, spleen and kidney remain normal. This proves that liposome increases the bioavailability of MNPs. In conclusion, LMNPs demonstrate superiority as a therapeutic regimen for the treatment of IDA among the tested iron formulations.
Collapse
Affiliation(s)
- Mohamed M Fathy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
| | - Asmaa M M Balah
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Faten F Mohamed
- Pathology Department, Faculty of Veterinary Medicine, Giza 12211, Egypt
| | - Wael M Elshemey
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Department of Physics, Faculty of Science, Islamic University in Madinah, Saudi Arabia
| |
Collapse
|
129
|
Rezaei G, Daghighi SM, Raoufi M, Esfandyari-Manesh M, Rahimifard M, Mobarakeh VI, Kamalzare S, Ghahremani MH, Atyabi F, Abdollahi M, Rezaee F, Dinarvand R. Synthetic and biological identities of polymeric nanoparticles influencing the cellular delivery: An immunological link. J Colloid Interface Sci 2019; 556:476-491. [PMID: 31473538 DOI: 10.1016/j.jcis.2019.08.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/10/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022]
Abstract
Enhanced understanding of bio-nano interaction requires recognition of hidden factors such as protein corona, a layer of adsorbed protein around nano-systems. This study compares the biological identity and fingerprint profile of adsorbed proteins on PLGA-based nanoparticles through nano-liquid chromatography-tandem mass spectrometry. The total proteins identified in the corona of nanoparticles (NPs) with different in size, charge and compositions were classified based on molecular mass, isoelectric point and protein function. A higher abundance of complement proteins was observed in modified NPs with an increased size, while NPs with a positive surface charge exhibited the minimum adsorption for immunoglobulin proteins. A correlation of dysopsonin/opsonin ratio was found with cellular uptake of NPs exposed to two positive and negative Fc receptor cell lines. Although the higher abundance of dysopsonins such as apolipoproteins may cover the active sites of opsonins causing a lower uptake, the correlation of adsorbed dysopsonin/opsonin proteins on the NPs surface has an opposite trend with the intensity of cell uptake. Despite the reduced uptake of corona-coated NPs in comparison with pristine NPs, the dysopsonin/opsonin ratio controlled by the physicochemistry properties of NPs could potentially be used to tune up the cellular delivery of polymeric NPs.
Collapse
Affiliation(s)
- Ghassem Rezaei
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran; Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Daghighi
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Raoufi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mahban Rahimifard
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sara Kamalzare
- Department of Hepatitis and HIV, Pasteur Institute of Iran, Tehran, IR, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mohammad Abdollahi
- Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaee
- Department of Gastroenterology-Hepatology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran.
| |
Collapse
|
130
|
Li F, Wang Y, Chen WL, Wang DD, Zhou YJ, You BG, Liu Y, Qu CX, Yang SD, Chen MT, Zhang XN. Co-delivery of VEGF siRNA and Etoposide for Enhanced Anti-angiogenesis and Anti-proliferation Effect via Multi-functional Nanoparticles for Orthotopic Non-Small Cell Lung Cancer Treatment. Am J Cancer Res 2019; 9:5886-5898. [PMID: 31534526 PMCID: PMC6735374 DOI: 10.7150/thno.32416] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Targeting tumor angiogenesis pathway via VEGF siRNA (siVEGF) has shown great potential in treating highly malignant and metastatic non-small cell lung cancer (NSCLC). However, anti-angiogenic monotherapy lacked sufficient antitumor efficacy which suffered from malignant tumor proliferation. Therefore, the combined application of siVEGF and chemotherapeutic agents for simultaneous targeting of tumor proliferation and angiogenesis has been a research hotspot to explore a promising NSCLC therapy regimen. Methods: We designed, for the first time, a rational therapy strategy via intelligently co-delivering siVEGF and chemotherapeutics etoposide (ETO) by multi-functional nanoparticles (NPs) directed against the orthotopic NSCLC. These NPs consisted of cationic liposomes loaded with siVEGF and ETO and then coated with versatile polymer PEGylated histidine-grafted chitosan-lipoic acid (PHCL). We then comprehensively evaluated the anti-angiogenic and anti-proliferation efficiency in the in vitro tumor cell model and in bioluminescent orthotopic lung tumor bearing mice model. Results: The NPs co-delivering siVEGF and ETO exhibited tailor-made surface charge reversal features in mimicking tumor extracellular environment with improved internal tumor penetration capacity and higher cellular internalization. Furthermore, these NPs with flexible particles size triggered by intracellular acidic environment and redox environment showed pinpointed and sharp intracellular cargo release guaranteeing adequate active drug concentration in tumor cells. Enhanced VEGF gene expression silencing efficacy and improved tumor cell anti-proliferation effect were demonstrated in vitro. In addition, the PHCL layer improved the stability of these NPs in neutral environment allowing enhanced orthotopic lung tumor targeting efficiency in vivo. The combined therapy by siVEGF and ETO co-delivered NPs for orthotopic NSCLC simultaneously inhibited tumor proliferation and tumor angiogenesis resulting in more significant suppression of tumor growth and metastasis than monotherapy. Conclusion: Combined application of siVEGF and ETO by the multi-functional NPs with excellent and on-demand properties exhibited the desired antitumor effect on the orthotopic lung tumor. Our work has significant potential in promoting combined anti-angiogenesis therapy and chemotherapy regimen for clinical NSCLC treatment.
Collapse
|
131
|
Quijia Quezada C, Azevedo CS, Charneau S, Santana JM, Chorilli M, Carneiro MB, Bastos IMD. Advances in nanocarriers as drug delivery systems in Chagas disease. Int J Nanomedicine 2019; 14:6407-6424. [PMID: 31496694 PMCID: PMC6691952 DOI: 10.2147/ijn.s206109] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Chagas disease is one of the most important public health problems in Latin America due to its high mortality and morbidity levels. There is no effective treatment for this disease since drugs are usually toxic with low bioavailability. Serious efforts to achieve disease control and eventual eradication have been unsuccessful to date, emphasizing the need for rapid diagnosis, drug development, and a reliable vaccine. Novel systems for drug and vaccine administration based on nanocarriers represent a promising avenue for Chagas disease treatment. Nanoparticulate systems can reduce toxicity, and increase the efficacy and bioavailability of active compounds by prolonging release, and therefore improve the therapeutic index. Moreover, nanoparticles are able to interact with the host's immune system, modulating the immune response to favour the elimination of pathogenic microorganisms. In addition, new advances in diagnostic assays, such as nanobiosensors, are beneficial in that they enable precise identification of the pathogen. In this review, we provide an overview of the strategies and nanocarrier-based delivery systems for antichagasic agents, such as liposomes, micelles, nanoemulsions, polymeric and non-polymeric nanoparticles. We address recent progress, with a particular focus on the advances of nanovaccines and nanodiagnostics, exploring new perspectives on Chagas disease treatment.
Collapse
Affiliation(s)
- Christian Quijia Quezada
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
- Department of Drugs and Medicines, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Clênia S Azevedo
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Sébastien Charneau
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Jaime M Santana
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marcella B Carneiro
- Electron Microscopy Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | - Izabela Marques Dourado Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| |
Collapse
|
132
|
Liu J, Zhang R, Xu ZP. Nanoparticle-Based Nanomedicines to Promote Cancer Immunotherapy: Recent Advances and Future Directions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900262. [PMID: 30908864 DOI: 10.1002/smll.201900262] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/21/2019] [Indexed: 05/27/2023]
Abstract
Cancer immunotherapy is a promising cancer terminator by directing the patient's own immune system in the fight against this challenging disorder. Despite the monumental therapeutic potential of several immunotherapy strategies in clinical applications, the efficacious responses of a wide range of immunotherapeutic agents are limited in virtue of their inadequate accumulation in the tumor tissue and fatal side effects. In the last decades, increasing evidences disclose that nanotechnology acts as an appealing solution to address these technical barriers via conferring rational physicochemical properties to nanomaterials. In this Review, an imperative emphasis will be drawn from the current understanding of the effect of a nanosystem's structure characteristics (e.g., size, shape, surface charge, elasticity) and its chemical modification on its transport and biodistribution behavior. Subsequently, rapid-moving advances of nanoparticle-based cancer immunotherapies are summarized from traditional vaccine strategies to recent novel approaches, including delivery of immunotherapeutics (such as whole cancer cell vaccines, immune checkpoint blockade, and immunogenic cell death) and engineered immune cells, to regulate tumor microenvironment and activate cellular immunity. The future prospects may involve in the rational combination of a few immunotherapies for more efficient cancer inhibition and elimination.
Collapse
Affiliation(s)
- Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
133
|
Patsula V, Horák D, Kučka J, Macková H, Lobaz V, Francová P, Herynek V, Heizer T, Páral P, Šefc L. Synthesis and modification of uniform PEG-neridronate-modified magnetic nanoparticles determines prolonged blood circulation and biodistribution in a mouse preclinical model. Sci Rep 2019; 9:10765. [PMID: 31341232 PMCID: PMC6656745 DOI: 10.1038/s41598-019-47262-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/05/2019] [Indexed: 12/19/2022] Open
Abstract
Magnetite (Fe3O4) nanoparticles with uniform sizes of 10, 20, and 31 nm were prepared by thermal decomposition of Fe(III) oleate or mandelate in a high-boiling point solvent (>320 °C). To render the particles with hydrophilic and antifouling properties, their surface was coated with a PEG-containing bisphosphonate anchoring group. The PEGylated particles were characterized by a range of physicochemical methods, including dynamic light scattering, transmission electron microscopy, thermogravimetric analysis, Fourier transform infrared spectroscopy, and magnetization measurements. As the particle size increased from 10 to 31 nm, the amount of PEG coating decreased from 28.5 to 9 wt.%. The PEG formed a dense brush-like shell on the particle surface, which prevented particles from aggregating in water and PBS (pH 7.4) and maximized the circulation time in vivo. Magnetic resonance relaxometry confirmed that the PEG-modified Fe3O4 nanoparticles had high relaxivity, which increased with increasing particle size. In the in vivo experiments in a mouse model, the particles provided visible contrast enhancement in the magnetic resonance images. Almost 70% of administrated 20-nm magnetic nanoparticles still circulated in the blood stream after four hours; however, their retention in the tumor was rather low, which was likely due to the antifouling properties of PEG.
Collapse
Affiliation(s)
- Vitalii Patsula
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Daniel Horák
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic.
| | - Jan Kučka
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Hana Macková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Volodymyr Lobaz
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague 6, Czech Republic
| | - Pavla Francová
- Center of Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| | - Vít Herynek
- Center of Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| | - Tomáš Heizer
- Center of Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| | - Petr Páral
- Center of Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| | - Luděk Šefc
- Center of Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Salmovská 3, 120 00, Prague 2, Czech Republic
| |
Collapse
|
134
|
Lynn GM, Laga R, Jewell CM. Induction of anti-cancer T cell immunity by in situ vaccination using systemically administered nanomedicines. Cancer Lett 2019; 459:192-203. [PMID: 31185250 DOI: 10.1016/j.canlet.2019.114427] [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: 01/16/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
Abstract
Patients with inadequate anti-cancer T cell responses experience limited benefit from immune checkpoint inhibitors and other immunotherapies that require T cells. Therefore, treatments that induce de novo anti-cancer T cell immunity are needed. One strategy - referred to as in situ vaccination - is to deliver chemotherapeutic or immunostimulatory drugs into tumors to promote cancer cell death and provide a stimulatory environment for priming T cells against antigens already present in the tumor. However, achieving sufficient drug concentrations in tumors without causing dose-limiting toxicities remains a major challenge. To address this challenge, nanomedicines based on nano-sized carriers ('nanocarriers') of chemotherapeutics and immunostimulants are being developed to improve drug accumulation in tumors following systemic (intravenous) administration. Herein, we present the rationale for using systemically administrable nanomedicines to induce anti-cancer T cell immunity via in situ vaccination and provide an overview of synthetic nanomedicines currently used clinically. We also describe general strategies for improving nanomedicine design to increase tumor uptake, including use of micelle- and star polymer-based nanocarriers. We conclude with perspectives for how nanomedicine properties, host factors and treatment combinations can be leveraged to maximize efficacy.
Collapse
Affiliation(s)
- Geoffrey M Lynn
- Fischell Department of Bioengineering, A. James Clark Hall, Room 5110, 8278 Paint Branch Drive, College Park, MD, 20742, USA; Avidea Technologies, Baltimore, MD, 21205, USA
| | - Richard Laga
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovského nám. 2, 162 06, Prague, Czech Republic
| | - Christopher M Jewell
- Fischell Department of Bioengineering, A. James Clark Hall, Room 5110, 8278 Paint Branch Drive, College Park, MD, 20742, USA; United States Department of Veterans Affairs, VA Maryland Health Care System, 10 North Greene Street, Baltimore, MD, 21201, USA; Robert E. Fischell Institute for Biomedical Devices, A. James Clark Hall, Room 5110, 8278 Paint Branch Drive, College Park, MD, 20742, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Cancer Center, Executive Office, Suite N9E17, 22 S. Greene Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
135
|
Self-Assembled Benznidazole-Loaded Cationic Nanoparticles Containing Cholesterol/Sialic Acid: Physicochemical Properties, In Vitro Drug Release and In Vitro Anticancer Efficacy. Int J Mol Sci 2019; 20:ijms20092350. [PMID: 31083590 PMCID: PMC6539689 DOI: 10.3390/ijms20092350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/12/2022] Open
Abstract
Cationic polymeric nanoparticles (NPs) have the ability to overcome biological membranes, leading to improved efficacy of anticancer drugs. The modulation of the particle-cell interaction is desired to control this effect and avoid toxicity to normal cells. In this study, we explored the surface functionalization of cationic polymethylmethacrylate (PMMA) NPs with two natural compounds, sialic acid (SA) and cholesterol (Chol). The performance of benznidazole (BNZ) was assessed in vitro in the normal renal cell line (HEK-293) and three human cancer cell lines, as follows: human colorectal cancer (HT-29), human cervical carcinoma (HeLa), and human hepatocyte carcinoma (HepG2). The structural properties and feasibility of NPs were evaluated and the changes induced by SA and Chol were determined by using multiple analytical approaches. Small (<200 nm) spherical NPs, with a narrow size distribution and high drug-loading efficiency were prepared by using a simple and reproducible emulsification solvent evaporation method. The drug interactions in the different self-assembled NPs were assessed by using Fourier transform-infrared spectroscopy. All formulations exhibited a slow drug-release profile and physical stability for more than 6 weeks. Both SA and Chol changed the kinetic properties of NPs and the anticancer efficacy. The feasibility and potential of SA/Chol-functionalized NPs has been demonstrated in vitro in the HEK-293, HepG2, HeLa, and HT-29 cell lines as a promising system for the delivery of BNZ.
Collapse
|
136
|
Li B, Lane LA. Probing the biological obstacles of nanomedicine with gold nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1542. [PMID: 30084539 PMCID: PMC6585966 DOI: 10.1002/wnan.1542] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Despite massive growth in nanomedicine research to date, the field still lacks fundamental understanding of how certain physical and chemical features of a nanoparticle affect its ability to overcome biological obstacles in vivo and reach its intended target. To gain fundamental understanding of how physical and chemical parameters affect the biological outcomes of administered nanoparticles, model systems that can systematically manipulate a single parameter with minimal influence on others are needed. Gold nanoparticles are particularly good model systems in this case as one can synthetically control the physical dimensions and surface chemistry of the particles independently and with great precision. Additionally, the chemical and physical properties of gold allow particles to be detected and quantified in tissues and cells with high sensitivity. Through systematic biological studies using gold nanoparticles, insights toward rationally designed nanomedicine for in vivo imaging and therapy can be obtained. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Bin Li
- Department of Biomedical Engineering, College of Engineering and Applied SciencesNanjing UniversityNanjingJiangsuChina
| | - Lucas A. Lane
- Department of Biomedical Engineering, College of Engineering and Applied SciencesNanjing UniversityNanjingJiangsuChina
| |
Collapse
|
137
|
Yan X, Sedykh A, Wang W, Zhao X, Yan B, Zhu H. In silico profiling nanoparticles: predictive nanomodeling using universal nanodescriptors and various machine learning approaches. NANOSCALE 2019; 11:8352-8362. [PMID: 30984943 DOI: 10.1039/c9nr00844f] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Rational nanomaterial design is urgently demanded for new nanomaterial development with desired properties. However, computational nanomaterial modeling and virtual nanomaterial screening are not applicable for this purpose due to the complexity of nanomaterial structures. To address this challenge, a new computational workflow is established in this study to virtually profile nanoparticles by (1) constructing a structurally diverse virtual gold nanoparticle (GNP) library and (2) developing novel universal nanodescriptors. The emphasis of this study is the second task by developing geometrical nanodescriptors that are suitable for the quantitative modeling of GNPs and virtual screening purposes. The feasibility, rigor and applicability of this novel computational method are validated by testing seven GNP datasets consisting of 191 unique GNPs of various nano-bioactivities and physicochemical properties. The high predictability of the developed GNP models suggests that this workflow can be used as a universal tool for nanomaterial profiling and rational nanomaterial design.
Collapse
Affiliation(s)
- Xiliang Yan
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | | | | | | | | | | |
Collapse
|
138
|
Xu X, Angioletti-Uberti S, Lu Y, Dzubiella J, Ballauff M. Interaction of Proteins with Polyelectrolytes: Comparison of Theory to Experiment. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:5373-5391. [PMID: 30095921 DOI: 10.1021/acs.langmuir.8b01802] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We discuss recent investigations of the interaction of polyelectrolytes with proteins. In particular, we review our recent studies on the interaction of simple proteins such as human serum albumin (HSA) and lysozyme with linear polyelectrolytes, charged dendrimers, charged networks, and polyelectrolyte brushes. In all cases discussed here, we combined experimental work with molecular dynamics (MD) simulations and mean-field theories. In particular, isothermal titration calorimetry (ITC) has been employed to obtain the respective binding constants Kb and the Gibbs free energy of binding. MD simulations with explicit counterions but implicit water demonstrate that counterion release is the main driving force for the binding of proteins to strongly charged polyelectrolytes: patches of positive charges located on the surface of the protein become multivalent counterions of the polyelectrolyte, thereby releasing a number of counterions condensed on the polyelectrolyte. The binding Gibbs free energy due to counterion release is predicted to scale with the logarithm of the salt concentration in the system, which is verified by both simulations and experiment. In several cases, namely, for the interaction of proteins with linear polyelectrolytes and highly charged hydrophilic dendrimers, the binding constant could be calculated from simulations to very good approximation. This finding demonstrated that in these cases explicit hydration effects do not contribute to the Gibbs free energy of binding. The Gibbs free energy can also be used to predict the kinetics of protein uptake by microgels for a given system by applying dynamic density functional theory. The entire discussion demonstrates that the direct comparison of theory with experiments can lead to a full understanding of the interaction of proteins with charged polymers. Possible implications for applications, such as drug design, are discussed.
Collapse
Affiliation(s)
- Xiao Xu
- School of Chemical Engineering , Nanjing University of Science and Technology , 200 Xiao Ling Wei , Nanjing 210094 , P. R. China
| | - Stefano Angioletti-Uberti
- Department of Materials , Imperial College London , London SW7 2AZ - UK , U.K
- International Research Centre for Soft Matter , Beijing University of Chemical Technology , 100099 Beijing , PR China
| | - Yan Lu
- Soft Matter and Functional Materials , Helmholtz-Zentrum Berlin für Materialien und Energie GmbH , 14109 Berlin , Germany
- Institute of Chemistry , University of Potsdam , 14467 Potsdam , Germany
| | - Joachim Dzubiella
- Soft Matter and Functional Materials , Helmholtz-Zentrum Berlin für Materialien und Energie GmbH , 14109 Berlin , Germany
- Physikalisches Institut , Albert-Ludwigs-Universität , 79104 Freiburg , Germany
| | - Matthias Ballauff
- Soft Matter and Functional Materials , Helmholtz-Zentrum Berlin für Materialien und Energie GmbH , 14109 Berlin , Germany
- Institut für Physik , Humboldt-Universität zu Berlin , 12489 Berlin , Germany
| |
Collapse
|
139
|
Tang JC, Partono A, Anvari B. Near-Infrared-Fluorescent Erythrocyte-Mimicking Particles: Physical and Optical Characteristics. IEEE Trans Biomed Eng 2019; 66:1034-1044. [PMID: 30130175 PMCID: PMC6382600 DOI: 10.1109/tbme.2018.2866368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exogenous fluorescent materials activated by near-infrared (NIR) light can offer deep optical imaging with subcellular resolution, and enhanced image contrast. We have engineered NIR particles by doping hemoglobin-depleted erythrocyte ghosts (EGs) with indocyanine green (ICG). We refer to these optical particles as NIR erythrocyte-mimicking transducers (NETs). A particular feature of NETs is that their diameters can be tuned from micrometer to nanometer scale, thereby, providing a capability for broad NIR biomedical imaging applications. Herein, we investigate the effects of ICG concentration on key material properties of micrometer-sized NETs, and nanometer-sized NETs fabricated by either sonication or mechanical extrusion of EGs. The zeta potentials of NETs do not vary significantly with ICG concentration, suggesting that ICG is encapsulated within NETs regardless of particle size or ICG concentration. Loading efficiency of ICG into the NETs monotonically decreases with increasing values of ICG concentration. Based on quantitative analyses of the fluorescence emission spectra of the NETs, we determine that 20 μM ICG utilized during fabrication of NETs presents an optimal concentration that maximizes the integrated fluorescence emission for micrometer- and nanometer-sized NETs. Encapsulation of the ICG in these constructs also enhances the fluorescence stability and quantum yield of ICG. These results guide the engineering of NETs with maximal NIR emission for imaging applications such as fluorescence-guided tumor resection and real-time angiography.
Collapse
|
140
|
Zhao Z, Ukidve A, Krishnan V, Mitragotri S. Effect of physicochemical and surface properties on in vivo fate of drug nanocarriers. Adv Drug Deliv Rev 2019; 143:3-21. [PMID: 30639257 DOI: 10.1016/j.addr.2019.01.002] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/07/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023]
Abstract
Over the years, a plethora of materials - natural and synthetic - have been engineered at a nanoscopic level and explored for drug delivery. Nanocarriers based on such materials could improve the payload's pharmacokinetics and achieve the desired pharmacological response at the target tissue. Despite the development of rationally designed drug nanocarriers, only a handful of such formulations have been successfully translated into the clinic. The physicochemical properties (size, shape, surface chemistry, porosity, elasticity, and many others) of these nanocarriers influence its biological identity, which in presence of biological barriers in vivo, could significantly modulate the therapeutic index of its cargo and alter the desired outcome. Further, complexities associated with developing effective drug nanocarriers have led to conflicting views of its safety, permeation of biological barriers and cellular uptake. Here, in this review, we emphasize the effect of physicochemical properties of nanocarriers on their interactions with the biological milieu. The review will discuss in depth, how modulating the physicochemical properties would influence a drug nanocarrier's behavior in vivo and the mechanisms underlying these effects. The goal of this review is to summarize the design considerations based on these properties and to provide a conceptual template for achieving improved therapeutic efficacy with enhanced patient compliance.
Collapse
|
141
|
Abstract
Bioorthogonal nanocatalysts in the form of 'nanozymes', are promising tools for generating imaging and therapeutic molecules in living systems. These systems use transformations developed by synthetic chemists to effect transformations that cannot be performed by cellular machinery. This emerging platform is rapidly evolving towards the creation of smart nanodevices featuring the capabilities of their enzyme prototypes, modulating catalytic activity through structure as well as chemical and physical signals. Here we describe different strategies to fabricate these nanocatalysts and their potential in diagnostic and therapeutic applications.
Collapse
|
142
|
Allen SD, Bobbala S, Karabin NB, Scott EA. On the advancement of polymeric bicontinuous nanospheres toward biomedical applications. NANOSCALE HORIZONS 2019; 4:258-272. [PMID: 32254084 DOI: 10.1039/c8nh00300a] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Self-assembled soft nanocarriers that are capable of simultaneous encapsulation of both lipophilic and water soluble payloads have significantly enhanced controlled delivery applications in biomedicine. These nanoarchitectures, such as liposomes, polymersomes and cubosomes, are primarily composed of either amphiphilic polymers or lipids, with the polymeric variants generally possessing greater stability and control over biodistribution and bioresponsive release. Polymersomes have long demonstrated such advantages over their lipid analogs, liposomes, but only recently have bicontinuous nanospheres emerged as a polymeric cubic phase alternative to lipid cubosomes. In this review, we summarize the current state of the field for bicontinuous nanosphere formulation and characterization and suggest future directions for this nascent delivery platform as it is adopted for biomedical applications.
Collapse
Affiliation(s)
- Sean D Allen
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, USA.
| | | | | | | |
Collapse
|
143
|
Krug P, Mielczarek L, Wiktorska K, Kaczyńska K, Wojciechowski P, Andrzejewski K, Ofiara K, Szterk A, Mazur M. Sulforaphane-conjugated selenium nanoparticles: towards a synergistic anticancer effect. NANOTECHNOLOGY 2019; 30:065101. [PMID: 30523968 DOI: 10.1088/1361-6528/aaf150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sulforaphane-modified selenium nanoparticles can be prepared in a simple aqueous-phase redox reaction through reduction of selenite with ascorbic acid. The sulforaphane molecules present in the reaction mixture adsorb on the nanoparticle surface, forming an adlayer. The resulting conjugate was examined with several physicochemical techniques, including microscopy, spectroscopy, x-ray diffraction, dynamic light scattering and zeta potential measurements. As shown in in vivo investigations on rats, the nanomaterial administered intraperitoneally is eliminated mainly in urine (and, to a lesser extent, in feces); however, it is also retained in the body. The modified nanoparticles mainly accumulate in the liver, but the basic parameters of blood and urine remain within normal limits. The sulforaphane-conjugated nanoparticles reveal considerable anticancer action, as demonstrated on several cancer cell cultures in vitro. This finding is due to the synergistic effect of elemental selenium and sulforaphane molecules assembled in one nanostructure (conjugate). On the other hand, the cytotoxic action on normal cells is relatively low. The high antitumor activity and selectivity of the conjugate with respect to diseased and healthy cells is extremely promising from the point of view of cancer treatment.
Collapse
Affiliation(s)
- Pamela Krug
- Department of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Pawar VK, Singh Y, Sharma K, Shrivastav A, Sharma A, Singh A, Meher JG, Singh P, Raval K, Kumar A, Bora HK, Datta D, Lal J, Chourasia MK. Improved chemotherapy against breast cancer through immunotherapeutic activity of fucoidan decorated electrostatically assembled nanoparticles bearing doxorubicin. Int J Biol Macromol 2019; 122:1100-1114. [DOI: 10.1016/j.ijbiomac.2018.09.059] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022]
|
145
|
Ghosh S, Lalani R, Patel V, Bardoliwala D, Maiti K, Banerjee S, Bhowmick S, Misra A. Combinatorial nanocarriers against drug resistance in hematological cancers: Opportunities and emerging strategies. J Control Release 2019; 296:114-139. [DOI: 10.1016/j.jconrel.2019.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/16/2022]
|
146
|
Zhang L, Hao P, Yang D, Feng S, Peng B, Appelhans D, Zhang T, Zan X. Designing nanoparticles with improved tumor penetration: surface properties from the molecular architecture viewpoint. J Mater Chem B 2019; 7:953-964. [PMID: 32255100 DOI: 10.1039/c8tb03034k] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer is the second most common cause of death, and nanomedicine is regarded as one of the strategies that may revolutionize cancer treatments. However, the tumor microenvironment (e.g., increased interstitial fluid pressure and dense extracellular matrix) hinders the penetration of nanomedicine into tumor cells, which leads to a short acting time and low drug concentration with tumors, eventually leading to a high recurrence rate and therapeutic failure in clinics. Developing a delivery system with deep penetration ability into the tumor has always been pursued and highly desirable for cancer treatments. Inspired by the high cellular uptake efficiency of enveloped viruses with rough and nanoscale surfaces, we constructed polystyrene nanoparticles (NPs) with similar sizes and charges, but with different surface topologies at the molecular level, by conjugating poly(propylene imine) (PPI) dendrimers with different generations onto the NPs. We found that subtle changes made to the surficial chemical properties led to changes in surface roughness and wettability, which considerably influenced the cellular internalization, endocytosis mechanism, and penetration into the tumor model both in vitro and in vivo. This will shed light on the future design of drug delivery vehicles and facilitate understanding the interactions between NP surfaces and cells, as well as tumor penetration.
Collapse
Affiliation(s)
- Long Zhang
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Yao Q, Yuan X, Chen T, Leong DT, Xie J. Engineering Functional Metal Materials at the Atomic Level. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1802751. [PMID: 30118559 DOI: 10.1002/adma.201802751] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/14/2018] [Indexed: 05/20/2023]
Abstract
With continuous research efforts devoted into synthesis and characterization chemistry of functional nanomaterials in the past decades, the development of metal materials is stepping into a new era, where atom-by-atom customization of property-dictating structural attributes is expected. Herein, the state-of-the-art modulation of functional metal nanomaterials at the atomic level, by size- and structure-controlled synthesis of thiolate-protected metal (e.g., Au and Ag) nanoclusters (NCs), is exemplified. Metal NCs are ultrasmall (<3 nm) particles with hierarchical primary, secondary, and tertiary structures, reminiscent of natural proteins or enzymes. Given the proven dependence of their physicochemical properties on their size and structure, documented synthetic methodologies delivering NCs with atomic-level monodispersity and tailorable size and structural attributes at individual hierarchical levels are categorized and discussed. Such assured atomic-level modulation could confer metal NCs with novel application opportunities in diverse fields, which are also exemplified by their size- and structure-dictated catalytic and biomedical performance. The precise synthesis and application chemistry developed based on the hierarchical structure scheme of metal NCs could increase the acceptance of metal NCs as a new family of functional materials.
Collapse
Affiliation(s)
- Qiaofeng Yao
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Xun Yuan
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Shibei District, Qingdao, Shandong Province, 266042, China
| | - Tiankai Chen
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Jianping Xie
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
148
|
Le Guével X, Henry M, Motto-Ros V, Longo E, Montañez MI, Pelascini F, de La Rochefoucauld O, Zeitoun P, Coll JL, Josserand V, Sancey L. Elemental and optical imaging evaluation of zwitterionic gold nanoclusters in glioblastoma mouse models. NANOSCALE 2018; 10:18657-18664. [PMID: 30264838 DOI: 10.1039/c8nr05299a] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
We report in this study the in vivo biodistribution of ultra-small luminescent gold (Au) particles (∼1.5 nm core size; 17 kDa), so-called nanoclusters (NCs), stabilized by bidentate zwitterionic molecules in subcutaneous (s.c.) and orthotopic glioblastoma mice models. Particular investigations on renal clearance and tumor uptake were performed using highly sensitive advanced imaging techniques such as multi-elemental Laser-Induced Breakdown Spectroscopy (LIBS) imaging and in-line X-ray Synchrotron Phase Contrast Tomography (XSPCT). Results show a blood circulation time of 6.5 ± 1.3 min accompanied by an efficient and fast renal clearance through the cortex of the kidney with a 66% drop between 1 h and 5 h. With a similar size range, these Au NCs are 5 times more fluorescent than the well-described Au25GSH18 NCs in the near-infrared (NIR) region and present significantly stronger tumor uptake and retention illustrated by an in vivo s.c. tumor-to-skin ratio of 1.8 measured by non-invasive optical imaging and an ex vivo tumor-to-muscle of 6.1. This work highlights the pivotal role of surface coating in designing optimum Au NC candidates for cancer treatment.
Collapse
Affiliation(s)
- Xavier Le Guével
- Cancer Targets & Experimental Therapeutics, Institute for Advanced Biosciences (IAB), University of Grenoble Alpes - INSERM U1209 - CNRS UMR 5309- 38000, Grenoble, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Raut S, Mooberry L, Sabnis N, Garud A, Dossou AS, Lacko A. Reconstituted HDL: Drug Delivery Platform for Overcoming Biological Barriers to Cancer Therapy. Front Pharmacol 2018; 9:1154. [PMID: 30374303 PMCID: PMC6196266 DOI: 10.3389/fphar.2018.01154] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Drug delivery to malignant tumors is limited by several factors, including off-target toxicities and suboptimal benefits to cancer patient. Major research efforts have been directed toward developing novel technologies involving nanoparticles (NPs) to overcome these challenges. Major obstacles, however, including, opsonization, transport across cancer cell membranes, multidrug-resistant proteins, and endosomal sequestration of the therapeutic agent continue to limit the efficiency of cancer chemotherapy. Lipoprotein-based drug delivery technology, "nature's drug delivery system," while exhibits highly desirable characteristics, it still needs substantial investment from private/government stakeholders to promote its eventual advance to the bedside. Consequently, this review focuses specifically on the synthetic (reconstituted) high-density lipoprotein rHDL NPs, evaluating their potential to overcome specific biological barriers and the challenges of translation toward clinical utilization and commercialization. This highly robust drug transport system provides site-specific, tumor-selective delivery of anti-cancer agents while reducing harmful off-target effects. Utilizing rHDL NPs for anti-cancer therapeutics and tumor imaging revolutionizes the future strategy for the management of a broad range of cancers and other diseases.
Collapse
Affiliation(s)
- Sangram Raut
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Linda Mooberry
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Nirupama Sabnis
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Ashwini Garud
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Akpedje Serena Dossou
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Andras Lacko
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
150
|
Gurnani P, Sanchez-Cano C, Abraham K, Xandri-Monje H, Cook AB, Hartlieb M, Lévi F, Dallmann R, Perrier S. RAFT Emulsion Polymerization as a Platform to Generate Well-Defined Biocompatible Latex Nanoparticles. Macromol Biosci 2018; 18:e1800213. [PMID: 30085410 DOI: 10.1002/mabi.201800213] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/03/2018] [Indexed: 12/17/2022]
Abstract
Current approaches to generate core-shell nanoparticles for biomedical applications are limited by factors such as synthetic scalability and circulatory desorption of cytotoxic surfactants. Developments in controlled radical polymerization, particularly in dispersed states, represent a promising method of overcoming these challenges. In this work, well-defined PEGylated nanoparticles are synthesized using reversible addition fragmentation chain transfer emulsion polymerization to control particle size and surface composition and were further characterized with light scattering, electron microscopy, and size exclusion chromatography. Importantly, the nanoparticles are found to be tolerated both in vitro and in vivo, without the need for any purification after particle synthesis. Pharmacokinetic and biodistribution studies in mice, following intraperitoneal injection of the nanoparticles, reveal a long (>76 h) circulation time and accumulation in the liver.
Collapse
Affiliation(s)
- Pratik Gurnani
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Carlos Sanchez-Cano
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Kristin Abraham
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Helena Xandri-Monje
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Alexander B Cook
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Matthias Hartlieb
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Francis Lévi
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Robert Dallmann
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Sébastien Perrier
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| |
Collapse
|