1
|
Meijlink B, Collado-Lara G, Bishard K, Conboy JP, Langeveld SAG, Koenderink GH, van der Steen AFW, de Jong N, Beekers I, Trietsch SJ, Kooiman K. Characterizing Microbubble-Mediated Permeabilization in a Vessel-on-a-Chip Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407550. [PMID: 39648449 DOI: 10.1002/smll.202407550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/29/2024] [Indexed: 12/10/2024]
Abstract
Drug transport from blood to extravascular tissue can locally be achieved by increasing the vascular permeability through ultrasound-activated microbubbles. However, the mechanism remains unknown, including whether short and long cycles of ultrasound induce the same onset rate, spatial distribution, and amount of vascular permeability increase. Accurate models are necessary for insights into the mechanism so a microvessel-on-a-chip is developed with a membrane-free extravascular space. Using these microvessels-on-a-chip, distinct differences between 2 MHz ultrasound treatments are shown with 10 or 1000 cycles. The onset rate is slower for 10 than 1000 cycles, while both cycle lengths increase the permeability in spot-wise patterns without affecting microvessel viability. Significantly less vascular permeability increase and sonoporation are induced for 10 versus 1000 cycles at 750 kPa (i.e., the highest studied peak negative acoustic pressure (PNP)). The PNP threshold for vascular permeability increases is 750 versus 550 kPa for 10 versus 1000 cycles, while this is 750 versus 220 kPa for sonoporation. Vascular permeability increases do not correlate with αvβ3-targeted microbubble behavior, while sonoporation correlates with αvβ3-targeted microbubble clustering. In conclusion, the further mechanistic unraveling of vascular permeability increase by ultrasound-activated microbubbles in a developed microvessel-on-a-chip model aids the safe and efficient development of microbubble-mediated drug transport.
Collapse
Affiliation(s)
- Bram Meijlink
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
| | - Gonzalo Collado-Lara
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
| | | | - James P Conboy
- Department of Bionanoscience, Delft University of Technology, Building 58, Van der Maasweg 9, Delft, 2629 HZ, The Netherlands
| | - Simone A G Langeveld
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
| | - Gijsje H Koenderink
- Department of Bionanoscience, Delft University of Technology, Building 58, Van der Maasweg 9, Delft, 2629 HZ, The Netherlands
| | - Antonius F W van der Steen
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
- Department of Imaging Physics, Delft University of Technology, Building 22, Lorentzweg 1, Delft, 2628 CJ, The Netherlands
| | - Nico de Jong
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
- Department of Imaging Physics, Delft University of Technology, Building 22, Lorentzweg 1, Delft, 2628 CJ, The Netherlands
| | - Inés Beekers
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
- Department of Health, ORTEC B.V., Houtsingel 5, Zoetermeer, 2719 EA, The Netherlands
| | | | - Klazina Kooiman
- Biomedical Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC, Wytemaweg 80, Rotterdam, 3015 CN, The Netherlands
| |
Collapse
|
2
|
Pattinson O, Keller SB, Evans ND, Pierron F, Carugo D. An Acoustic Device for Ultra High-Speed Quantification of Cell Strain During Cell-Microbubble Interaction. ACS Biomater Sci Eng 2023; 9:5912-5923. [PMID: 37747762 PMCID: PMC10565720 DOI: 10.1021/acsbiomaterials.3c00757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023]
Abstract
Microbubbles utilize high-frequency oscillations under ultrasound stimulation to induce a range of therapeutic effects in cells, often through mechanical stimulation and permeabilization of cells. One of the largest challenges remaining in the field is the characterization of interactions between cells and microbubbles at therapeutically relevant frequencies. Technical limitations, such as employing sufficient frame rates and obtaining sufficient image resolution, restrict the quantification of the cell's mechanical response to oscillating microbubbles. Here, a novel methodology was developed to address many of these limitations and improve the image resolution of cell-microbubble interactions at high frame rates. A compact acoustic device was designed to house cells and microbubbles as well as a therapeutically relevant acoustic field while being compatible with a Shimadzu HPV-X camera. Cell viability tests confirmed the successful culture and proliferation of cells, and the attachment of DSPC- and cationic DSEPC-microbubbles to osteosarcoma cells was quantified. Microbubble oscillation was observed within the device at a frame rate of 5 million FPS, confirming suitable acoustic field generation and ultra high-speed image capture. High spatial resolution in these images revealed observable deformation in cells following microbubble oscillation and supported the first use of digital image correlation for strain quantification in a single cell. The novel acoustic device provided a simple, effective method for improving the spatial resolution of cell-microbubble interaction images, presenting the opportunity to develop an understanding of the mechanisms driving the therapeutic effects of oscillating microbubbles upon ultrasound exposure.
Collapse
Affiliation(s)
- Oliver Pattinson
- Faculty
of Engineering and Physical Sciences, University
of Southampton, University Road, Southampton SO17 1BJ, United Kingdom
| | - Sara B. Keller
- Department
of Engineering Science, University of Oxford, Old Road, Headington, Oxford OX3 7LD, U.K.
| | - Nicholas D. Evans
- Faculty
of Engineering and Physical Sciences, University
of Southampton, University Road, Southampton SO17 1BJ, United Kingdom
| | - Fabrice Pierron
- Faculty
of Engineering and Physical Sciences, University
of Southampton, University Road, Southampton SO17 1BJ, United Kingdom
| | - Dario Carugo
- Nuffield
Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences
(NDORMS), University of Oxford, Old Road, Headington, Oxford OX3 7LD, United Kingdom
| |
Collapse
|
3
|
Graceffa V. Intracellular protein delivery: New insights into the therapeutic applications and emerging technologies. Biochimie 2023; 213:82-99. [PMID: 37209808 DOI: 10.1016/j.biochi.2023.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
The inability to cross the plasma membranes traditionally limited the therapeutic use of recombinant proteins. However, in the last two decades, novel technologies made delivering proteins inside the cells possible. This allowed researchers to unlock intracellular targets, once considered 'undruggable', bringing a new research area to emerge. Protein transfection systems display a large potential in a plethora of applications. However, their modality of action is often unclear, and cytotoxic effects are elevated, whereas experimental conditions to increase transfection efficacy and cell viability still need to be identified. Furthermore, technical complexity often limits in vivo experimentation, while challenging industrial and clinical translation. This review highlights the applications of protein transfection technologies, and then critically discuss the current methodologies and their limitations. Physical membrane perforation systems are compared to systems exploiting cellular endocytosis. Research evidence of the existence of either extracellular vesicles (EVs) or cell-penetrating peptides (CPPs)- based systems, that circumvent the endosomal systems is critically analysed. Commercial systems, novel solid-phase reverse protein transfection systems, and engineered living intracellular bacteria-based mechanisms are finally described. This review ultimately aims at finding new methodologies and possible applications of protein transfection systems, while helping the development of an evidence-based research approach.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Centre for Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), Atlantic Technological University (ATU), Sligo, Ireland.
| |
Collapse
|
4
|
Eck M, Aronovich R, Ilovitsh T. Efficacy optimization of low frequency microbubble-mediated sonoporation as a drug delivery platform to cancer cells. Int J Pharm X 2022; 4:100132. [PMID: 36189459 PMCID: PMC9520274 DOI: 10.1016/j.ijpx.2022.100132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Ultrasound insonation of microbubbles can be used to form pores in cell membranes and facilitate the local trans-membrane transport of drugs and genes. An important factor in efficient delivery is the size of the delivered target compared to the generated membrane pores. Large molecule delivery remains a challenge, and can affect the resulting therapeutic outcomes. To facilitate large molecule delivery, large pores need to be formed. While ultrasound typically uses megahertz frequencies, it was recently shown that when microbubbles are excited at a frequency of 250 kHz (an order of magnitude below the resonance frequency of these agents), their oscillations are significantly enhanced as compared to the megahertz range. Here, to promote the delivery of large molecules, we suggest using this low frequency and inducing large pore formation through the high-amplitude oscillations of microbubbles. We assessed the impact of low frequency microbubble-mediated sonoporation on breast cancer cell uptake by optimizing the delivery of 4 fluorescent molecules ranging from 1.2 to 70 kDa in size. The optimal ultrasound peak negative pressure was found to be 500 kPa. Increasing the pressure did not enhance the fraction of fluorescent cells, and in fact reduced cell viability. For the smaller molecule sizes, 1.2 kDa and 4 kDa, the groups treated with an ultrasound pressure of 500 kPa and MB resulted in a fraction of 58% and 29% of fluorescent cells respectively, whereas delivery of 20 kDa and 70 kDa molecules yielded 10% and 5%, respectively. These findings suggest that low-frequency (e.g., 250 kHz) insonation of microbubbles results in high amplitude oscillation in vitro that increase the uptake of large molecules. Successful ultrasound-mediated molecule delivery requires the careful selection of insonation parameters to maximize the therapeutic effect by increasing cell uptake.
Collapse
Affiliation(s)
- Michal Eck
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ramona Aronovich
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
5
|
Langeveld SAG, Meijlink B, Beekers I, Olthof M, van der Steen AFW, de Jong N, Kooiman K. Theranostic Microbubbles with Homogeneous Ligand Distribution for Higher Binding Efficacy. Pharmaceutics 2022; 14:pharmaceutics14020311. [PMID: 35214044 PMCID: PMC8878664 DOI: 10.3390/pharmaceutics14020311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/05/2023] Open
Abstract
Phospholipid-coated targeted microbubbles are used for ultrasound molecular imaging and locally enhanced drug delivery, with the binding efficacy being an important trait. The use of organic solvent in microbubble production makes the difference between a heterogeneous or homogeneous ligand distribution. This study demonstrates the effect of ligand distribution on the binding efficacy of phospholipid-coated ανβ3-targeted microbubbles in vitro using a monolayer of human umbilical-vein endothelial cells and in vivo using chicken embryos. Microbubbles with a homogeneous ligand distribution had a higher binding efficacy than those with a heterogeneous ligand distribution both in vitro and in vivo. In vitro, 1.55× more microbubbles with a homogeneous ligand distribution bound under static conditions, while this was 1.49× more under flow with 1.25 dyn/cm2, 1.56× more under flow with 2.22 dyn/cm2, and 1.25× more in vivo. The in vitro dissociation rate of bound microbubbles with homogeneous ligand distribution was lower at low shear stresses (1–5 dyn/cm2). The internalized depth of bound microbubbles was influenced by microbubble size, not by ligand distribution. In conclusion, for optimal binding the use of organic solvent in targeted microbubble production is preferable over directly dispersing phospholipids in aqueous medium.
Collapse
Affiliation(s)
- Simone A. G. Langeveld
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
- Correspondence:
| | - Bram Meijlink
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
| | - Inés Beekers
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
- Department of Health, ORTEC B.V., 2719 EA Zoetermeer, The Netherlands
| | - Mark Olthof
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
| | - Antonius F. W. van der Steen
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
| | - Nico de Jong
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
- Imaging Physics, Delft University of Technology, 2628 CJ Delft, The Netherlands
| | - Klazina Kooiman
- Thorax Center, Biomedical Engineering, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (B.M.); (I.B.); (M.O.); (A.F.W.v.d.S.); (N.d.J.); (K.K.)
| |
Collapse
|
6
|
Sulheim E, Hanson I, Snipstad S, Vikedal K, Mørch Y, Boucher Y, Davies CDL. Sonopermeation with Nanoparticle‐Stabilized Microbubbles Reduces Solid Stress and Improves Nanomedicine Delivery to Tumors. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Einar Sulheim
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Ingunn Hanson
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Sofie Snipstad
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Krister Vikedal
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
| | - Yves Boucher
- Edwin L. Steele Laboratory for Tumor Biology Massachusetts General Hospital Boston MA 02114 USA
| | - Catharina de Lange Davies
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| |
Collapse
|
7
|
Yang W, Xu H, Liu Q, Liu C, Hu J, Liu P, Fang T, Bai Y, Zhu J, Xie R. 5-Aminolevulinic acid hydrochloride loaded microbubbles-mediated sonodynamic therapy in pancreatic cancer cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 48:1178-1188. [PMID: 32924612 DOI: 10.1080/21691401.2020.1813743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
5-Aminolevulinic acid hydrochloride (ALA)-mediated sonodynamic therapy (SDT) had anti-tumour effect on pancreatic cancer cells. Hence, ALA loaded lipid/poly(lactic-co-glycolic acid) (PLGA) microbubbles (MBs)-mediated SDT for pancreatic cancer has great potential. The average size of ALA-lipid MBs and ALA-PLGA MBs was about 3.0 µm. The two kinds of MBs had good biocompatibility to normal HPDE6-C7 cells and were not toxic to pancreatic cancer cells. Compared with ALA-induced SDT, a statistically significant decrease in cell viability was observed in ALA lipid/PLGA MBs combined with ultrasound groups in AsPC-1 and BxPC-3 cells (p < .05). Obvious effect on the apoptotic rate, apoptosis and pyroptosis morphology, enhanced reactive oxygen species was found in ALA-lipid/PLGA MBs mediated SDT in vitro. Through in vivo study, we found ALA-lipid/PLGA MBs-mediated SDT was a promise treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Wei Yang
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Haitao Xu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Qinghao Liu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Chunxun Liu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiahe Hu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Liu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianyi Fang
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuxian Bai
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiuxin Zhu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratories of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Rui Xie
- Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
8
|
Skachkov I, Luan Y, van der Steen AFW, Jong ND, Kooiman K. Corrections to "Targeted Microbubble Mediated Sonoporation of Endothelial Cells In Vivo". IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:2320. [PMID: 33687842 DOI: 10.1109/tuffc.2021.3064763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In the above article [1], the authors regret that there was a mistake in calculating the mol% of the microbubble coating composition. For all experiments, the unit in mg/mL was utilized and the conversion mistake only came when converting to mol% in order to define the ratio between the coating formulation components. The correct molecular weight of PEG-40 stearate is 2046.54 g/mol [2], [3], not 328.53 g/mol. On page 1661, paragraph II-A, it should read "The coating was composed of DSPC (84.8 mol%; P 6517; Sigma-Aldrich, Zwijndrecht, The Netherlands);PEG-40 stearate (8.2 mol%; P 3440; Sigma-Aldrich); DSPE-PEG(2000) (5.9 mol%; 880125 P; Avanti Polar Lipids, Alabaster, AL, USA); and DSPE-PEG(2000)-biotin (1.1 mol%; 880129 C; Avanti Polar Lipids)".
Collapse
|
9
|
Snipstad S, Hanstad S, Bjørkøy A, Mørch Ý, de Lange Davies C. Sonoporation Using Nanoparticle-Loaded Microbubbles Increases Cellular Uptake of Nanoparticles Compared to Co-Incubation of Nanoparticles and Microbubbles. Pharmaceutics 2021; 13:640. [PMID: 33946327 PMCID: PMC8146007 DOI: 10.3390/pharmaceutics13050640] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022] Open
Abstract
Therapeutic agents can benefit from encapsulation in nanoparticles, due to improved pharmacokinetics and biodistribution, protection from degradation, increased cellular uptake and sustained release. Microbubbles in combination with ultrasound have been shown to improve the delivery of nanoparticles and drugs to tumors and across the blood-brain barrier. Here, we evaluate two different microbubbles for enhancing the delivery of polymeric nanoparticles to cells in vitro: a commercially available lipid microbubble (Sonazoid) and a microbubble with a shell composed of protein and nanoparticles. Various ultrasound parameters are applied and confocal microscopy is employed to image cellular uptake. Ultrasound enhanced cellular uptake depending on the pressure and duty cycle. The responsible mechanisms are probably sonoporation and sonoprinting, followed by uptake, and to a smaller degree enhanced endocytosis. The use of commercial Sonazoid microbubbles leads to significantly lower uptake than when using nanoparticle-loaded microbubbles, suggesting that proximity between cells, nanoparticles and microbubbles is important, and that mainly nanoparticles in the shell are taken up, rather than free nanoparticles in solution.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
- Cancer Clinic, St. Olav’s Hospital, Prinsesse Kristinas Gate 1, 7030 Trondheim, Norway
| | - Sigurd Hanstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| |
Collapse
|
10
|
Ho YJ, Chang HC, Lin CW, Fan CH, Lin YC, Wei KC, Yeh CK. Oscillatory behavior of microbubbles impacts efficacy of cellular drug delivery. J Control Release 2021; 333:316-327. [PMID: 33811982 DOI: 10.1016/j.jconrel.2021.03.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 01/16/2023]
Abstract
Drug-loaded microbubbles have been proven to be an effective strategy for non-invasive and local drug delivery when combined with ultrasound excitation for targeted drug release. Inertial cavitation is speculated to be a major mechanism for releasing drugs from drug-loaded microbubbles, but it results in lethal cellular pore damage that greatly limits its application. Thus, we investigated the cellular vesicle attachment and uptake to evaluate the efficiency of drug delivery by modulating the behaviors of targeted microbubble oscillation. The efficiency of vesicle attachment on the targeted cell membrane was 36.5 ± 15.9% and 3.8 ± 2.3% under stable and inertial cavitation, respectively. Further, stable cavitation enhanced cell permeability (26.8 ± 3.2%), maintained cell viability (90.8 ± 2.1%), and showed 7.9 ± 1.9-fold enhancement of in vivo vesicle release on tumor vessels. Therefore, our results reveal the ability to improve drug delivery via stable cavitation induced by targeted microbubbles. We propose that this strategy might be suitable for tissue repair or neuromodulation.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ho-Chun Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan; Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan.
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, New Taipei City, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
11
|
Mashel TV, Tarakanchikova YV, Muslimov AR, Zyuzin MV, Timin AS, Lepik KV, Fehse B. Overcoming the delivery problem for therapeutic genome editing: Current status and perspective of non-viral methods. Biomaterials 2020; 258:120282. [PMID: 32798742 DOI: 10.1016/j.biomaterials.2020.120282] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/22/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
|
12
|
Presset A, Bonneau C, Kazuyoshi S, Nadal-Desbarats L, Mitsuyoshi T, Bouakaz A, Kudo N, Escoffre JM, Sasaki N. Endothelial Cells, First Target of Drug Delivery Using Microbubble-Assisted Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1565-1583. [PMID: 32331799 DOI: 10.1016/j.ultrasmedbio.2020.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 06/11/2023]
Abstract
Microbubble-assisted ultrasound has emerged as a promising method for local drug delivery. Microbubbles are intravenously injected and locally activated by ultrasound, thus increasing the permeability of vascular endothelium for facilitating extravasation and drug uptake into the treated tissue. Thereby, endothelial cells are the first target of the effects of ultrasound-driven microbubbles. In this review, the in vitro and in vivo bioeffects of this method on endothelial cells are described and discussed, including aspects on the permeabilization of biologic barriers (endothelial cell plasma membranes and endothelial barriers), the restoration of their integrity, the molecular and cellular mechanisms involved in both these processes, and the resulting intracellular and intercellular consequences. Finally, the influence of the acoustic settings, microbubble parameters, treatment schedules and flow parameters on these bioeffects are also reviewed.
Collapse
Affiliation(s)
- Antoine Presset
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | - Sasaoka Kazuyoshi
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | - Takigucho Mitsuyoshi
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Nobuki Kudo
- Laboratory of Biological Engineering, Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | | | - Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
13
|
Ilovitsh T, Feng Y, Foiret J, Kheirolomoom A, Zhang H, Ingham ES, Ilovitsh A, Tumbale SK, Fite BZ, Wu B, Raie MN, Zhang N, Kare AJ, Chavez M, Qi LS, Pelled G, Gazit D, Vermesh O, Steinberg I, Gambhir SS, Ferrara KW. Low-frequency ultrasound-mediated cytokine transfection enhances T cell recruitment at local and distant tumor sites. Proc Natl Acad Sci U S A 2020; 117:12674-12685. [PMID: 32430322 PMCID: PMC7293655 DOI: 10.1073/pnas.1914906117] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Robust cytotoxic T cell infiltration has proven to be difficult to achieve in solid tumors. We set out to develop a flexible protocol to efficiently transfect tumor and stromal cells to produce immune-activating cytokines, and thus enhance T cell infiltration while debulking tumor mass. By combining ultrasound with tumor-targeted microbubbles, membrane pores are created and facilitate a controllable and local transfection. Here, we applied a substantially lower transmission frequency (250 kHz) than applied previously. The resulting microbubble oscillation was significantly enhanced, reaching an effective expansion ratio of 35 for a peak negative pressure of 500 kPa in vitro. Combining low-frequency ultrasound with tumor-targeted microbubbles and a DNA plasmid construct, 20% of tumor cells remained viable, and ∼20% of these remaining cells were transfected with a reporter gene both in vitro and in vivo. The majority of cells transfected in vivo were mucin 1+/CD45- tumor cells. Tumor and stromal cells were then transfected with plasmid DNA encoding IFN-β, producing 150 pg/106 cells in vitro, a 150-fold increase compared to no-ultrasound or no-plasmid controls and a 50-fold increase compared to treatment with targeted microbubbles and ultrasound (without IFN-β). This enhancement in secretion exceeds previously reported fourfold to fivefold increases with other in vitro treatments. Combined with intraperitoneal administration of checkpoint inhibition, a single application of IFN-β plasmid transfection reduced tumor growth in vivo and recruited efficacious immune cells at both the local and distant tumor sites.
Collapse
Affiliation(s)
- Tali Ilovitsh
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Yi Feng
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Biomedical Engineering, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Josquin Foiret
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Azadeh Kheirolomoom
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Hua Zhang
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Asaf Ilovitsh
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Spencer K Tumbale
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Brett Z Fite
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Bo Wu
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Marina N Raie
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Nisi Zhang
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Aris J Kare
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Michael Chavez
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Gadi Pelled
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Dan Gazit
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ophir Vermesh
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Idan Steinberg
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
- Department of Radiology, Stanford University, Stanford, CA 94305
| | - Katherine W Ferrara
- Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305;
- Department of Radiology, Stanford University, Stanford, CA 94305
| |
Collapse
|
14
|
Opening of endothelial cell-cell contacts due to sonoporation. J Control Release 2020; 322:426-438. [PMID: 32246975 DOI: 10.1016/j.jconrel.2020.03.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 01/06/2023]
Abstract
Ultrasound insonification of microbubbles can locally increase vascular permeability to enhance drug delivery. To control and optimize the therapeutic potential, we need to better understand the underlying biological mechanisms of the drug delivery pathways. The aim of this in vitro study was to elucidate the microbubble-endothelial cell interaction using the Brandaris 128 ultra-high-speed camera (up to 25 Mfps) coupled to a custom-built Nikon confocal microscope, to visualize both microbubble oscillation and the cellular response. Sonoporation and opening of cell-cell contacts by single αVβ3-targeted microbubbles (n = 152) was monitored up to 4 min after ultrasound insonification (2 MHz, 100-400 kPa, 10 cycles). Sonoporation occurred when microbubble excursion amplitudes exceeded 0.7 μm. Quantification of the influx of the fluorescent model drug propidium iodide upon sonoporation showed that the size of the created pore increased for larger microbubble excursion amplitudes. Microbubble-mediated opening of cell-cell contacts occurred as a cellular response upon sonoporation and did not correlate with the microbubble excursion amplitude itself. The initial integrity of the cell-cell contacts affected the susceptibly to drug delivery, since cell-cell contacts opened more often when cells were only partially attached to their neighbors (48%) than when fully attached (14%). The drug delivery outcomes were independent of nonlinear microbubble behavior, microbubble location, and cell size. In conclusion, by studying the microbubble-cell interaction at nanosecond and nanometer resolution the relationship between drug delivery pathways and their underlying mechanisms was further unraveled. These novel insights will aid the development of safe and efficient microbubble-mediated drug delivery.
Collapse
|
15
|
Koda R, Origasa T, Nakajima T, Yamakoshi Y. Observing Bubble Cavitation by Back-Propagation of Acoustic Emission Signals. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2019; 66:823-833. [PMID: 30735990 DOI: 10.1109/tuffc.2019.2897983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Temporal- and spatial-resolved observations of microbubble cavitation generated through high-intensity ultrasound irradiation are key in improving both the efficiency and efficacy of ultrasound-assisted drug delivery systems. A method of measuring bubble cavitation applying an image-reconstruction technique of back-propagation of an acoustic cavitation emission (ACE) signal is proposed. A high-intensity focused ultrasound wave (pump wave) irradiates the bubble synchronously using ultrasound recording equipment to acquire the timing of the RF signal, which is produced when the bubble radiates a secondary wave during bubble cavitation. The ACE signal source is reconstructed through ultrasound-wave back-propagation followed by amplitude deconvolution. The proposed method was applied to microbubbles of an ultrasound contrast agent by changing the sound pressure of the pump wave. The method reliability of the temporal resolution was verified by simulating the amplitude-modulated signal of the virtual sound source. The temporal transition of the ACE signal exhibited sub-microsecond-order fluctuations in the signal intensity. From the amplitude signal image and the instantaneous frequency image reconstruction of the proposed method, two different ACE phenomena were visualized. One is the periodic pattern by the beat signals from the harmonic and ultraharmonic component of nonlinear oscillation under low-intensity ultrasound conditions. The other is the nonperiodic temporal and spatial distributions of this irradiation under high-intensity ultrasound conditions.
Collapse
|
16
|
Du X, Wang J, Zhou Q, Zhang L, Wang S, Zhang Z, Yao C. Advanced physical techniques for gene delivery based on membrane perforation. Drug Deliv 2018; 25:1516-1525. [PMID: 29968512 PMCID: PMC6058615 DOI: 10.1080/10717544.2018.1480674] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Gene delivery as a promising and valid tool has been used for treating many serious diseases that conventional drug therapies cannot cure. Due to the advancement of physical technology and nanotechnology, advanced physical gene delivery methods such as electroporation, magnetoporation, sonoporation and optoporation have been extensively developed and are receiving increasing attention, which have the advantages of briefness and nontoxicity. This review introduces the technique detail of membrane perforation, with a brief discussion for future development, with special emphasis on nanoparticles mediated optoporation that have developed as an new alternative transfection technique in the last two decades. In particular, the advanced physical approaches development and new technology are highlighted, which intends to stimulate rapid advancement of perforation techniques, develop new delivery strategies and accelerate application of these techniques in clinic.
Collapse
Affiliation(s)
- Xiaofan Du
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Jing Wang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Quan Zhou
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Luwei Zhang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Sijia Wang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Zhenxi Zhang
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Cuiping Yao
- a Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Analytical Technology and Instrumentation , School of Life Science and Technology, Xi'an Jiaotong University , Xi'an , People's Republic of China
| |
Collapse
|
17
|
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: 87] [Impact Index Per Article: 12.4] [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
|
18
|
Skachkov I, Luan Y, van Tiel ST, van der Steen AFW, de Jong N, Bernsen MR, Kooiman K. SPIO labeling of endothelial cells using ultrasound and targeted microbubbles at diagnostic pressures. PLoS One 2018; 13:e0204354. [PMID: 30235336 PMCID: PMC6147550 DOI: 10.1371/journal.pone.0204354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/06/2018] [Indexed: 02/07/2023] Open
Abstract
In vivo cell tracking of therapeutic, tumor, and endothelial cells is an emerging field and a promising technique for imaging cardiovascular disease and cancer development. Site-specific labeling of endothelial cells with the MRI contrast agent superparamagnetic iron oxide (SPIO) in the absence of toxic agents is challenging. Therefore, the aim of this in vitro study was to find optimal parameters for efficient and safe SPIO-labeling of endothelial cells using ultrasound-activated CD31-targeted microbubbles for future MRI tracking. Ultrasound at a frequency of 1 MHz (10,000 cycles, repetition rate of 20 Hz) was used for varying applied peak negative pressures (10–160 kPa, i.e. low mechanical index (MI) of 0.01–0.16), treatment durations (0–30 s), time of SPIO addition (-5 min– 15 min with respect to the start of the ultrasound), and incubation time after SPIO addition (5 min– 3 h). Iron specific Prussian Blue staining in combination with calcein-AM based cell viability assays were applied to define the most efficient and safe conditions for SPIO-labeling. Optimal SPIO labeling was observed when the ultrasound parameters were 40 kPa peak negative pressure (MI 0.04), applied for 30 s just before SPIO addition (0 min). Compared to the control, this resulted in an approximate 12 times increase of SPIO uptake in endothelial cells in vitro with 85% cell viability. Therefore, ultrasound-activated targeted ultrasound contrast agents show great potential for effective and safe labeling of endothelial cells with SPIO.
Collapse
Affiliation(s)
- Ilya Skachkov
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Ying Luan
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
| | - Sandra T. van Tiel
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Antonius F. W. van der Steen
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Nico de Jong
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Monique R. Bernsen
- Department of Radiology & Nucleair Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, the Netherlands
- * E-mail:
| |
Collapse
|
19
|
D'Arrigo JS. Targeting Early Dementia: Using Lipid Cubic Phase Nanocarriers to Cross the Blood⁻Brain Barrier. Biomimetics (Basel) 2018; 3:E4. [PMID: 31105226 PMCID: PMC6352688 DOI: 10.3390/biomimetics3010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, a frequent co-morbidity of cerebrovascular pathology and Alzheimer's disease has been observed. Numerous published studies indicate that the preservation of a healthy cerebrovascular endothelium can be an important therapeutic target. By incorporating the appropriate drug(s) into biomimetic (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic, which targets certain cell surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood⁻brain barrier. This targeting allows for various cell types related to Alzheimer's to be simultaneously searched out for localized drug treatment in vivo.
Collapse
|
20
|
Nanotherapy for Alzheimer's disease and vascular dementia: Targeting senile endothelium. Adv Colloid Interface Sci 2018; 251:44-54. [PMID: 29274774 DOI: 10.1016/j.cis.2017.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022]
Abstract
Due to the complexity of Alzheimer's disease, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted lipid nanoemulsion) are available. Versatile small molecule drug(s) targeting multiple pathways of Alzheimer's disease pathogenesis are known. By incorporating such drug(s) into the targeted "lipid-coated microbubble" [LCM]/"nanoparticle-derived" [ND] (or LCM/ND) nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly class B type I), or SR-BI, making possible for various Alzheimer's-related cell types to be simultaneously searched out for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble [LCM] subpopulation (i.e., a stable LCM suspension); such film-stabilized microbubbles are well known to substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer's patient.
Collapse
|
21
|
Alzheimer’s Disease, Brain Injury, and C.N.S. Nanotherapy in Humans: Sonoporation Augmenting Drug Targeting. Med Sci (Basel) 2017. [PMCID: PMC5753658 DOI: 10.3390/medsci5040029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Owing to the complexity of neurodegenerative diseases, multiple cellular types need to be targeted simultaneously in order for a given therapy to demonstrate any major effectiveness. Ultrasound-sensitive coated microbubbles (in a targeted nanoemulsion) are available. Versatile small-molecule drug(s) targeting multiple pathways of Alzheimer’s disease pathogenesis are known. By incorporating such drug(s) into the targeted lipid-coated microbubble/nanoparticle-derived (LCM/ND) lipid nanoemulsion type, one obtains a multitasking combination therapeutic for translational medicine. This multitasking therapeutic targets cell-surface scavenger receptors (mainly scavenger receptor class B type I (SR-BI)), making it possible for various Alzheimer’s-related cell types to be simultaneously sought for localized drug treatment in vivo. Besides targeting cell-surface SR-BI, the proposed LCM/ND-nanoemulsion combination therapeutic(s) include a characteristic lipid-coated microbubble (LCM) subpopulation (i.e., a stable LCM suspension); such LCM substantially reduce the acoustic power levels needed for accomplishing temporary noninvasive (transcranial) ultrasound treatment, or sonoporation, if additionally desired for the Alzheimer’s patient.
Collapse
|
22
|
van Rooij T, Beekers I, Lattwein KR, van der Steen AFW, de Jong N, Kooiman K. Vibrational Responses of Bound and Nonbound Targeted Lipid-Coated Single Microbubbles. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2017; 64:785-797. [PMID: 28287967 DOI: 10.1109/tuffc.2017.2679160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
One of the main challenges for ultrasound molecular imaging is acoustically distinguishing nonbound microbubbles from those bound to their molecular target. In this in vitro study, we compared two types of in-house produced targeted lipid-coated microbubbles, either consisting of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, C16:0 (DPPC) or 1,2-distearoyl-sn-glycero-3-phosphocholine, C18:0 (DSPC) as the main lipid, using the Brandaris 128 ultrahigh-speed camera to determine vibrational response differences between bound and nonbound biotinylated microbubbles. In contrast to previous studies that studied vibrational differences upon binding, we used a covalently bound model biomarker (i.e., streptavidin) rather than physisorption, to ensure binding of the biomarker to the membrane. The microbubbles were insonified at frequencies between 1 and 4 MHz at pressures of 50 and 150 kPa. This paper shows lower acoustic stability of bound microbubbles, of which DPPC-based microbubbles deflated most. For DPPC microbubbles with diameters between 2 and [Formula: see text] driven at 50 kPa, resonance frequencies of bound microbubbles were all higher than 1.8 MHz, whereas those of nonbound microbubbles were significantly lower. In addition, the relative radial excursions at resonance were also higher for bound DPPC microbubbles. These differences did not persist when the pressure was increased to 150 kPa, except for the acoustic stability which further decreased. No differences in resonance frequencies were observed between bound and nonbound DSPC microbubbles. Nonlinear responses in terms of emissions at the subharmonic and second harmonic frequencies were similar for bound and nonbound microbubbles at both pressures. In conclusion, we identified differences in vibrational responses of bound DPPC microbubbles with diameters between 2 and [Formula: see text] that distinguish them from nonbound ones.
Collapse
|
23
|
Ho YJ, Yeh CK. Theranostic Performance of Acoustic Nanodroplet Vaporization-Generated Bubbles in Tumor Intertissue. Am J Cancer Res 2017; 7:1477-1488. [PMID: 28529631 PMCID: PMC5436507 DOI: 10.7150/thno.19099] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 02/10/2017] [Indexed: 12/27/2022] Open
Abstract
Solid tumors with poorly perfused regions reveal some of the treatment limitations that restrict drug delivery and therapeutic efficacy. Acoustic droplet vaporization (ADV) has been applied to directly disrupt vessels and release nanodroplets, ADV-generated bubbles (ADV-Bs), and drugs into tumor tissue. In this study, we investigated the in vivo behavior of ADV-Bs stimulated by US, and evaluated the possibility of moving intertissue ADV-Bs into the poorly perfused regions of solid tumors. Intravital imaging revealed intertissue ADV-B formation, movement, and cavitation triggered by US, where the distance of intertissue ADV-B movement was 33-99 µm per pulse. When ADV-Bs were applied to tumor cells, the cell membrane was damaged, increasing cellular permeability or inducing cell death. The poorly perfused regions within solid tumors show enhancement due to ADV-B accumulation after application of US-triggered ADV-B. The intratumoral nanodroplet or ADV-B distribution around the poorly perfused regions with tumor necrosis or hypoxia were demonstrated by histological assessment. ADV-B formation, movement and cavitation could induce cell membrane damage by mechanical force providing a mechanism to overcome treatment limitations in poorly perfused regions of tumors.
Collapse
|
24
|
Fix SM, Novell A, Yun Y, Dayton PA, Arena CB. An evaluation of the sonoporation potential of low-boiling point phase-change ultrasound contrast agents in vitro. J Ther Ultrasound 2017; 5:7. [PMID: 28127427 PMCID: PMC5260003 DOI: 10.1186/s40349-017-0085-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 01/06/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Phase-change ultrasound contrast agents (PCCAs) offer a solution to the inherent limitations associated with using microbubbles for sonoporation; they are characterized by prolonged circulation lifetimes, and their nanometer-scale sizes may allow for passive accumulation in solid tumors. As a first step towards the goal of extravascular cell permeabilization, we aim to characterize the sonoporation potential of a low-boiling point formulation of PCCAs in vitro. METHODS Parameters to induce acoustic droplet vaporization and subsequent microbubble cavitation were optimized in vitro using high-speed optical microscopy. Sonoporation of pancreatic cancer cells in suspension was then characterized at a range of pressures (125-600 kPa) and pulse lengths (5-50 cycles) using propidium iodide as an indicator molecule. RESULTS We achieved sonoporation efficiencies ranging from 8 ± 1% to 36 ± 4% (percent of viable cells), as evidenced by flow cytometry. Increasing sonoporation efficiency trended with increasing pulse length and peak negative pressure. CONCLUSIONS We conclude that PCCAs can be used to induce the sonoporation of cells in vitro, and our results warrant further investigation into the use of PCCAs as extravascular sonoporation agents in vivo.
Collapse
Affiliation(s)
- Samantha M Fix
- Eshelman School of Pharmacy, University of North Carolina Chapel Hill, Chapel Hill, NC USA
| | - Anthony Novell
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Chapel Hill, NC USA
| | - Yeoheung Yun
- FIT BEST Laboratory, Chemical, Biological and Bioengineering Department, North Carolina A&T State University, Greensboro, NC USA
| | - Paul A Dayton
- Eshelman School of Pharmacy, University of North Carolina Chapel Hill, Chapel Hill, NC USA.,Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Chapel Hill, NC USA
| | - Christopher B Arena
- Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Chapel Hill, NC USA.,Laboratory for Therapeutic Directed Energy, Department of Physics, Elon University, Elon, NC USA
| |
Collapse
|
25
|
Dimcevski G, Kotopoulis S, Bjånes T, Hoem D, Schjøtt J, Gjertsen BT, Biermann M, Molven A, Sorbye H, McCormack E, Postema M, Gilja OH. A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release 2016; 243:172-181. [PMID: 27744037 DOI: 10.1016/j.jconrel.2016.10.007] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND The primary aim of our study was to evaluate the safety and potential toxicity of gemcitabine combined with microbubbles under sonication in inoperable pancreatic cancer patients. The secondary aim was to evaluate a novel image-guided microbubble-based therapy, based on commercially available technology, towards improving chemotherapeutic efficacy, preserving patient performance status, and prolonging survival. METHODS Ten patients were enrolled and treated in this Phase I clinical trial. Gemcitabine was infused intravenously over 30min. Subsequently, patients were treated using a commercial clinical ultrasound scanner for 31.5min. SonoVue® was injected intravenously (0.5ml followed by 5ml saline every 3.5min) during the ultrasound treatment with the aim of inducing sonoporation, thus enhancing therapeutic efficacy. RESULTS The combined therapeutic regimen did not induce any additional toxicity or increased frequency of side effects when compared to gemcitabine chemotherapy alone (historical controls). Combination treated patients (n=10) tolerated an increased number of gemcitabine cycles compared with historical controls (n=63 patients; average of 8.3±6.0cycles, versus 13.8±5.6cycles, p=0.008, unpaired t-test). In five patients, the maximum tumour diameter was decreased from the first to last treatment. The median survival in our patients (n=10) was also increased from 8.9months to 17.6months (p=0.011). CONCLUSIONS It is possible to combine ultrasound, microbubbles, and chemotherapy in a clinical setting using commercially available equipment with no additional toxicities. This combined treatment may improve the clinical efficacy of gemcitabine, prolong the quality of life, and extend survival in patients with pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Georg Dimcevski
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Spiros Kotopoulis
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Tormod Bjånes
- Laboratory of Clinical Biochemistry, Section of Clinical Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Dag Hoem
- Department of Surgical Sciences, Haukeland University Hospital, Norway
| | - Jan Schjøtt
- Laboratory of Clinical Biochemistry, Section of Clinical Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bjørn Tore Gjertsen
- Centre for Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Martin Biermann
- Department of Radiology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Anders Molven
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Emmet McCormack
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Michiel Postema
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warszawa, Poland; School of Electrical and Information Engineering, Chamber of Mines Building, University of the Witwatersrand, Johannesburg, South Africa
| | - Odd Helge Gilja
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
26
|
van Rooij T, Skachkov I, Beekers I, Lattwein KR, Voorneveld JD, Kokhuis TJ, Bera D, Luan Y, van der Steen AF, de Jong N, Kooiman K. Viability of endothelial cells after ultrasound-mediated sonoporation: Influence of targeting, oscillation, and displacement of microbubbles. J Control Release 2016; 238:197-211. [DOI: 10.1016/j.jconrel.2016.07.037] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/20/2016] [Accepted: 07/24/2016] [Indexed: 10/21/2022]
|
27
|
Hua X, Ding J, Li R, Zhang Y, Huang Z, Guo Y, Chen Q. Anti-tumor effect of ultrasound-induced Nordy-loaded microbubbles destruction. J Drug Target 2016; 24:703-8. [PMID: 26811100 DOI: 10.3109/1061186x.2016.1144058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Synthesized dl-Nordihydroguaiaretic acid (dl-NGDA or "Nordy") can inhibit the growth of malignant human tumors, especially the tumor angiogenesis. However, its liposoluble nature limits its in vivo efficacy in the hydrosoluble circulation of human. PURPOSE We tried to use the ultrasonic microbubble as the carrier and the ultrasound-induced destruction for the targeted release of Nordy and evaluate its in vitro and in vivo anti-tumor effect. METHODS Nordy-loaded lipid microbubbles were prepared by mechanical vibration. Effects of ultrasound-induced Nordy-loaded microbubbles destruction on proliferation of human umbilical vein endothelial cells (HUVECs), tumor derived endothelial cells (Td-ECs), and rabbit transplanted VX2 tumor models were evaluated. RESULTS The ultrasound-induced Nordy-loaded microbubbles destruction inhibited the proliferations of HUVECs and Td-ECs in vitro, and inhibited the tumor growth and the microvasculature in vivo. Its efficacy was higher than those of Nordy used only and Nordy with ultrasound exposure. CONCLUSION Ultrasonic microbubbles can be used as the carrier of Nordy and achieve its targeted release with improved anti-tumor efficacy in the condition of ultrasound-induced microbubbles destruction.
Collapse
Affiliation(s)
- Xing Hua
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| | - Jun Ding
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| | - Rui Li
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| | - Ying Zhang
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| | - Zejun Huang
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| | - Yanli Guo
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| | - Qinghai Chen
- a Third Military Medical University, Southwest Hospital , Chongqing , China
| |
Collapse
|
28
|
Maciulevicius M, Tamosiunas M, Jurkonis R, Venslauskas MS, Satkauskas S. Analysis of Metrics for Molecular Sonotransfer in Vitro. Mol Pharm 2015; 12:3620-7. [PMID: 26312556 DOI: 10.1021/acs.molpharmaceut.5b00347] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Ultrasound induced microbubble (MB) cavitation is used to significantly enhance cell membrane permeabilization, thereby allowing delivery of various therapeutic agents into cells. In order to monitor and quantitatively control the extent of cavitation the uniform dosimetry model is needed. In present study we have simultaneously performed quantitative evaluation of three main sonoporation factors: (1) MB concentration, (2) MB cavitation extent, and (3) doxorubicin (DOX) sonotransfer into Chinese hamster ovary cells. MB concentration measurement results and passively recorded MB cavitation signals were used for MB sonodestruction rate and spectral root-mean-square (RMS) calculations, respectively. Subsequently, time to maximum value of RMS and inertial cavitation dose (ICD) quantifications were performed for every acoustic pressure value. This comprehensive research has led not only to explanation of relation of ICD and MB sonodestruction rate but also to the development of a new sonoporation metric: the inverse of time to maximum value of RMS (1/time to maximum value of RMS). ICD and MB sonodestruction rate intercorrelation and correlation with DOX sonotransfer suggest inertial cavitation to be the key mechanism for cell sonoporation. All these metrics were successfully used for doxorubicin sonotransfer prediction (R(2) > 0.9, p < 0.01) and therefore shows feasibility to be applied for future dosimetric applications for ultrasound-mediated drug and gene delivery.
Collapse
Affiliation(s)
| | - Mindaugas Tamosiunas
- Biophysical Research Group, Vytautas Magnus University , Kaunas 44248, Lithuania
| | - Rytis Jurkonis
- Biomedical Engineering Institute, Kaunas University of Technology , Kaunas 44249, Lithuania
| | | | - Saulius Satkauskas
- Biophysical Research Group, Vytautas Magnus University , Kaunas 44248, Lithuania
| |
Collapse
|
29
|
van Rooij T, Daeichin V, Skachkov I, de Jong N, Kooiman K. Targeted ultrasound contrast agents for ultrasound molecular imaging and therapy. Int J Hyperthermia 2015; 31:90-106. [PMID: 25707815 DOI: 10.3109/02656736.2014.997809] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ultrasound contrast agents (UCAs) are used routinely in the clinic to enhance contrast in ultrasonography. More recently, UCAs have been functionalised by conjugating ligands to their surface to target specific biomarkers of a disease or a disease process. These targeted UCAs (tUCAs) are used for a wide range of pre-clinical applications including diagnosis, monitoring of drug treatment, and therapy. In this review, recent achievements with tUCAs in the field of molecular imaging, evaluation of therapy, drug delivery, and therapeutic applications are discussed. We present the different coating materials and aspects that have to be considered when manufacturing tUCAs. Next to tUCA design and the choice of ligands for specific biomarkers, additional techniques are discussed that are applied to improve binding of the tUCAs to their target and to quantify the strength of this bond. As imaging techniques rely on the specific behaviour of tUCAs in an ultrasound field, it is crucial to understand the characteristics of both free and adhered tUCAs. To image and quantify the adhered tUCAs, the state-of-the-art techniques used for ultrasound molecular imaging and quantification are presented. This review concludes with the potential of tUCAs for drug delivery and therapeutic applications.
Collapse
Affiliation(s)
- Tom van Rooij
- Department of Biomedical Engineering, Thoraxcenter , Erasmus MC, Rotterdam , the Netherlands
| | | | | | | | | |
Collapse
|