101
|
Jackson J, Leung D, Burt H. The use of ultrasound to increase the uptake and cytotoxicity of dual taxane and P-glycoprotein inhibitor loaded, solid core nanoparticles in drug resistant cells. ULTRASONICS 2020; 101:106033. [PMID: 31561207 DOI: 10.1016/j.ultras.2019.106033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/02/2019] [Accepted: 09/17/2019] [Indexed: 06/10/2023]
Abstract
The objective of this study was to use ultrasound in combination with nanoparticulate formulations of taxane drugs for an additive approach to overcome multidrug resistance (MDR). Polymeric nanoparticulate formulations containing both chemotherapeutic taxane drugs and a polymeric inhibitor (MePEG17-b-PCL5) of drug resistant proteins have been previously developed in an attempt to overcome MDR in cells. High frequency (>1 MHz) ultrasound has been shown to increase the uptake of cytotoxic drugs in MDR proliferating cells and has been suggested as a different way to overcome MDR, resensitize drug resistant cancer cells and allow for chemotherapeutic efficacy. MDCK-MDR cells were incubated with docetaxel (DTX) or paclitaxel (PTX) loaded, solid core, nanoparticles made from a 50:50 ratio of two diblock copolymers, MePEG114-b-PCL200 and MePEG17-b-PCL5 (PCL200/PCL5). The accumulation of drug in MDCK-MDR cells was measured using radiolabeled drug and the viability of cells was determined using an MTS cell proliferation assay. The effect of ultrasound (4 MHz, 32 W/cm2, 10 s, 25% duty cycle) on drug uptake and cell viability was studied. Using free DTX or PTX, MDCK-MDR cells were killed at sublethal doses of drug with the P-gp inhibitor (MePEG17-b-PCL5) present at a concentration of just 0.006% (m/v) and cell death began after just 3 h of incubation. Using sublethal incubation doses of PTX or DTX in PCL200/PCL5 nanoparticles for 90 min, followed by a second exposure to blank PCL200/PCL5 nanoparticles, cell viability dropped by approximately 60% at 24 h. Drug accumulation increased by 1.43-1.9 fold following five bursts of ultrasound applied at 90 min. Both, increased ultrasound exposure and increased concentrations of blank nanoparticles during the second incubation allowed for increased levels of cell death. The combined use of ultrasound with taxane and P-gp inhibitor loaded polymeric nanoparticles may allow for increased accumulation of drug and inhibitor which may then release both agents inside cells in a controlled manner to overcome drug resistance in MDR cells.
Collapse
Affiliation(s)
- John Jackson
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, B.C., Canada.
| | - Donna Leung
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, B.C., Canada
| | - Helen Burt
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, B.C., Canada
| |
Collapse
|
102
|
Lucchetti D, Perelli L, Colella F, Ricciardi-Tenore C, Scoarughi GL, Barbato G, Boninsegna A, De Maria R, Sgambato A. Low-intensity pulsed ultrasound affects growth, differentiation, migration, and epithelial-to-mesenchymal transition of colorectal cancer cells. J Cell Physiol 2020; 235:5363-5377. [PMID: 31967331 DOI: 10.1002/jcp.29423] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022]
Abstract
Ultrasound (US) offers potentially important opportunities from a therapeutic point of view. Thus, the study of the biological effects of US on cancer cells is important to understand the consequences of these changes on the malignant phenotype. This study aimed to investigate the effects of low-intensity ultrasound (LIPUS) on the phenotype of colorectal cancer cell lines. Cell proliferation was evaluated by viability test and by evaluation of pERK expression, while cell motility using the scratch test. Cell differentiation was evaluated assessing alkaline phosphatase activity. Epithelial mesenchymal transition was assessed by analyzing the expression of Vimentin and E-Cadherin. Release and uptake of extracellular vesicles (EVs) were evaluated by flow cytometry. LIPUS effects on the organization of cytoskeleton were analyzed by confocal microscopy and by evaluation of Rho GTPase expression. No alterations in vitality and clonogenicity were observed when the intermediate (0.4 MPa) and the lowest (0.035 MPa) acoustic intensities were administered while the treatment with high intensity (1 MPa) induced a reduction of both cell viability and clonogenicity in both cell lines in a frequency-dependent manner. LIPUS promoted the differentiation of colon cancer cells, affected epithelial-to-mesenchymal transition, promoted the closure of a wound as well as increased the release of EVs compared with untreated cells. LIPUS-induced increase in cell motility was likely due to a Rho GTPase-dependent mechanism. Overall, the results obtained warrant further studies on the potential combined effect of LIPUS with differentiating agents and on their potential use in a clinical setting.
Collapse
Affiliation(s)
- Donatella Lucchetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Institute of General Pathology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Luigi Perelli
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Filomena Colella
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Roma, Italy
| | | | | | | | - Alma Boninsegna
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ruggero De Maria
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.,Institute of General Pathology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Alessandro Sgambato
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Roma, Italy.,Scientific Direction, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| |
Collapse
|
103
|
Zhong S, Ling Z, Zhou Z, He J, Ran H, Wang Z, Zhang Q, Song W, Zhang Y, Luo J. Herceptin-decorated paclitaxel-loaded poly(lactide- co-glycolide) nanobubbles: ultrasound-facilitated release and targeted accumulation in breast cancers. Pharm Dev Technol 2020; 25:454-463. [PMID: 31873051 DOI: 10.1080/10837450.2019.1709500] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Ultrasound can promote the drug release from drug-loaded substances and alter the tumor local microenvironment to facilitate the transport of drug carriers into the tumor tissues. Based on the altered tumor microenvironment, nanobubbles (NBs) as drug carriers with surfaces functionalized with targeting ligands can reach the tumor sites, thereby increasing the efficacy of chemotherapy. Herein, paclitaxel (PTX)-loaded poly(lactide-co-glycolide) (PLGA) NBs are prepared as drug carriers with covalently conjugated herceptin (anti-HER2 monoclonal antibody) on the surface to guide the target. The effect of ultrasound on the drug release and targeting of the herceptin-conjugated drug-loaded nanobubbles (PTX-NBs-HER) on the cancerous cells is determined. The use of ultrasound significantly improves the cell targeting capability in vitro, and efficiency of enhanced permeability and retention in vivo. The combination of PTX-NBs-HER and ultrasound facilitates the release of PTX, as well as the uptake and cell apoptosis in vitro. The in vivo application of both PTX-NBs-HER and ultrasound enhances the PTX targeting and accumulation in breast cancers while reducing the transmission and distribution of PTX in healthy organs. The combination of ultrasound with PTX-NBs-HER as contrast agents and drug carriers affords an image-guided drug delivery system for the precise targeted therapy of tumors.
Collapse
Affiliation(s)
- Shigen Zhong
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Zhiyu Ling
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Zhiyi Zhou
- Department of Ultrasound, The General Hospital of Chongqing, Chongqing, China
| | - Jin He
- Department of Ultrasound, The General Hospital of Chongqing, Chongqing, China
| | - Haitao Ran
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Zhigang Wang
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Qunxia Zhang
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Weixiang Song
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing, China
| | - Yong Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Luo
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
104
|
Abstract
Ultrasound and magneto-responsive nanosized drug delivery systems have been designed as novel carriers for controlled release. Colloidal bubbles (CBs) could be designed to incorporate different materials, such as protein, lipid, polymer, surfactants, and even nanoparticles in their shell, which makes them suitable for a wide range of drug delivery applications. The interior of CBs may be filled with different gases, which is essential for conferring the characteristics of an ultrasounds contrasting agent. Manipulating the core of CBs enhances features such as stability and duration of the echogenic effect. Thus CBs derivatized with nanoparticles combine functional properties of CBs and NPs to yield a versatile theranostics platform technology.
Collapse
|
105
|
Žid L, Zeleňák V, Girman V, Bednarčík J, Zeleňáková A, Szűcsová J, Hornebecq V, Hudák A, Šuleková M, Váhovská L. Doxorobicin as cargo in a redox-responsive drug delivery system capped with water dispersible ZnS nanoparticles. RSC Adv 2020; 10:15825-15835. [PMID: 35493685 PMCID: PMC9052939 DOI: 10.1039/d0ra02091e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/13/2020] [Indexed: 11/29/2022] Open
Abstract
In this work, we have prepared and investigated a redox-responsive drug delivery system (DDS) based on a porous carrier. Doxorubicin (DOX), a chemotherapy medication for treatment of different kinds of cancer, was used as a model drug in the study. DOX was loaded in ordered hexagonal mesoporous silica SBA-15, a nanoporous material with good biocompatibility, stability, large pore size and specific surface area (SBET = 908 m2 g−1, VP = 0.79 cm3 g−1, d = 5.9 nm) and easy surface modification. To prepare the redox-responsive system, cystamine derivative ligands, with redox active disulphide linkers were grafted onto the surface of SBA-15. To ensure no significant premature release of DOX from the porous system, thioglycolic acid modified ZnS nanoparticles (ZnS–COOH NPs) were used as pore capping agents. The grafted redox-responsive cystamine derivative ligand containing disulphide linkers was bonded by a peptide bond to the thioglycolic acid groups of ZnS–COOH NPs, capping the pores. Once the disulphide bond was cleaved, the ZnS–COOH NPs caps were released and pores were opened to deliver the DOX cargo. The dithiol bond was cleavable by redox active molecules such as dithiothreitol (DTT) or glutathione, the concentration of which in cancer cells is 4 times higher than in healthy cells. The redox release of DOX was studied in two different media, physiological saline solution with DTT and saline without DTT. The prepared DDS proved the concept of redox responsive release. All samples were characterised by powder X-ray diffraction (XRD), transition electron microscopy (TEM), nitrogen adsorption/desorption at 77 K, Fourier-transform infrared spectroscopy (FTIR), thermal analysis and zeta potential measurements. The presence of semiconducting ZnS nanoparticle caps on the pore openings was detected by magnetic measurements using SQUID magnetometry showing that such cargo systems could be monitored using magnetic measurements which opens up the possibilities of using such drug delivery systems as theranostic agents. Redox-responsive drug delivery system was studied. ZnS nanoparticles served as pore capping agent to prevent premature release of anticancer drug. Such cargo can be monitored by magnetic field which opens possibilities its use in theranostics.![]()
Collapse
Affiliation(s)
- Lukáš Žid
- Department of Inorganic Chemistry Faculty of Science
- P. J. Šafárik University
- SK-041 54 Košice
- Slovak Republic
| | - Vladimír Zeleňák
- Department of Inorganic Chemistry Faculty of Science
- P. J. Šafárik University
- SK-041 54 Košice
- Slovak Republic
| | - Vladimír Girman
- Institute of Physics
- P. J. Šafárik University
- 04001 Košice
- Slovakia
| | - Jozef Bednarčík
- Institute of Physics
- P. J. Šafárik University
- 04001 Košice
- Slovakia
| | | | | | | | - Alexander Hudák
- Department of Chemistry
- Biochemistry and Biophysics
- Institute of Pharmaceutical Chemistry
- The University of Veterinary Medicine and Pharmacy in Košice
- Košice 041 81
| | - Monika Šuleková
- Department of Chemistry
- Biochemistry and Biophysics
- Institute of Pharmaceutical Chemistry
- The University of Veterinary Medicine and Pharmacy in Košice
- Košice 041 81
| | - Lucia Váhovská
- Department of Chemistry
- Biochemistry and Biophysics
- Institute of Pharmaceutical Chemistry
- The University of Veterinary Medicine and Pharmacy in Košice
- Košice 041 81
| |
Collapse
|
106
|
Silvani G, Scognamiglio C, Caprini D, Marino L, Chinappi M, Sinibaldi G, Peruzzi G, Kiani MF, Casciola CM. Reversible Cavitation-Induced Junctional Opening in an Artificial Endothelial Layer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1905375. [PMID: 31762158 DOI: 10.1002/smll.201905375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Indexed: 06/10/2023]
Abstract
Targeting pharmaceuticals through the endothelial barrier is crucial for drug delivery. In this context, cavitation-assisted permeation shows promise for effective and reversible opening of intercellular junctions. A vessel-on-a-chip is exploited to investigate and quantify the effect of ultrasound-excited microbubbles-stable cavitation-on endothelial integrity. In the vessel-on-a-chip, the endothelial cells form a complete lumen under physiological shear stress, resulting in intercellular junctions that exhibit barrier functionality. Immunofluorescence microscopy is exploited to monitor vascular integrity following vascular endothelial cadherin staining. It is shown that microbubbles amplify the ultrasound effect, leading to the formation of interendothelial gaps that cause barrier permeabilization. The total gap area significantly increases with pressure amplitude compared to the control. Gap opening is fully reversible with gap area distribution returning to the control levels 45 min after insonication. The proposed integrated platform allows for precise and repeatable in vitro measurements of cavitation-enhanced endothelium permeability and shows potential for validating irradiation protocols for in vivo applications.
Collapse
Affiliation(s)
- Giulia Silvani
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Chiara Scognamiglio
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Davide Caprini
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Luca Marino
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
| | - Mauro Chinappi
- Department of Industrial Engineering, Università di Roma Tor Vergata, Via del Politecnico 1, 00133, Rome, Italy
| | - Giorgia Sinibaldi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Mohammad F Kiani
- Department of Mechanical Engineering, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Carlo M Casciola
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| |
Collapse
|
107
|
Yemane PT, Åslund AKO, Snipstad S, Bjørkøy A, Grendstad K, Berg S, Mørch Y, Torp SH, Hansen R, Davies CDL. Effect of Ultrasound on the Vasculature and Extravasation of Nanoscale Particles Imaged in Real Time. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:3028-3041. [PMID: 31474384 DOI: 10.1016/j.ultrasmedbio.2019.07.683] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Ultrasound and microbubbles have been found to improve the delivery of drugs and nanoparticles to tumor tissue. To obtain new knowledge on the influence of vascular parameters on extravasation and to elucidate the effect of acoustic pressure on extravasation and penetration of nanoscale particles into the extracellular matrix, real-time intravital multiphoton microscopy was performed during sonication of tumors growing in dorsal window chambers. The impact of vessel diameter, vessel structure and blood flow was characterized. Fluorescein isothiocyanate-dextran (2 MDa) was injected to visualize blood vessels. Mechanical indexes (MI) of 0.2-0.8 and in-house-made, nanoparticle-stabilized microbubbles or Sonovue were applied. The rate and extent of penetration into the extracellular matrix increased with increasing MI. However, to achieve extravasation, smaller vessels required MIs (0.8) higher than those of blood vessels with larger diameters. Ultrasound changed the blood flow rate and direction. Interestingly, the majority of extravasations occurred at vessel branching points.
Collapse
Affiliation(s)
- Petros T Yemane
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andreas K O Åslund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Stroke Unit, Department of Internal Medicine, St. Olav's Hospital, Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristin Grendstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Sverre H Torp
- Department of Pathology, St. Olav's Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Hansen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | | |
Collapse
|
108
|
Bioinspired nanoplatform for enhanced delivery efficiency of doxorubicin into nucleus with fast endocytosis, lysosomal pH-triggered drug release, and reduced efflux. Colloids Surf B Biointerfaces 2019; 183:110413. [DOI: 10.1016/j.colsurfb.2019.110413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 11/20/2022]
|
109
|
Roovers S, Deprez J, Priwitaningrum D, Lajoinie G, Rivron N, Declercq H, De Wever O, Stride E, Le Gac S, Versluis M, Prakash J, De Smedt SC, Lentacker I. Sonoprinting liposomes on tumor spheroids by microbubbles and ultrasound. J Control Release 2019; 316:79-92. [PMID: 31676384 DOI: 10.1016/j.jconrel.2019.10.051] [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: 08/12/2019] [Revised: 10/24/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022]
Abstract
Ultrasound-triggered drug-loaded microbubbles have great potential for drug delivery due to their ability to locally release drugs and simultaneously enhance their delivery into the target tissue. We have recently shown that upon applying ultrasound, nanoparticle-loaded microbubbles can deposit nanoparticles onto cells grown in 2D monolayers, through a process that we termed "sonoprinting". However, the rigid surfaces on which cell monolayers are typically growing might be a source of acoustic reflections and aspherical microbubble oscillations, which can influence microbubble-cell interactions. In the present study, we aim to reveal whether sonoprinting can also occur in more complex and physiologically relevant tissues, by using free-floating 3D tumor spheroids as a tissue model. We show that both monospheroids (consisting of tumor cells alone) and cospheroids (consisting of tumor cells and fibroblasts, which produce an extracellular matrix) can be sonoprinted. Using doxorubicin-liposome-loaded microbubbles, we show that sonoprinting allows to deposit large amounts of doxorubicin-containing liposomes to the outer cell layers of the spheroids, followed by doxorubicin release into the deeper layers of the spheroids, resulting in a significant reduction in cell viability. Sonoprinting may become an attractive approach to deposit drug patches at the surface of tissues, thereby promoting the delivery of drugs into target tissues.
Collapse
Affiliation(s)
- S Roovers
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - J Deprez
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - D Priwitaningrum
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - N Rivron
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, Vienna, Austria
| | - H Declercq
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Tissue Engineering Group, Department of Human Structure and Repair, Ghent University, Belgium
| | - O De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Laboratory Experimental Cancer Research (LECR), Ghent University, Ghent, Belgium
| | - E Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - S Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - M Versluis
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - J Prakash
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - S C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
110
|
Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: Designing a new perspective in nanomedicine delivery. Theranostics 2019; 9:8091-8108. [PMID: 31754383 PMCID: PMC6857058 DOI: 10.7150/thno.37204] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
The enhanced permeability and retention (EPR) effect has underlain the predominant nanomedicine design philosophy for the past three decades. However, growing evidence suggests that it is over-represented in preclinical models, and agents designed solely using its principle of passive accumulation can only be applied to a narrow subset of clinical tumors. For this reason, strategies that can improve upon the EPR effect to facilitate nanomedicine delivery to otherwise non-responsive tumors are required for broad clinical translation. EPR-adaptive nanomedicine delivery comprises a class of chemical and physical techniques that modify tumor accessibility in an effort to increase agent delivery and therapeutic effect. In the present review, we overview the primary benefits and limitations of radiation, ultrasound, hyperthermia, and photodynamic therapy as physical strategies for EPR-adaptive delivery to EPR-insensitive tumor phenotypes, and we reflect upon changes in the preclinical research pathway that should be implemented in order to optimally validate and develop these delivery strategies.
Collapse
Affiliation(s)
- Alexander Dhaliwal
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- MD/PhD Program, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON Canada
| |
Collapse
|
111
|
Prabhakar A, Banerjee R. Nanobubble Liposome Complexes for Diagnostic Imaging and Ultrasound-Triggered Drug Delivery in Cancers: A Theranostic Approach. ACS OMEGA 2019; 4:15567-15580. [PMID: 31572858 PMCID: PMC6761614 DOI: 10.1021/acsomega.9b01924] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/27/2019] [Indexed: 05/19/2023]
Abstract
The ability of ultrasound contrast agents to enhance the cell membrane permeability in response to an ultrasound pulse has unveiled avenues to facilitate the delivery of a higher intracellular payload at target sites. In light of the above, we report the development of submicron-sized (528.7 ± 31.7 nm) nanobubble-paclitaxel liposome (NB-PTXLp) complexes for ultrasound imaging and ultrasound responsive drug delivery in cancer cells. With a paclitaxel entrapment efficiency of 85.4 ± 4.39%, the 200 nm-sized liposomes tethered efficiently (conjugation efficiency ∼98.7 ± 0.14%) with the nanobubbles to form conjugates. Sonoporation of MiaPaCa-2 cells upon treatment with nanobubbles and ultrasound enhanced cellular permeability, resulting in 2.5-fold higher uptake of liposomes in comparison to only liposome treatment. This manifested into more than 300-fold higher anticancer activity of NB-PTXLps in the presence of ultrasound in MiaPaCa-2, Panc-1, MDA-MB-231, and AW-8507 cell lines, compared to commercial formulation ABRAXANE. Also, the NB-PTXLp conjugates were found to exhibit echogenicity comparable to the commercial ultrasound contrast agent SonoVue. In addition, the developed nanobubbles were found to exhibit more than 1 week echogenic stability as opposed to 6 h stability of the commercially available ultrasound contrast agent SonoVue. Hence, the NB-PTXLps developed herein could prove to be a promising and minimally invasive theranostic platform for cancer treatments in the future.
Collapse
|
112
|
Khayamian MA, Shalileh S, Vanaei S, Salemizadeh Parizi M, Ansaryan S, Saghafi M, Abbasvandi F, Ebadi A, Soltan Khamsi P, Abdolahad M. Electrochemical generation of microbubbles by carbon nanotube interdigital electrodes to increase the permeability and material uptakes of cancer cells. Drug Deliv 2019; 26:928-934. [PMID: 31526074 PMCID: PMC6758649 DOI: 10.1080/10717544.2019.1662514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Artificial cavitation as a prerequisite of sonoporation, plays an important role on the ultrasound (US) assisted drug delivery systems. In this study, we have proposed a new method of microbubble (MB) generation by local electrolysis of the medium. An integrated interdigital array of three-electrode system was designed and patterned on a nickel-coated quartz substrate and then, a short DC electrical pulse was applied that consequently resulted in distributed generation of microbubbles at the periphery of the electrodes. Growth of the carbon nanotube (CNT) nanostructures on the surface of the electrodes approximately increased the number of generated microbubbles up to 7-fold and decreased their average size from ∼20 µm for bare to ∼7 µm for CNT electrodes. After optimizing the three-electrode system, biocompatibility assays of the CNT electrodes stimulated by DC electrical micropulses were conducted. Finally, the effect of the proposed method on the sonoporation efficiency and drug uptake of breast cells were assessed using cell cycle and Annexin V/PI flow cytometry analysis. These results show the potential of electrochemical generation of MBs by CNT electrodes as an easy, available and promising technique for artificial cavitation and ultrasound assisted drug delivery.
Collapse
Affiliation(s)
- Mohammad Ali Khayamian
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,School of Mechanical Engineering, College of Engineering, University of Tehran , Tehran , Iran
| | - Shahriar Shalileh
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Shohreh Vanaei
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,School of Biology, College of Science, University of Tehran , Tehran , Iran
| | - Mohammad Salemizadeh Parizi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Saeid Ansaryan
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Mohammad Saghafi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Fereshteh Abbasvandi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR , Tehran , Iran
| | - Amirali Ebadi
- MEMS and NEMS Laboratory, Department of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran , Tehran , Iran
| | - Pouya Soltan Khamsi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Mohammad Abdolahad
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| |
Collapse
|
113
|
Li S, Yin G, Pu X, Huang Z, Liao X, Chen X. A novel tumor-targeted thermosensitive liposomal cerasome used for thermally controlled drug release. Int J Pharm 2019; 570:118660. [PMID: 31491484 DOI: 10.1016/j.ijpharm.2019.118660] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/06/2019] [Accepted: 09/01/2019] [Indexed: 12/22/2022]
Abstract
Drug carriers with tumor targeting and controlled release have strong prospects for application in safe and efficient chemotherapy. Among various carriers, liposomes have good biocompatibility and can enhance the uptake of drugs by cancer cells. However, traditional liposomes have no specific targeting to cancer cells and are prone to insufficient stability, causing early leakage of the drug. Accordingly, organic-inorganic hybrid phospholipid and thermosensitive phospholipid are deliberately introduced into a liposome system to enhance the morphological and structural stability of the liposomes while realizing thermally controlled drug release. Furthermore, modification with a targeting ligand (WSG-peptide) can endow liposomes with active targeting to ovarian carcinoma cells. First, WSG-peptide was grafted onto the hydrophilic terminal of phospholipid molecules, and the organic-inorganic hybrid cerasome-forming lipid (CFL) was synthesized via a two-step chemical reaction. Then, the WSG-grafted thermosensitive liposomal cerasome (c-LIP-WSG) was prepared by thin-film hydration method. The results showed that the c-LIP-WSG had excellent structural stability both in storage and in a simulated circulation environment. In vitro drug release confirmed that the liposomes exhibited thermally controlled release. Cell uptake experiments and living fluorescence imaging of SKOV-3 tumor-bearing nude mice confirmed that the WSG-peptide modified liposomes were provided with specific targeting properties for ovarian carcinoma.
Collapse
Affiliation(s)
- Sixie Li
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Guangfu Yin
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, PR China.
| | - Ximing Pu
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Zhongbin Huang
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Xiaoming Liao
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, PR China
| | - Xianchun Chen
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, PR China
| |
Collapse
|
114
|
Zhang Y, Ou Y, Guo J, Huang X. Ultrasound-triggered breast tumor sonodynamic therapy through hematoporphyrin monomethyl ether-loaded liposome. J Biomed Mater Res B Appl Biomater 2019; 108:948-957. [PMID: 31389180 DOI: 10.1002/jbm.b.34447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/18/2019] [Accepted: 07/09/2019] [Indexed: 01/23/2023]
Abstract
Sonodynamic therapy (SDT) which employs ultrasound-triggered sonosensitizers to generate reactive oxygen species (ROS) has been proved to be effective for treatment of cancers. However, it is still desirable for sonosensitizers to be delivered to tumors as effectively as possible. In this study, we prepared the hematoporphyrin monomethyl ether (HMME)-loaded liposome as the sonosensitizers for SDT and evaluated their effects on human MCF-7 breast cancer cells in vitro and in vivo. Liposomes prepared by thin film hydration technique were about 100 nm in size with positive zeta potential and exhibited spherical in shape. Following irradiation of ultrasound which generates intracellular ROS, the liposome facilitated the delivery of HMME to tumor cells. HMME-loaded liposomes showed low cytotoxicity under basal condition but significant sonodynamic effects under ultrasonic irradiation. Notably, HMME-loaded liposomes exhibited spatial distribution of HMME in tumor tissues of mice. The promoted delivery of HMME into the tumors by liposomes was shown by the greater tumor growth inhibition than free HMME after 20-day treatment. Taken together, these results show that HMME-loaded liposome functions as a promising sonosensitizer for SDT, implying the efficient antitumor effects of HMME-based SDT on breast tumor.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Pharmacy, Danyang People's Hospital, Danyang, China
| | - Yulong Ou
- Department of Pharmacy, Danyang People's Hospital, Danyang, China
| | - Jia Guo
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, China
| | - Xiaojia Huang
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, China
| |
Collapse
|
115
|
The crucial role of macromolecular engineering, drug encapsulation and dilution on the thermoresponsiveness of UCST diblock copolymer nanoparticles used for hyperthermia. Eur J Pharm Biopharm 2019; 142:281-290. [PMID: 31279918 DOI: 10.1016/j.ejpb.2019.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 11/21/2022]
Abstract
Poly(acrylamide-co-acrylonitrile) (P(AAm-co-AN)), an upper critical solution temperature (UCST)-type copolymer in water, was synthesized by reversible addition fragmentation chain transfer (RAFT) copolymerization and used as a macro-RAFT agent for the polymerization of oligo(ethylene glycol) methyl ether methacrylate (OEGMA) to yield amphiphilic diblock P(AAm-co-AN)-b-POEGMA copolymer. A series of copolymers with different AN content was obtained allowing to finely tune the UCST behavior (cloud point (Tt-UCST) from 35 to 78 °C). Addition of the POEGMA block did not modify the Tt-UCST regardless its Mn but provided a lower critical solution temperature behavior at high temperature. Nanoparticles were then formulated by the nanoprecipitation technique revealing that copolymers with higher Tt-UCST yield smaller, better-defined nanoparticles. Eventually, doxorubicin (Dox) was encapsulated into nanoparticles made from the copolymer having a Tt-UCST close to mild hyperthermia (~43 °C). Surprisingly, Dox encapsulation increased Tt-UCST and gave smaller nanoparticles as opposed to their unloaded counterparts. The dilution of the suspension also led to a decrease of Tt-UCST. No obvious hyperthermia effect was observed on the cytotoxicity of Dox-loaded nanoparticles. Our study highlighted the influence of macromolecular engineering, drug encapsulation and nanoparticle dilution on UCST behavior, important features often overlooked despite their crucial impact in the development of thermosensitive nanoscale drug delivery systems.
Collapse
|
116
|
Liu JF, Jang B, Issadore D, Tsourkas A. Use of magnetic fields and nanoparticles to trigger drug release and improve tumor targeting. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1571. [PMID: 31241251 DOI: 10.1002/wnan.1571] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/29/2019] [Accepted: 05/31/2019] [Indexed: 12/21/2022]
Abstract
Drug delivery strategies aim to maximize a drug's therapeutic index by increasing the concentration of drug at target sites while minimizing delivery to off-target tissues. Because biological tissues are minimally responsive to magnetic fields, there has been a great deal of interest in using magnetic nanoparticles in combination with applied magnetic fields to selectively control the accumulation and release of drug in target tissues while minimizing the impact on surrounding tissue. In particular, spatially variant magnetic fields have been used to encourage accumulation of drug-loaded magnetic nanoparticles at target sites, while time-variant magnetic fields have been used to induce drug release from thermally sensitive nanocarriers. In this review, we discuss nanoparticle formulations and approaches that have been developed for magnetic targeting and/or magnetically induced drug release, as well as ongoing challenges in using magnetism for therapeutic applications. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Jessica F Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bian Jang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
117
|
Takemae K, Okamoto J, Horise Y, Masamune K, Muragaki Y. Function of Epirubicin-Conjugated Polymeric Micelles in Sonodynamic Therapy. Front Pharmacol 2019; 10:546. [PMID: 31164824 PMCID: PMC6536629 DOI: 10.3389/fphar.2019.00546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/30/2019] [Indexed: 02/03/2023] Open
Abstract
The combinatory use of high-intensity focused ultrasound (HIFU) and epirubicin (EPI)-conjugated polymeric micellar nanoparticles (NC-6300) is thought to be a less invasive and more efficient method of cancer therapy. To investigate the mechanism underlying the combination effect, we examined the effect of trigger-pulsed HIFU (TP-HIFU) and NC-6300 from the perspective of reactive oxygen species (ROS) generation, which is considered the primary function of sonodynamic therapy (SDT), and changes in drug characteristics. TP-HIFU is an effective sequence for generating hydroxyl radicals to kill cancer cells. EPI was susceptible to degradation by TP-HIFU through the production of hydroxyl radicals. In contrast, EPI degradation of NC-6300 was suppressed by the hydrophilic shell of the micelles. NC-6300 also exhibited a sonosensitizer function, which promoted the generation of superoxide anions by TP-HIFU irradiation. The amount of ROS produced by TP-HIFU reached a level that caused structural changes to the cellular membrane. In conclusion, drug-conjugated micellar nanoparticles are more desirable for SDT because of accelerated ROS production and drug protection from ROS. Furthermore, a combination of NC-6300 and TP-HIFU is useful for minimally invasive cancer therapy with cooperative effects of HIFU-derived features, antitumor activity of EPI, and increased ROS generation to cause damage to cancer cells.
Collapse
Affiliation(s)
- Kazuhisa Takemae
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Pharmaceutical Division, Kowa Company, Ltd., Tokyo, Japan
| | - Jun Okamoto
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yuki Horise
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ken Masamune
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| | - Yoshihiro Muragaki
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
118
|
Fan P, Yang D, Wu J, Yang Y, Guo X, Tu J, Zhang D. Cell-cycle-dependences of membrane permeability and viability observed for HeLa cells undergoing multi-bubble-cell interactions. ULTRASONICS SONOCHEMISTRY 2019; 53:178-186. [PMID: 30642802 DOI: 10.1016/j.ultsonch.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/30/2018] [Accepted: 01/05/2019] [Indexed: 05/07/2023]
Abstract
Microbubble-mediated sonoporation is a promising strategy for intracellular gene/drug delivery, but the biophysical mechanisms involved in the interactions between microbubbles and cells are not well understood. Here, HeLa cells were synchronized in individual cycle phases, then the cell-cycle-dependences of the membrane permeability and viability of HeLa cells undergoing multi-bubble sonoporation were evaluated using focused ultrasound exposure apparatus coupled passive cavitation detection system. The results indicated that: (1) the microbubble cavitation activity should be independent on cell cycle phases; (2) G1-phase cells with the largest Young's modulus were the most robust against microbubble-mediated sonoporation; (3) G2/M-phase cells exhibited the greatest accumulated FITC uptake with the lowest viability, which should be mainly attributed to the chemical effect of synchronization drugs; and (4) more important, S-phase cells with the lowest stiffness seemed to be the most susceptible to the mechanical effect generated by microbubble cavitation activity, which resulted in the greatest enhancement in sonoporation-facilitated membrane permeabilization without further scarifying their viability. The current findings may benefit ongoing efforts aiming to pursue rational utilization of microbubble-mediated sonoporation in cell-cycle-targeted gene/drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Pengfei Fan
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Dongxin Yang
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Jun Wu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yanye Yang
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China.
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), School of Physics, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China.
| |
Collapse
|
119
|
Zykova Y, Kudryavtseva V, Gai M, Kozelskaya A, Frueh J, Sukhorukov G, Tverdokhlebov S. Free-standing microchamber arrays as a biodegradable drug depot system for implant coatings. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.02.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
120
|
Almekkawy M, Chen J, Ellis MD, Haemmerich D, Holmes DR, Linte CA, Panescu D, Pearce J, Prakash P, Zderic V. Therapeutic Systems and Technologies: State-of-the-Art Applications, Opportunities, and Challenges. IEEE Rev Biomed Eng 2019; 13:325-339. [PMID: 30951478 PMCID: PMC7341980 DOI: 10.1109/rbme.2019.2908940] [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] [Indexed: 11/08/2022]
Abstract
In this review, we present current state-of-the-art developments and challenges in the areas of thermal therapy, ultrasound tomography, image-guided therapies, ocular drug delivery, and robotic devices in neurorehabilitation. Additionally, intellectual property and regulatory aspects pertaining to therapeutic systems and technologies are addressed.
Collapse
|
121
|
Mannaris C, Bau L, Grundy M, Gray M, Lea-Banks H, Seth A, Teo B, Carlisle R, Stride E, Coussios CC. Microbubbles, Nanodroplets and Gas-Stabilizing Solid Particles for Ultrasound-Mediated Extravasation of Unencapsulated Drugs: An Exposure Parameter Optimization Study. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:954-967. [PMID: 30655109 DOI: 10.1016/j.ultrasmedbio.2018.10.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 06/09/2023]
Abstract
Ultrasound-induced cavitation has been proposed as a strategy to tackle the challenge of inadequate extravasation, penetration and distribution of therapeutics into tumours. Here, the ability of microbubbles, droplets and solid gas-trapping particles to facilitate mass transport and extravasation of a model therapeutic agent following ultrasound-induced cavitation is investigated. Significant extravasation and penetration depths on the order of millimetres are achieved with all three agents, including the range of pressures and frequencies achievable with existing clinical ultrasound systems. Deeper but highly directional extravasation was achieved with frequencies of 1.6 and 3.3 MHz compared with 0.5 MHz. Increased extravasation was observed with increasing pulse length and exposure time, while an inverse relationship is observed with pulse repetition frequency. No significant cell death or any haemolytic activity in human blood was observed at clinically relevant concentrations for any of the agents. Overall, solid gas-trapping nanoparticles were found to enable the most extensive extravasation for the lowest input acoustic energy, followed by microbubbles and then droplets. The ability of these agents to produce sustained inertial cavitation activity whilst being small enough to follow the drug out of the circulation and into diseased tissue, combined with a good safety profile and the possibility of real-time monitoring, offers considerable potential for enhanced drug delivery of unmodified drugs in oncological and other biomedical applications.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Megan Grundy
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Harriet Lea-Banks
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Anjali Seth
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Boon Teo
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Constantin C Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom.
| |
Collapse
|
122
|
Li X, Wang Z, Xia H. Ultrasound Reversible Response Nanocarrier Based on Sodium Alginate Modified Mesoporous Silica Nanoparticles. Front Chem 2019; 7:59. [PMID: 30805332 PMCID: PMC6378627 DOI: 10.3389/fchem.2019.00059] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Mesoporous silica nanoparticles (MSN) covered by polymer coatings, cross-linked by weak coordination bonds were expected to present a reversible responsiveness under on-off ultrasound stimuli. Herein, we prepared a sodium alginate (SA) modified MSN with carboxyl-calcium (COO--Ca2+) coordination bonds in the modified layer, which could block the mesopores of MSN and effectively prevent the cargo from pre-releasing before stimulation. The coordination bonds would be destroyed under the stimulation of low intensity ultrasound (20 kHz) or high intensity focused ultrasound (HIFU, 1.1 MHz), leading to a rapid and significant cargo release, and then they could be reformed when ultrasound was turned off, resulting in an instant cargo release stopping. The reversible cleavage and reformation of this coordination bonds under on-off ultrasound stimulus were confirmed by the gel-sol transition behaviors of the SA-CaCl2 gels. An excellent real-time control of rhodamine B (RhB) release performance was obtained under the ultrasound stimuli. Obviously, the cargo release ratio could reach to nearly 40% when HIFU (80 W) was turned on for 5 min, and remained basically constant when ultrasound was turned off, which would finally reach to nearly 100% within 30 min under this on-off pulsatile status. These hybrid MSN based nanoparticles with excellent reversible ultrasound on-off responsiveness were of great interest in on-demand drug delivery applications in the future.
Collapse
Affiliation(s)
| | - Zhanhua Wang
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute, Sichuan University, Chengdu, China
| | | |
Collapse
|
123
|
Ishihara H, Sakai-Kato K. [Characterization and Analytical Techniques for Nano-DDS Formulations]. YAKUGAKU ZASSHI 2019; 139:235-236. [PMID: 30713233 DOI: 10.1248/yakushi.18-00171-f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
124
|
Liu J, Chen Y, Wang G, Jin Q, Sun Z, Lv Q, Wang J, Yang Y, Zhang L, Xie M. Improving acute cardiac transplantation rejection therapy using ultrasound-targeted FK506-loaded microbubbles in rats. Biomater Sci 2019; 7:3729-3740. [DOI: 10.1039/c9bm00301k] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
FK506-MBs combined with the UTMD technique increased drug concentrations in transplanted hearts and enhanced the therapeutic effect.
Collapse
|
125
|
Bordat A, Boissenot T, Nicolas J, Tsapis N. Thermoresponsive polymer nanocarriers for biomedical applications. Adv Drug Deliv Rev 2019; 138:167-192. [PMID: 30315832 DOI: 10.1016/j.addr.2018.10.005] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/12/2018] [Accepted: 10/08/2018] [Indexed: 12/21/2022]
Abstract
Polymer nanocarriers allow drug encapsulation leading to fragile molecule protection from early degradation/metabolization, increased solubility of poorly soluble drugs and improved plasmatic half-life. However, efficiently controlling the drug release from nanocarriers is still challenging. Thermoresponsive polymers exhibiting either a lower critical solution temperature (LCST) or an upper critical solution temperature (UCST) in aqueous medium may be the key to build spatially and temporally controlled drug delivery systems. In this review, we provide an overview of LCST and UCST polymers used as building blocks for thermoresponsive nanocarriers for biomedical applications. Recent nanocarriers based on thermoresponsive polymer exhibiting unprecedented features useful for biomedical applications are also discussed. While LCST nanocarriers have been studied for over two decades, UCST nanocarriers have recently emerged and already show great potential for effective thermoresponsive drug release.
Collapse
Affiliation(s)
- Alexandre Bordat
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Tanguy Boissenot
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Julien Nicolas
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Nicolas Tsapis
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92290 Châtenay-Malabry, France.
| |
Collapse
|
126
|
Cui X, Cheng W, Dong M, Han X. A multifunctional biomimetic hybrid nanocarrier for the controlled delivery of chemotherapy drugs by near-infrared light. NEW J CHEM 2019. [DOI: 10.1039/c8nj05879b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have constructed a biocompatible magnetic nanoparticle-based nanocomposite material by integrating the features of Fe3O4 nanoparticles, mesoporous silica and lipid bilayer for the treatment of tumor.
Collapse
Affiliation(s)
- Xinyu Cui
- State Key Laboratory of Urban Water Resource and Environment
- School of Chemical Engineering and Technology
- Harbin Institute of Technology
- Harbin
- China
| | - Wenlong Cheng
- Department of Chemical Engineering
- Monash University
- Victoria
- Australia
| | - Mingdong Dong
- Interdisciplinary Nanoscience Center (iNANO)
- Aarhus University
- 8000 Aarhus
- Denmark
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment
- School of Chemical Engineering and Technology
- Harbin Institute of Technology
- Harbin
- China
| |
Collapse
|
127
|
Snipstad S, Sulheim E, de Lange Davies C, Moonen C, Storm G, Kiessling F, Schmid R, Lammers T. Sonopermeation to improve drug delivery to tumors: from fundamental understanding to clinical translation. Expert Opin Drug Deliv 2018; 15:1249-1261. [PMID: 30415585 DOI: 10.1080/17425247.2018.1547279] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Ultrasound in combination with microbubbles can make cells and tissues more accessible for drugs, thereby achieving improved therapeutic outcomes. In this review, we introduce the term 'sonopermeation', covering mechanisms such as pore formation (traditional sonoporation), as well as the opening of intercellular junctions, stimulated endocytosis/transcytosis, improved blood vessel perfusion and changes in the (tumor) microenvironment. Sonopermeation has gained a lot of interest in recent years, especially for delivering drugs through the otherwise impermeable blood-brain barrier, but also to tumors. AREAS COVERED In this review, we summarize various in vitro assays and in vivo setups that have been employed to unravel the fundamental mechanisms involved in ultrasound-enhanced drug delivery, as well as clinical trials that are ongoing in patients with brain, pancreatic, liver and breast cancer. We summarize the basic principles of sonopermeation, describe recent findings obtained in (pre-) clinical trials, and discuss future directions. EXPERT OPINION We suggest that an improved mechanistic understanding, and microbubbles and ultrasound equipment specialized for drug delivery (and not for imaging) are key aspects to create more effective treatment regimens by sonopermeation. Real-time feedback and tools to predict therapeutic outcome and which tumors/patients will benefit from sonopermeation-based interventions will be important to promote clinical translation.
Collapse
Affiliation(s)
- Sofie Snipstad
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Einar Sulheim
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Catharina de Lange Davies
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway
| | - Chrit Moonen
- d Imaging Division , University Medical Center , Utrecht , The Netherlands
| | - Gert Storm
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands
| | - Fabian Kiessling
- g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| | - Ruth Schmid
- b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway
| | - Twan Lammers
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands.,g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
128
|
Goutal S, Gerstenmayer M, Auvity S, Caillé F, Mériaux S, Buvat I, Larrat B, Tournier N. Physical blood-brain barrier disruption induced by focused ultrasound does not overcome the transporter-mediated efflux of erlotinib. J Control Release 2018; 292:210-220. [PMID: 30415015 DOI: 10.1016/j.jconrel.2018.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022]
Abstract
Overcoming the efflux mediated by ATP-binding cassette (ABC) transporters at the blood-brain barrier (BBB) remains a challenge for the delivery of small molecule tyrosine kinase inhibitors (TKIs) such as erlotinib to the brain. Inhibition of ABCB1 and ABCG2 at the mouse BBB improved the BBB permeation of erlotinib but could not be achieved in humans. BBB disruption induced by focused ultrasound (FUS) was investigated as a strategy to overcome the efflux transport of erlotinib in vivo. In rats, FUS combined with microbubbles allowed for a large and spatially controlled disruption of the BBB in the left hemisphere. ABCB1/ABCG2 inhibition was performed using elacridar (10 mg/kg i.v). The brain kinetics of erlotinib was studied using 11C-erlotinib Positron Emission Tomography (PET) imaging in 5 groups (n = 4-5 rats per group) including a baseline group, immediately after sonication (FUS), 48 h after FUS (FUS + 48 h), elacridar (ELA) and their combination (FUS + ELA). BBB integrity was assessed using the Evan's Blue (EB) extravasation test. Brain exposure to 11C-erlotinib was measured as the area under the curve (AUC) of the brain kinetics (% injected dose (%ID) versus time (min)) in volumes corresponding to the disrupted (left) and the intact (right) hemispheres, respectively. EB extravasation highlighted BBB disruption in the left hemisphere of animals of the FUS and FUS + ELA groups but not in the control and ELA groups. EB extravasation was not observed 48 h after FUS suggesting recovery of BBB integrity. Compared with the control group (AUCBaseline = 1.4 ± 0.5%ID.min), physical BBB disruption did not impact the brain kinetics of 11C-erlotinib in the left hemisphere (p > .05) either immediately (AUCFUS = 1.2 ± 0.1%ID.min) or 48 h after FUS (AUCFUS+48h = 1.1 ± 0.3%ID.min). Elacridar similarly increased 11C-erlotinib brain exposure to the left hemisphere in the absence (AUCELA = 2.2 ± 0.5%ID.min, p < .001) and in the presence of BBB disruption (AUCFUS+ELA = 2.1 ± 0.5%ID.min, p < .001). AUCleft was never significantly different from AUCright (p > .05), in any of the tested conditions. BBB integrity is not the rate limiting step for erlotinib delivery to the brain which is mainly governed by ABC-mediated efflux. Efflux transport of erlotinib persisted despite BBB disruption.
Collapse
Affiliation(s)
- Sébastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France; Molecular Imaging Research Center, MIRCen, Institut de Biologie François Jacob, Direction de la Recherche Fondamentale, CEA, Fontenay-Aux-Roses, France
| | - Matthieu Gerstenmayer
- Neurospin, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Université Paris Saclay, Gif sur Yvette, France
| | - Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Sébastien Mériaux
- Neurospin, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Université Paris Saclay, Gif sur Yvette, France
| | - Irène Buvat
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Benoit Larrat
- Neurospin, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Université Paris Saclay, Gif sur Yvette, France
| | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France.
| |
Collapse
|
129
|
|
130
|
Zhou L, Qiu T, Lv F, Liu L, Ying J, Wang S. Self-Assembled Nanomedicines for Anticancer and Antibacterial Applications. Adv Healthc Mater 2018; 7:e1800670. [PMID: 30080319 DOI: 10.1002/adhm.201800670] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/03/2018] [Indexed: 01/28/2023]
Abstract
Self-assembly strategies have been widely applied in the nanomedicine field, which provide a convenient approach for building various structures for delivery carriers. When cooperating with biomolecules, self-assembly systems have significant influence on the cell activity and life process and could be used for regulating nanodrug activity. In this review, self-assembled nanomedicines are introduced, including materials, encapsulation, and releasing strategies, where self-assembly strategies are involved. Furthermore, as a promising and emerging area for nanomedicine, in situ self-assembly of anticancer drugs and supramolecular antibiotic switches is also discussed about how to regulate drug activity. Selective pericellular assembly can block mass transformation of cancer cells inducing cell apoptosis, and the intracellular assembly can either cause cell death or effectively avoid drug elimination from cytosol of cancer cells because of the assembly-induced retention (AIR) effect. Host-guest interactions of drug and competitive molecules offer reversible regulations of antibiotic activity, which can reduce drug-resistance and inhibit the generation of drug-resistant bacteria. Finally, the challenges and development trend in the field are discussed.
Collapse
Affiliation(s)
- Lingyun Zhou
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- College of Chemistry; University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Tian Qiu
- Department of Pathology; National Cancer Center/National Clinical Research Center for; Cancer/Cancer Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing 100021 P. R. China
| | - Fengting Lv
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Libing Liu
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Jianming Ying
- Department of Pathology; National Cancer Center/National Clinical Research Center for; Cancer/Cancer Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing 100021 P. R. China
| | - Shu Wang
- Beijing National Laboratory for Molecular Sciences; Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
- College of Chemistry; University of Chinese Academy of Sciences; Beijing 100049 P. R. China
| |
Collapse
|
131
|
Nan N, Si D, Hu G. Nanoscale cavitation in perforation of cellular membrane by shock-wave induced nanobubble collapse. J Chem Phys 2018; 149:074902. [DOI: 10.1063/1.5037643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Nan Nan
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai Key Laboratory of Mechanics in Energy Engineering, Shanghai University, Shanghai 200072, China
| | - Dongqing Si
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai Key Laboratory of Mechanics in Energy Engineering, Shanghai University, Shanghai 200072, China
| | - Guohui Hu
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai Key Laboratory of Mechanics in Energy Engineering, Shanghai University, Shanghai 200072, China
| |
Collapse
|
132
|
Houvenagel S, Moine L, Picheth G, Dejean C, Brûlet A, Chennevière A, Faugeras V, Huang N, Couture O, Tsapis N. Comb-Like Fluorophilic-Lipophilic-Hydrophilic Polymers for Nanocapsules as Ultrasound Contrast Agents. Biomacromolecules 2018; 19:3244-3256. [DOI: 10.1021/acs.biomac.8b00506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Sophie Houvenagel
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Laurence Moine
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Guilherme Picheth
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Camille Dejean
- BioCIS, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Annie Brûlet
- Laboratoire Léon Brillouin, UMR12 CEA-CNRS, CEA Saclay, Gif sur Yvette, F-91191, France
| | - Alexis Chennevière
- Laboratoire Léon Brillouin, UMR12 CEA-CNRS, CEA Saclay, Gif sur Yvette, F-91191, France
| | - Vincent Faugeras
- Institut Langevin, ESPCI Paris, CNRS (UMR 7587), INSERM (U979), Paris 75238 CEDEX 05, France
| | - Nicolas Huang
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Olivier Couture
- Institut Langevin, ESPCI Paris, CNRS (UMR 7587), INSERM (U979), Paris 75238 CEDEX 05, France
| | - Nicolas Tsapis
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| |
Collapse
|
133
|
Elhelf IS, Albahar H, Shah U, Oto A, Cressman E, Almekkawy M. High intensity focused ultrasound: The fundamentals, clinical applications and research trends. Diagn Interv Imaging 2018; 99:349-359. [DOI: 10.1016/j.diii.2018.03.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 02/22/2018] [Accepted: 03/06/2018] [Indexed: 02/06/2023]
|
134
|
Mannaris C, Teo BM, Seth A, Bau L, Coussios C, Stride E. Gas-Stabilizing Gold Nanocones for Acoustically Mediated Drug Delivery. Adv Healthc Mater 2018; 7:e1800184. [PMID: 29696808 DOI: 10.1002/adhm.201800184] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/15/2018] [Indexed: 01/27/2023]
Abstract
The efficient penetration of drugs into tumors is a major challenge that remains unmet. Reported herein is a strategy to promote extravasation and enhanced penetration using inertial cavitation initiated by focused ultrasound and cone-shaped gold nanoparticles that entrap gas nanobubbles. The cones are capable of initiating inertial cavitation under pressures and frequencies achievable with existing clinical ultrasound systems and of promoting extravasation and delivery of a model large therapeutic molecule in an in vitro tissue mimicking flow phantom, achieving penetration depths in excess of 2 mm. Ease of functionalization and intrinsic imaging capabilities provide gold with significant advantages as a material for biomedical applications. The cones show neither cytotoxicity in Michigan Cancer Foundation (MCF)-7 cells nor hemolytic activity in human blood at clinically relevant concentrations and are found to be colloidally stable for at least 5 d at 37 °C and several months at 4 °C.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Boon M Teo
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
- Interdisciplinary Nanoscience Center (iNANO), The iNANO House, Aarhus University, Gustav Wieds Vej 14, DK-8000, Aarhus C, Denmark
- School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Anjali Seth
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Luca Bau
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
135
|
Garello F, Terreno E. Sonosensitive MRI Nanosystems as Cancer Theranostics: A Recent Update. Front Chem 2018; 6:157. [PMID: 29868560 PMCID: PMC5949352 DOI: 10.3389/fchem.2018.00157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/19/2018] [Indexed: 11/13/2022] Open
Abstract
In the tireless search for innovative and more efficient cancer therapies, sonosensitive Magnetic Resonance Imaging (MRI) agents play an important role. Basically, these systems consist of nano/microvesicles composed by a biocompatible membrane, responsive to ultrasound-induced thermal or mechanical effects, and an aqueous core, filled up with a MRI detectable probe and a therapeutic agent. They offer the possibility to trigger and monitor in real time drug release in a spatio-temporal domain, with the expectation to predict the therapeutic outcome. In this review, the key items to design sonosensitive MRI agents will be examined and an overview on the different approaches available so far will be given. Due to the extremely wide range of adopted ultrasound settings and formulations conceived, it is hard to compare the numerous preclinical studies reported. However, in general, a significantly better therapeutic outcome was noticed when exploiting ultrasound triggered drug release in comparison to traditional therapies, thus paving the way to the possible clinical translation of optimized sonosensitive MRI agents.
Collapse
Affiliation(s)
- Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Enzo Terreno
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
136
|
Ding S, O'Banion CP, Welfare JG, Lawrence DS. Cellular Cyborgs: On the Precipice of a Drug Delivery Revolution. Cell Chem Biol 2018; 25:648-658. [PMID: 29628434 DOI: 10.1016/j.chembiol.2018.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/17/2017] [Accepted: 02/28/2018] [Indexed: 12/12/2022]
Abstract
Cell-based drug delivery systems offer the prospect of biocompatibility, large-loading capacity, long in vivo lifespan, and active targeting of diseased sites. However, these opportunities are offset by an array of challenges, including safeguarding the integrity of the drug cargo and the cellular host, as well as ensuring that drug release occurs at the appropriate time and place. Emerging strategies that address these, and related, issues, are described herein.
Collapse
Affiliation(s)
- Song Ding
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Colin P O'Banion
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joshua G Welfare
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David S Lawrence
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
137
|
Peruzzi G, Sinibaldi G, Silvani G, Ruocco G, Casciola CM. Perspectives on cavitation enhanced endothelial layer permeability. Colloids Surf B Biointerfaces 2018; 168:83-93. [PMID: 29486912 DOI: 10.1016/j.colsurfb.2018.02.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 12/20/2022]
Abstract
Traditional drug delivery systems, where pharmaceutical agents are conveyed to the target tissue through the blood circulation, suffer of poor therapeutic efficiency and limited selectivity largely due to the low permeability of the highly specialised biological interface represented by the endothelial layer. Examples concern cancer therapeutics or degenerative disorders where drug delivery is inhibited by the blood-brain barrier (BBB). Microbubbles injected into the bloodstream undergo volume oscillations under localised ultrasound irradiation and possibly collapse near the site of interest, with no effect on the rest of the endothelium. The resulting mechanical action induces a transient increase of the inter-cellular spaces and facilitates drug extravasation. This approach, already pursed in in vivo animal models, is extremely expensive and time-consuming. On the other hand in vitro studies using different kinds of microfluidic networks are firmly established in the pharmaceutical industry for drug delivery testing. The combination of the in vitro approach with ultrasound used to control microbubbles oscillations is expected to provide crucial information for developing cavitation enhanced drug delivery protocols and for screening the properties of the biological interface in presence of healthy or diseased tissues. Purpose of the present review is providing the state of the art in this rapidly growing field where cavitation is exploited as a viable technology to transiently modify the permeability of the biological interface. After describing current in vivo studies, particular emphasis will be placed on illustrating characteristics of micro-devices, biological functionalisation, properties of the artificial endothelium and ultrasound irradiation techniques.
Collapse
Affiliation(s)
- Giovanna Peruzzi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giorgia Sinibaldi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Italy
| | - Giulia Silvani
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy; Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy; Department of Physics, Sapienza University of Rome, Italy.
| | - Carlo Massimo Casciola
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy; Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Italy
| |
Collapse
|
138
|
Bosca F, Bielecki PA, Exner AA, Barge A. Porphyrin-Loaded Pluronic Nanobubbles: A New US-Activated Agent for Future Theranostic Applications. Bioconjug Chem 2018; 29:234-240. [PMID: 29365258 DOI: 10.1021/acs.bioconjchem.7b00732] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sonodynamic therapy (SDT) has become a promising noninvasive approach for cancer therapy. The treatment exploits the ability of particular molecules (i.e., porphyrins) to be excited by ultrasound and produce reactive oxygen species (ROS) during their decay process. These reactive species, in turn, result in cell death. To capitalize on the real-time visualization and on-demand delivery of ultrasound contrast agents, this study aims to combine porphyrins with nanobubbles (NBs) to obtain an ultrasound-activated theranostic agent that exploits the SDT activity in vitro. Two porphyrin classes, exposing different hydrophobic side chains, were synthesized. NB size and encapsulation efficiency were markedly dependent on the porphyrin structure. The combination of these porphyrin and NBs resulted in a significant reduction in cell viability upon sonication in pilot studies performed on the LS 174T colorectal cancer cell line.
Collapse
Affiliation(s)
- Federica Bosca
- Department of Drug Science and Technology, University of Turin , Via Giuria 9, 10125 Turin, Italy
| | | | | | - Alessandro Barge
- Department of Drug Science and Technology, University of Turin , Via Giuria 9, 10125 Turin, Italy
| |
Collapse
|
139
|
Yu F, Wu H, Tang Y, Xu Y, Qian X, Zhu W. Temperature-sensitive copolymer-coated fluorescent mesoporous silica nanoparticles as a reactive oxygen species activated drug delivery system. Int J Pharm 2018; 536:11-20. [DOI: 10.1016/j.ijpharm.2017.11.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 10/25/2017] [Accepted: 11/10/2017] [Indexed: 10/18/2022]
|
140
|
Zhou L, Wang H, Li Y. Stimuli-Responsive Nanomedicines for Overcoming Cancer Multidrug Resistance. Theranostics 2018; 8:1059-1074. [PMID: 29463999 PMCID: PMC5817110 DOI: 10.7150/thno.22679] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy is still a main option for cancer therapy, but its efficacy is often unsatisfying due to multidrug resistance (MDR). The tumor microenvironment is considered a dominant factor causing MDR. Stimuli-responsive nanomedicines exhibit many superiorities for reversal of MDR. As smart systems, stimuli-responsive nanomedicines are desirable for achieving site-specific accumulation and triggered drug release in response to slight changes in physicochemical properties in pathological conditions or to exogenous stimuli. In this review, we highlight the current progress of various nanomedicines with different stimuli-responsive capabilities for overcoming MDR. The materials, design, construction as well as efficacy in overcoming MDR of these nanomedicines are discussed. Eventually, we look forward to forthcoming intelligent nanoparticle systems with new mechanisms to deliver drugs for practical applications in conquering cancer MDR.
Collapse
Affiliation(s)
- Lei Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Hao Wang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
141
|
Abstract
Integration of nanotechnology and biomedicine has offered great opportunities for the development of nanoscaled therapeutic platforms. Amongst various nanocarriers, mesoporous silica nanoparticles (MSNs) is one of the most developed and promising inorganic materials-based drug delivery system for clinical translations due to their simple composition and nanoporous structure. MSNs possess unique structural features, for example, well-defined morphology, large surface areas, uniform size, controllable structure, flexible pore volume, tunable pore sizes, extraordinarily high loading efficiency, and excellent biocompatibility. Progress in structure control and functionalization may endow MSNs with functionalities that enable medical applications of these integrated nanoparticles such as molecularly targeted drug delivery, multicomponent synergistic therapy, in vivo imaging and therapeutic capability, on-demand/stimuli-responsive drug release, etc. In this chapter, the authors overview MSNs' characteristics and the scientific efforts developed till date involving drug delivery and biomedical applications.
Collapse
|
142
|
Ni R, Liu J, Chau Y. Ultrasound-facilitated assembly and disassembly of a pH-sensitive self-assembly peptide. RSC Adv 2018; 8:29482-29487. [PMID: 35548023 PMCID: PMC9084454 DOI: 10.1039/c8ra04391d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/12/2018] [Indexed: 12/23/2022] Open
Abstract
In this report, we investigated the impact of external stimulus (ultrasound) and internal stimulus (pH) on peptide assembly and disassembly. Two short rationally designed peptides K3C6SPD and F20H differing in the presence of a single pH-sensitive histidine residue were studied as the model peptides. The assembly kinetics studies demonstrated that the substitution of phenylalanine in K3C6SPD with histidine (F20H) significantly slowed down the peptide assembly rate at all three tested pHs (pH 9.5, pH 7.4 and pH 5.0). At the same time, this F to H substitution led to the increased pH-responsive assembly kinetics. By treating the peptide sample at the beginning of the assembly process at pH 9.5 for 5 min with the ultrasound power of 2.1 W cm−2, the assembly rate of peptide F20H was significantly accelerated with the lag phase being shortened from 10 days to 2 days. For the disassembly of F20H peptide nanofibrils preformed at pH 9.5, upon pH adjustment from pH 9.5 to pH 5.0, 5 min ultrasonication of the nanofibrils resulted in the nanofibril disassembly within 6 hours, instead of 5 days in the absence of ultrasound. On the contrary, similar ultrasound treatment of the peptide K3C6SPD did not produce any obvious effect on both assembly and disassembly processes. This study offers an effective strategy to modulate the stimuli-responsiveness of the peptide-based biomaterials. We present an efficient strategy to enhance the stimuli-responsiveness of peptide-based biomaterials by combination of different stimuli.![]()
Collapse
Affiliation(s)
- Rong Ni
- Department of Chemical and Biological Engineering
- The Hong Kong University of Science and Technology
- Kowloon
- China
- Institute for Advanced Study
| | - Jianhui Liu
- Department of Chemical and Biological Engineering
- The Hong Kong University of Science and Technology
- Kowloon
- China
| | - Ying Chau
- Department of Chemical and Biological Engineering
- The Hong Kong University of Science and Technology
- Kowloon
- China
| |
Collapse
|
143
|
Shen N, Lei B, Wang Y, Xu S, Liu H. Redox/ultrasound dual stimuli-responsive nanogel for precisely controllable drug release. NEW J CHEM 2018. [DOI: 10.1039/c8nj00392k] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Schematic representation of the preparation and the strategy of redox/ultrasound triggered drug release of the nanogel system.
Collapse
Affiliation(s)
- Nengwei Shen
- Key Laboratory for Advanced Materials
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Bin Lei
- Key Laboratory for Advanced Materials
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Yizhou Wang
- Key Laboratory for Advanced Materials
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Shouhong Xu
- Key Laboratory for Advanced Materials
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| | - Honglai Liu
- Key Laboratory for Advanced Materials
- School of Chemistry and Molecular Engineering
- East China University of Science and Technology
- Shanghai
- P. R. China
| |
Collapse
|
144
|
Thermo-responsive mesoporous silica/lipid bilayer hybrid nanoparticles for doxorubicin on-demand delivery and reduced premature release. Colloids Surf B Biointerfaces 2017; 160:527-534. [DOI: 10.1016/j.colsurfb.2017.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/20/2017] [Accepted: 10/02/2017] [Indexed: 11/19/2022]
|
145
|
Manaspon C, Hernandez C, Nittayacharn P, Jeganathan S, Nasongkla N, Exner AA. Increasing Distribution of Drugs Released from In Situ Forming PLGA Implants Using Therapeutic Ultrasound. Ann Biomed Eng 2017; 45:2879-2887. [PMID: 28929267 PMCID: PMC5693652 DOI: 10.1007/s10439-017-1926-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022]
Abstract
One of the challenges in developing sustained-release local drug delivery systems is the limited treatment volume that can be achieved. In this work, we examine the effectiveness of using low frequency, high intensity ultrasound to promote the spatial penetration of drug molecules away from the implant/injection site boundary upon release from injectable, phase inverting poly(lactic acid-co-glycolic acid) (PLGA) implants. Fluorescein-loaded PLGA solutions were injected into poly(acrylamide) phantoms, and the constructs were treated daily for 14 days with ultrasound at 2.2 W/cm2 for 10 min. The 2D distribution of fluorescein within the phantoms was quantified using fluorescence imaging. Implants receiving ultrasound irradiation showed a 1.7-5.6 fold increase (p < 0.05) in fluorescence intensity and penetration distance, with the maximum increase observed 5 days post-implantation. However, this evidence was not seen when the same experiment was also carried out in phosphate buffer saline (pH 7.4). Results suggest an active role of ultrasound in local molecular transport in the phantom. An increase of fluorescein release and penetration depth in phantoms can be accomplished through brief application of ultrasound. This simple technique offers an opportunity to eventually enhance the therapeutic efficacy and broaden the application of local drug delivery systems.
Collapse
Affiliation(s)
- Chawan Manaspon
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakorn Pathom, 73170, Thailand
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Christopher Hernandez
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Pinunta Nittayacharn
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Selva Jeganathan
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Norased Nasongkla
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakorn Pathom, 73170, Thailand
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
- Department of Biomedical Engineering, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
| |
Collapse
|
146
|
Fan P, Zhang Y, Guo X, Cai C, Wang M, Yang D, Li Y, Tu J, Crum LA, Wu J, Zhang D. Cell-cycle-specific Cellular Responses to Sonoporation. Am J Cancer Res 2017; 7:4894-4908. [PMID: 29187912 PMCID: PMC5706108 DOI: 10.7150/thno.20820] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022] Open
Abstract
Microbubble-mediated sonoporation has shown its great potential in facilitating intracellular uptake of gene/drugs and other therapeutic agents that are otherwise difficult to enter cells. However, the biophysical mechanisms underlying microbubble-cell interactions remain unclear. Particularly, it is still a major challenge to get a comprehensive understanding of the impact of cell cycle phase on the cellular responses simultaneously occurring in cell membrane and cytoskeleton induced by microbubble sonoporation. Methods: Here, efficient synchronizations were performed to arrest human cervical epithelial carcinoma (HeLa) cells in individual cycle phases. The, topography and stiffness of synchronized cells were examined using atomic force microscopy. The variations in cell membrane permeabilization and cytoskeleton arrangement induced by sonoporation were analyzed simultaneously by a real-time fluorescence imaging system. Results: The results showed that G1-phase cells typically had the largest height and elastic modulus, while S-phase cells were generally the flattest and softest ones. Consequently, the S-Phase was found to be the preferred cycle for instantaneous sonoporation treatment, due to the greatest enhancement of membrane permeability and the fastest cytoskeleton disassembly at the early stage after sonoporation. Conclusion: The current findings may benefit ongoing efforts aiming to pursue rational utilization of microbubble-mediated sonoporation in cell cycle-targeted gene/drug delivery for cancer therapy.
Collapse
|
147
|
Franco MS, Oliveira MC. Ratiometric drug delivery using non-liposomal nanocarriers as an approach to increase efficacy and safety of combination chemotherapy. Biomed Pharmacother 2017; 96:584-595. [PMID: 29035823 DOI: 10.1016/j.biopha.2017.10.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/27/2017] [Accepted: 10/02/2017] [Indexed: 10/18/2022] Open
Abstract
The observation that different drug ratios of the same drug combination can lead to synergistic or antagonistic effects when tested against the same cancer cell line in vitro gave rise to a new trend, the ratiometric delivery. This strategy consists of co-encapsulating a specific synergistic ratio of a drug combination into a nanocarrier so that synergism observed in vitro will be faithfully translated to in vivo, optimizing combination therapy. In this review we focus on how to quantify synergism in vitro, followed by how this affected the evolution of nanocarriers culminating in the ratiometric delivery, and finally we summarize the results of the non-liposomal formulations that were built upon this concept.
Collapse
Affiliation(s)
- Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
148
|
Luo Z, Jin K, Pang Q, Shen S, Yan Z, Jiang T, Zhu X, Yu L, Pang Z, Jiang X. On-Demand Drug Release from Dual-Targeting Small Nanoparticles Triggered by High-Intensity Focused Ultrasound Enhanced Glioblastoma-Targeting Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:31612-31625. [PMID: 28861994 DOI: 10.1021/acsami.7b10866] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Glioblastoma is one of the most challenging and intractable tumors with the difficult treatment and poor prognosis. Unsatisfactory traditional systemic chemotherapies for glioblastoma are mainly attributed to the insufficient and nonspecific drug delivery into the brain tumors as well as the incomplete drug release at the tumor sites. Inspired by the facts that angiopep-2 peptide is an acknowledged dual-targeting moiety for brain tumor-targeting delivery and high-intensity focused ultrasound (HIFU) is an ideal trigger for drug release with an ultrahigh energy and millimeter-sized focus ability, in the present study, a novel HIFU-responsive angiopep-2-modified small poly(lactic-co-glycolic acid) (PLGA) hybrid nanoparticle (NP) drug delivery system holding doxorubicin/perfluorooctyl bromide (ANP-D/P) was designed to increase the intratumoral drug accumulation, further trigger on-demand drug release at the glioblastoma sites, and enhance glioblastoma therapy. It was shown that the ANP-D/P was stable and had a small size of 41 nm. The angiopep-2 modification endowed the ANP-D/P with improved blood-brain barrier transportation and specific accumulation in glioblastoma tissues by 17 folds and 13.4 folds compared with unmodified NPs, respectively. Under HIFU irradiation, the ANP-D/P could release 47% of the drug within 2 min and induce the apoptosis of most tumor cells. HIFU-triggered instantaneous drug release at the glioblastoma sites eventually enabled the ANP-D/P to achieve the strongest antiglioblastoma efficacy with the longest median survival time (56 days) of glioblastoma-bearing mice and the minimum vestiges of tumor cells in the pathological slices among all groups. In conclusion, the HIFU-responsive ANP-D/P in this study provided a new way for glioblastoma therapy with a great potential for clinical applications.
Collapse
Affiliation(s)
- Zimiao Luo
- Biomedical Engineering and Technology Institute, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , 3663 N. Zhongshan Rd., Shanghai 200062, PR China
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Kai Jin
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Qiang Pang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Shun Shen
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Zhiqiang Yan
- Biomedical Engineering and Technology Institute, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , 3663 N. Zhongshan Rd., Shanghai 200062, PR China
| | - Ting Jiang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Xiaoyan Zhu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Lei Yu
- Biomedical Engineering and Technology Institute, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University , 3663 N. Zhongshan Rd., Shanghai 200062, PR China
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| | - Xinguo Jiang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & PLA, Fudan University , 826 N. Zhangheng Rd., Shanghai 201203, PR China
| |
Collapse
|
149
|
Boissenot T, Bordat A, Larrat B, Varna M, Chacun H, Paci A, Poinsignon V, Fattal E, Tsapis N. Ultrasound-induced mild hyperthermia improves the anticancer efficacy of both Taxol® and paclitaxel-loaded nanocapsules. J Control Release 2017; 264:219-227. [PMID: 28867377 DOI: 10.1016/j.jconrel.2017.08.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/22/2017] [Accepted: 08/30/2017] [Indexed: 11/26/2022]
Abstract
We study the influence of ultrasound on paclitaxel-loaded nanocapsules in vitro and in vivo. These nanocapsules possess a shell of poly(dl-lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG) and a liquid core of perfluorooctyl bromide (PFOB). In vitro experiments show that mechanical effects such as cavitation are negligible for nanocapsules due to their small size and thick and rigid shell. As the mechanical effects were unable to increase paclitaxel delivery, we focused on the thermal effects of ultrasound in the in vivo studies. A focused ultrasound sequence was therefore optimized in vivo under magnetic resonance imaging guidance to obtain localized mild hyperthermia with high acoustic pressure. Ultrasound-induced mild hyperthermia (41-43°C) was then tested in vivo in a subcutaneous CT-26 colon cancer murine model. As hyperthermia is applied, an inhibition of tumor growth for both paclitaxel-loaded nanocapsules and the commercial formulation of paclitaxel, namely Taxol® have been observed (p<0.05). Ultrasound-induced mild hyperthermia at high acoustic pressure appears as an interesting strategy to enhance cytotoxic efficacy locally.
Collapse
Affiliation(s)
- Tanguy Boissenot
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Alexandre Bordat
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Benoît Larrat
- Commissariat à l'Energie Atomique (CEA), Institut d'Imagerie Biomédicale (I(2)BM), Neurospin, Saclay, France
| | - Mariana Varna
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Hélène Chacun
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Angelo Paci
- Gustave Roussy Cancer Campus, Service interdépartemental de Pharmacologie et d'Analyse du Médicament (SIPAM), 94800 Villejuif, France
| | - Vianney Poinsignon
- Gustave Roussy Cancer Campus, Service interdépartemental de Pharmacologie et d'Analyse du Médicament (SIPAM), 94800 Villejuif, France
| | - Elias Fattal
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Nicolas Tsapis
- Institut Galien Paris-Sud, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| |
Collapse
|
150
|
Wu P, Jia Y, Qu F, Sun Y, Wang P, Zhang K, Xu C, Liu Q, Wang X. Ultrasound-Responsive Polymeric Micelles for Sonoporation-Assisted Site-Specific Therapeutic Action. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25706-25716. [PMID: 28741924 DOI: 10.1021/acsami.7b05469] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Targeting drug delivery remains a challenge in various disease treatment including cancer. The local drug deposit could be greatly enhanced by some external stimuli-responsive systems. Here we develop pluronic P123/F127 polymeric micelles (M) encapsulating curcumin (Cur) that are permeabilized directly by focused ultrasound, in which ultrasound triggers drug release. Tumor preferential accumulation and site-specific sonochemotherapy were then evaluated. Cur-loaded P123/F127 mixed micelles (Cur-M) exhibited longer circulating time and increased cellular uptake compared to free Cur. With the assistance of focused ultrasound treatment, Cur-M showed tumor-targeting deposition in a time-dependent manner following systemic administration. This was due to enhanced permeabilization of tumor regions and increased penetration of Cur-M in irradiated tumor cells by ultrasound sonoporation. Furthermore, Cur-M self-assembly could be regulated by ultrasound irradiation. In vitro Cur release from mixed micelles was greatly dependent on ultrasound intensity but not on duration, suggesting the cavitational threshold was necessary to initiate subsequent sonochemotherapy. In vivo site-specific drug release was demonstrated in dual-tumor models, which showed spatial-temporal release of entrapped drugs following intratumoral injection. The sonoporation-assisted site-specific chemotherapy significantly inhibited tumor growth and the decrease in tumor weight was approximately 6.5-fold more than without exposure to ultrasound irradiation. In conclusion, the established ultrasound-guided nanomedicine targeting deposit and local release may represent a new strategy to improve chemotherapy efficiency.
Collapse
Affiliation(s)
- Pengying Wu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Yali Jia
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Fei Qu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Yue Sun
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Pan Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Kun Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Chuanshan Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong , Shatin, Hong Kong 999077, China
| | - Quanhong Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Xiaobing Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| |
Collapse
|