1
|
Estifeeva TM, Nechaeva AM, Le-Deygen IM, Adelyanov AM, Grigoryan IV, Petrovskii VS, Potemkin II, Abramov AA, Prosvirnin AV, Sencha EA, Borozdenko DA, Barmin RA, Mezhuev YO, Gorin DA, Rudakovskaya PG. Ultrasound protein-copolymer microbubble library engineering through poly(vinylpyrrolidone-co-acrylic acid) structure. BIOMATERIALS ADVANCES 2025; 166:214074. [PMID: 39447238 DOI: 10.1016/j.bioadv.2024.214074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
HYPOTHESIS While albumin-coated microbubbles are routine contrast agents for ultrasound imaging, their short duration of contrast enhancement limits their use, yet can be improved by incorporating protein-copolymer hybrids into microbubble shells. The incorporation of N-vinyl-2-pyrrolidone and acrylic acid copolymer (P(VP-AA)) has been shown to enhance the performance of bovine serum albumin (BSA)-coated microbubbles. However, the impact of the copolymer structural properties on key microbubble characteristics (i.e., concentration, mean diameter and acoustic response) remains poorly understood. Therefore, we hypothesize that the copolymer structure affects its capacity to form micelle-like nanoaggregates, protein-copolymer hybrids, and microbubble shells, ultimately influencing the physicochemical and acoustic properties of the microbubbles. EXPERIMENTS Here we evaluate the production and performance of BSA@P(VP-AA) microbubbles synthesized using a series of P(VP-AA) copolymers with -C8H17 and -C18H37 end groups and molecular weight cutoffs between 3.5 and 15 kDa. Both simulation and experimental data demonstrate that interactions between BSA and the copolymers significantly influence the performance of the resulting microbubbles across the library of 60 formulations. FINDINGS The introduction of -C8H17 terminated copolymers into microbubble shells resulted in up to 200-fold higher concentration, 7-fold greater acoustic response, and 5-fold longer ultrasound contrast enhancement compared to plain BSA microbubbles. The enhanced acoustic performance was sustained during in vivo cardiac ultrasound imaging, without altering liver accumulation after copolymer introduction. These findings underscore how optimizing copolymer structure (specifically the terminal end group and molecular weight) can tailor the formation and performance of protein-copolymer-coated microbubbles, offering valuable insights for designing ultrasound contrast agents.
Collapse
Affiliation(s)
- Tatiana M Estifeeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Anna M Nechaeva
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russia
| | - Irina M Le-Deygen
- Chemical Enzymology Department, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Artem M Adelyanov
- Physics Department, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Ilya V Grigoryan
- Physics Department, Lomonosov Moscow State University, Moscow 119991, Russia
| | | | - Igor I Potemkin
- Physics Department, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Alexander A Abramov
- Laboratory of Experimental Heart Pathology, Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Ministry of Health of Russia, Moscow 121552, Russia
| | - Anton V Prosvirnin
- Laboratory of Experimental Heart Pathology, Institute of Experimental Cardiology, Chazov National Medical Research Center for Cardiology, Ministry of Health of Russia, Moscow 121552, Russia
| | - Ekaterina A Sencha
- Department of Clinical Ultrasound and Functional Diagnostics, M.F. Vladimirsky Moscow Regional Clinical Research Institute (MONIKI), Moscow 129110, Russia
| | - Denis A Borozdenko
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, Ministry of Health of Russia, Moscow 117997, Russia
| | - Roman A Barmin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| | - Yaroslav O Mezhuev
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Moscow 125047, Russia; A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Moscow 119334, Russia
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Polina G Rudakovskaya
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| |
Collapse
|
2
|
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
|
3
|
Wang X, Tan Y, Gao L, Gao H. Study on ultrasound-enhanced molecular transport in articular cartilage. Drug Deliv Transl Res 2024; 14:3621-3639. [PMID: 39145819 DOI: 10.1007/s13346-024-01695-7] [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] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
Local intra-articular administration with minimal side effects and rapid efficacy is a promising strategy for treating osteoarthritis(OA). Most drugs are rapidly cleared from the joint space by capillaries and lymphatic vessels before free diffusion into cartilage. Ultrasound, as a non-invasive therapy, enhances molecular transport within cartilage through the mechanisms of microbubble cavitation and thermal effects. This study investigated the mass transfer behavior of solute molecules with different molecular weights (479 Da, 40 kDa, 150 kDa) within porcine articular cartilage under low-frequency ultrasound conditions of 40 kHz and ultrasound intensities of 0.189 W/cm2 and 0.359 W/cm2. The results revealed that under the conditions of 0.189 W/cm2 ultrasound intensity, the mass transfer concentration of solute molecules were higher compared to passive diffusion, and with an increase in ultrasound intensity to 0.359 W/cm2, the mass transfer effect within the cartilage was further enhanced. Ultrasound promotes molecular transport in different layers of cartilage. Under static conditions, after 2 h of mass transfer, the concentration of small molecules in the superficial layer is lower than that in the middle layer. After applying ultrasound at 0.189 W/cm2, the molecular concentration in the superficial layer significantly increases. Under conditions of 0.359 W/cm2, after 12 h of mass transfer, the concentration of medium and large molecules in the deep layer region increased by more than two times. In addition, this study conducted an assessment of damage to porcine articular cartilage under ultrasound exposure, revealing the significant potential of low-frequency, low-intensity ultrasound in drug delivery and treatment of OA.
Collapse
Affiliation(s)
- Xiaoyu Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Yansong Tan
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300382, China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300382, China
| | - Lilan Gao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300382, China.
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300382, China.
| | - Hong Gao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China.
| |
Collapse
|
4
|
Chuang CF, Lin CW, Yeh CK. Ultrasound-triggered drug release and cytotoxicity of microbubbles with diverse drug attributes. ULTRASONICS SONOCHEMISTRY 2024; 112:107182. [PMID: 39631357 DOI: 10.1016/j.ultsonch.2024.107182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Ultrasound (US)-triggered cavitation of drug-loaded microbubbles (MBs) represents a promising approach for targeted drug delivery, with substantial benefits attainable through precise control over drug release dosage and form. This study investigates Camptothecin-loaded MBs (CPT-MBs) and Doxorubicin-loaded MBs (DOX-MBs), focusing on how properties such as hydrophilicity, hydrophobicity, and charged functional groups affect their interaction with the lipid surfaces of MBs, thereby influencing the fundamental characteristics and acoustic properties of the drug-loaded MBs. In comparison to DOX-MBs, CPT-MBs showed larger MB size (2.2 ± 0.3 and 1.4 ± 0.1 μm, respectively), a 2-fold increase in drug loading, and an 18 % reduction in leakage after 2 h at 37℃. Under 1 MHz US with a 100 ms pulse repetition interval (PRI), 1000 cycles, 5-minute duration, and 550 kPa acoustic pressure, CPT-MBs undergo inertial cavitation, while DOX-MBs undergo stable cavitation. Drug particles released from these MBs under US-induced cavitation were analyzed using dynamic light scattering, NanoSight, cryo-electron microscopy, and density gradient ultracentrifugation. Results showed that CPT-MBs mainly release free CPT, while DOX-MBs release multilayered DOX-lipid aggregates. The cytotoxicity to C6 cells induced by US-triggered cavitation of these two types of MBs also differed. DOX-lipid aggregates delayed initial uptake, leading to less pronounced short-term (2 h) effects compared to the rapid release of free CPT from CPT-MBs. These findings underscore the need to optimize drug delivery strategies by fine-tuning MB composition and US parameters to control drug release kinetics and achieve the best tumoricidal outcomes.
Collapse
Affiliation(s)
- Chi-Fen Chuang
- 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
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Fox J, Batchelor DVB, Coletta PL, Valleley EM, Evans SD. Microbubble Enhanced Delivery of Vitamin C for Treatment of Colorectal Cancer. ACS OMEGA 2024; 9:45270-45278. [PMID: 39554410 PMCID: PMC11561761 DOI: 10.1021/acsomega.4c06779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/19/2024]
Abstract
During chemotherapy treatment for cancer, often only a fraction of the administered dose reaches the tumor site, with the remaining drug spreading throughout the body, producing unwanted side-effects and restricting how much drug can be safely administered. A potential solution to reduce this problem is the use of microbubbles. The interaction between microbubbles and ultrasound generates pores in the tumor cells, permitting enhanced drug uptake. This study investigates the delivery of the ascorbic acid derivative, palmitoyl ascorbate, to KRAS-mutated colorectal cancer cells in vitro. Ultrasound-triggered microbubbles enhanced the efficacy of liposomal palmitoyl ascorbate treatments by 1.7- and 2.2-fold in LS174T and HCT116 CRC cell lines, respectively. This enhancement was achieved without increasing the drug dosage, and the therapeutic effect was shown to be localized to the area that received the ultrasound pulse, aiding in the reduction of off-site toxicity.
Collapse
Affiliation(s)
- Joseph Fox
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K.
| | - Damien V. B. Batchelor
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K.
| | - Patricia Louise Coletta
- Leeds
Institute of Medical Research, St James’s
University Hospital, Wellcome
Trust Brenner Building, Leeds LS9 7TF, U.K.
| | - Elizabeth M.A. Valleley
- Leeds
Institute of Medical Research, St James’s
University Hospital, Wellcome
Trust Brenner Building, Leeds LS9 7TF, U.K.
| | - Stephen D. Evans
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
6
|
Mur J, Reuter F, Agrež V, Petkovšek R, Ohl CD. Optic generation and perpetuation of acoustic bubble clusters. ULTRASONICS SONOCHEMISTRY 2024; 110:107023. [PMID: 39153420 PMCID: PMC11378257 DOI: 10.1016/j.ultsonch.2024.107023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Laser-induced cavitation bubbles offer precise control of the flow in space and time, but they are rarely used for the mechanical and chemical processing of liquids. Instead, strong acoustic fields are commonly used to nucleate and drive cavitation bubbles for liquid process applications. While acoustic field creates many more cavitation events, the resulting chaotic dynamics offers little control on the fluid mechanics, i.e., where and how bubbles deliver their energy. Here we present a method that utilizes a laser to nucleate a single cavitation bubble, which is then driven into violent oscillations by the ultrasound field, resulting in splitting of the bubble followed by formation of a cluster of cavitation bubbles. This combination offers means for cavitation control not available in conventional acoustic cavitation. Here, the cavitation bubble is generated with a custom build pulsed laser that is focused below a sonotrode driven at 20 kHz. In absence of the acoustic driving the bubble reaches a maximum diameter of 130 µm with a lifetime of approximately 10 µs. In the presence of the acoustic field the first few expansions and bubble collapses are strongly affected by the phase of nucleation. Over successive acoustic cycles a small bubble cluster develops that loses its connection with the phase of generation. We study the dynamics in the free field and constrained by a rigid boundary. For both geometries the cluster over many acoustic cycles dies off, yet through repetitive optical bubble seeding the cluster lifetime and its location can be controlled.
Collapse
Affiliation(s)
- Jaka Mur
- Faculty of Natural Sciences, Institute for Physics, Otto-von-Guericke-University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany; Faculty of Mechanical Engineering, University of Ljubljana, Aškerčeva 6, SI-1000 Ljubljana, Slovenia
| | - Fabian Reuter
- Faculty of Natural Sciences, Institute for Physics, Otto-von-Guericke-University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Vid Agrež
- Faculty of Mechanical Engineering, University of Ljubljana, Aškerčeva 6, SI-1000 Ljubljana, Slovenia
| | - Rok Petkovšek
- Faculty of Mechanical Engineering, University of Ljubljana, Aškerčeva 6, SI-1000 Ljubljana, Slovenia.
| | - Claus-Dieter Ohl
- Faculty of Natural Sciences, Institute for Physics, Otto-von-Guericke-University Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany.
| |
Collapse
|
7
|
Dauba A, Spitzlei C, Bautista KJB, Jourdain L, Selingue E, VanTreeck KE, Mattern JA, Denis C, Ouldali M, Arteni AA, Truillet C, Larrat B, Tsuruta J, Durham PG, Papadopoulou V, Dayton PA, Tsapis N, Novell A. Low-boiling-point perfluorocarbon nanodroplets for adaptable ultrasound-induced blood-brain barrier opening. J Control Release 2024; 376:441-456. [PMID: 39419451 DOI: 10.1016/j.jconrel.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Low-boiling point perfluorocarbon nanodroplets (NDs) are valued as effective sonosensitive agents, encapsulating a liquid perfluorocarbon that would instantaneously vaporize at body temperature without the NDs shell. Those NDs have been explored for both therapeutic and diagnostic purposes. Here, phospholipid-shelled nanodroplets containing octafluoropropane (C3F8) or decafluorobutane (C4F10) formed by condensation of microbubbles were thoroughly characterized before blood-brain (BBB) permeabilization. Transmission electron microscopy (TEM) and cryo-TEM were employed to confirm droplet formation while providing high-resolution insights into the droplet surface and lipid arrangement assessed from electron density observation after condensation. The vaporization threshold of NDs was determined with a high-speed camera, and the frequency signal emitted by the freshly vaporized bubbles was analyzed using cavitation detection. C3F8 NDs exhibited vaporization at 0.3 MPa (f0 = 1.5 MHz, 50 cycles), and emitted signals at 2 f0 and 1.5 f0 from 0.45 MPa onwards (f0 = 1.5 MHz, 50 cycles), while broadband noise was measured starting from 0.55 MPa. NDs with the higher boiling point C4F10 vaporized at 1.15 MPa and emitted signals at 2 f0 from 0.65 MPa and 1.5 f0 from 0.9 MPa, while broadband noise was detected starting from 0.95 MPa. Both ND formulations were used to permeabilize the BBB in healthy mice using tailored ultrasound sequences, allowing for the identification of optimal applications for each NDs type. C3F8 NDs proved suitable and safe for permeabilizing a large area, potentially the entire brain, at low acoustic pressure. Meanwhile, C4F10 droplets facilitated very localized (400 μm isotropic) permeabilization at higher pressure. This study prompts a closer examination of the structural rearrangements occurring during the condensation of microbubbles into NDs and highlights the potential to tailor solutions for different brain pathologies by choosing the composition of the NDs and adjusting the ultrasound sequence.
Collapse
Affiliation(s)
- Ambre Dauba
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Claire Spitzlei
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France; Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Kathlyne Jayne B Bautista
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Laurène Jourdain
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Erwan Selingue
- Université Paris-Saclay, CEA, CNRS, Baobab, NeuroSpin, Gif-sur-Yvette 91191, France
| | - Kelly E VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jacob A Mattern
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Caroline Denis
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Malika Ouldali
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Cryo-electron Microscopy Facility, CRYOEM-Gif, 91198 Gif-sur-Yvette, France
| | - Ana-Andreea Arteni
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Cryo-electron Microscopy Facility, CRYOEM-Gif, 91198 Gif-sur-Yvette, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, Baobab, NeuroSpin, Gif-sur-Yvette 91191, France
| | - James Tsuruta
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Phillip G Durham
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA; Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France.
| |
Collapse
|
8
|
Ishak O, Breton E, Cabras P, Dumont E, Mondou P, Novell A, Larrat B, Vappou J. Magnetic resonance cavitation imaging for the monitoring of ultrasound therapies. Phys Med Biol 2024; 69:215018. [PMID: 39378906 DOI: 10.1088/1361-6560/ad84b4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
Objective.Focused ultrasound (FUS) is a promising non-invasive therapeutic approach that can be used to generate thermal and non-thermal bioeffects. Several non-thermal FUS therapies rely on FUS-induced oscillations of microbubbles (MBs), a phenomenon referred to as cavitation. Cavitation monitoring in real time is essential to ensure both the efficacy and the safety of FUS therapies. This study aims to introduce a new magnetic resonance (MR) method for cavitation monitoring during FUS therapies.Approach.By finely synchronizing the FUS pulse with an accelerated turbo spin-echo MR sequence, the cavitation effect could be quantitatively estimated on the acquired images at 1-Hz refresh rate. The proposed method was assessed in vitro in a water bath. A series of FUS pulses were generated on a silicone tube filled with MBs at different acoustic pressures (0.07-2.07 MPa) and pulse durations (20-2000μs). MR images and passive cavitation detection (PCD) signals were simultaneously acquired for each FUS pulse.Main results.Inertial cavitation was found to induce a quantitatively interpretable signal loss on the MR image. The transition from stable to inertial cavitation was identified on MR cavitation maps with high repeatability. These results were found to be in good agreement with PCD measurements in terms of pressure thresholds between stable and inertial cavitation. MR cavitation imaging was shown to be sensitive to short and even ultrashort FUS pulses, from 2 ms down to 20μs. The presented theoretical model suggests that the signal loss in MR cavitation imaging relies on susceptibility changes related to the diameter of the oscillating MBs.Significance.The proposed MR cavitation imaging method can both locate and characterize cavitation activity. It has therefore the potential to improve the efficacy and safety of FUS therapies, particularly for localized drug delivery applications.
Collapse
Affiliation(s)
- Ounay Ishak
- Université de Strasbourg, CNRS, INSERM, ICube, UMR7357, Strasbourg, France
| | - Elodie Breton
- Université de Strasbourg, CNRS, INSERM, ICube, UMR7357, Strasbourg, France
| | - Paolo Cabras
- Université de Strasbourg, CNRS, INSERM, ICube, UMR7357, Strasbourg, France
- Image Guided Therapy, Pessac, France
| | | | - Paul Mondou
- Université de Strasbourg, CNRS, INSERM, ICube, UMR7357, Strasbourg, France
- Université Paris-Saclay, CEA, CNRS, BAOBAB, Neurospin, 91191 Gif-sur-Yvette, France
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, INSERM, BioMaps, SHFJ, 91401 Orsay, France
| | - Benoît Larrat
- Université Paris-Saclay, CEA, CNRS, BAOBAB, Neurospin, 91191 Gif-sur-Yvette, France
| | - Jonathan Vappou
- Université de Strasbourg, CNRS, INSERM, ICube, UMR7357, Strasbourg, France
| |
Collapse
|
9
|
Shen L, Tian Z, Yang K, Rich J, Xia J, Upreti N, Zhang J, Chen C, Hao N, Pei Z, Huang TJ. Joint subarray acoustic tweezers enable controllable cell translation, rotation, and deformation. Nat Commun 2024; 15:9059. [PMID: 39428395 PMCID: PMC11491459 DOI: 10.1038/s41467-024-52686-8] [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: 02/26/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Contactless microscale tweezers are highly effective tools for manipulating, patterning, and assembling bioparticles. However, current tweezers are limited in their ability to comprehensively manipulate bioparticles, providing only partial control over the six fundamental motions (three translational and three rotational motions). This study presents a joint subarray acoustic tweezers platform that leverages acoustic radiation force and viscous torque to control the six fundamental motions of single bioparticles. This breakthrough is significant as our manipulation mechanism allows for controlling the three translational and three rotational motions of single cells, as well as enabling complex manipulation that combines controlled translational and rotational motions. Moreover, our tweezers can gradually increase the load on an acoustically trapped cell to achieve controllable cell deformation critical for characterizing cell mechanical properties. Furthermore, our platform allows for three-dimensional (3D) imaging of bioparticles without using complex confocal microscopy by rotating bioparticles with acoustic tweezers and taking images of each orientation using a standard microscope. With these capabilities, we anticipate the JSAT platform to play a pivotal role in various applications, including 3D imaging, tissue engineering, disease diagnostics, and drug testing.
Collapse
Affiliation(s)
- Liang Shen
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
- Department of Mechanical Engineering, Virginia Polytechnical Institute and State University, Blacksburg, VA, USA
| | - Zhenhua Tian
- Department of Mechanical Engineering, Virginia Polytechnical Institute and State University, Blacksburg, VA, USA.
| | - Kaichun Yang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Joseph Rich
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jianping Xia
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Neil Upreti
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Jinxin Zhang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Chuyi Chen
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Nanjing Hao
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Zhichao Pei
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA.
| |
Collapse
|
10
|
Liu JD, VanTreeck KE, Marston WA, Papadopoulou V, Rowe SE. Ultrasound-Mediated Antibiotic Delivery to In Vivo Biofilm Infections: A Review. Chembiochem 2024; 25:e202400181. [PMID: 38924307 PMCID: PMC11483220 DOI: 10.1002/cbic.202400181] [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: 02/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Bacterial biofilms are a significant concern in various medical contexts due to their resilience to our immune system as well as antibiotic therapy. Biofilms often require surgical removal and frequently lead to recurrent or chronic infections. Therefore, there is an urgent need for improved strategies to treat biofilm infections. Ultrasound-mediated drug delivery is a technique that combines ultrasound application, often with the administration of acoustically-active agents, to enhance drug delivery to specific target tissues or cells within the body. This method involves using ultrasound waves to assist in the transportation or activation of medications, improving their penetration, distribution, and efficacy at the desired site. The advantages of ultrasound-mediated drug delivery include targeted and localized delivery, reduced systemic side effects, and improved efficacy of the drug at lower doses. This review scrutinizes recent advances in the application of ultrasound-mediated drug delivery for treating biofilm infections, focusing on in vivo studies. We examine the strengths and limitations of this technology in the context of wound infections, device-associated infections, lung infections and abscesses, and discuss current gaps in knowledge and clinical translation considerations.
Collapse
Affiliation(s)
- Jamie D. Liu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kelly E. VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William A. Marston
- Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Department of Radiology, The University of North Carolina at Chapel Hill, NC, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
11
|
Moosavifar M, Barmin RA, Rama E, Rix A, Gumerov RA, Lisson T, Bastard C, Rütten S, Avraham‐Radermacher N, Koehler J, Pohl M, Kulkarni V, Baier J, Koletnik S, Zhang R, Dasgupta A, Motta A, Weiler M, Potemkin II, Schmitz G, Kiessling F, Lammers T, Pallares RM. Polymeric Microbubble Shell Engineering: Microporosity as a Key Factor to Enhance Ultrasound Imaging and Drug Delivery Performance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404385. [PMID: 39207095 PMCID: PMC11516050 DOI: 10.1002/advs.202404385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/15/2024] [Indexed: 09/04/2024]
Abstract
Microbubbles (MB) are widely used as contrast agents for ultrasound (US) imaging and US-enhanced drug delivery. Polymeric MB are highly suitable for these applications because of their acoustic responsiveness, high drug loading capability, and ease of surface functionalization. While many studies have focused on using polymeric MB for diagnostic and therapeutic purposes, relatively little attention has thus far been paid to improving their inherent imaging and drug delivery features. This study here shows that manipulating the polymer chemistry of poly(butyl cyanoacrylate) (PBCA) MB via temporarily mixing the monomer with the monomer-mimetic butyl cyanoacetate (BCC) during the polymerization process improves the drug loading capacity of PBCA MB by more than twofold, and the in vitro and in vivo acoustic responses of PBCA MB by more than tenfold. Computer simulations and physisorption experiments show that BCC manipulates the growth of PBCA polymer chains and creates nanocavities in the MB shell, endowing PBCA MB with greater drug entrapment capability and stronger acoustic properties. Notably, because BCC can be readily and completely removed during MB purification, the resulting formulation does not include any residual reagent beyond the ones already present in current PBCA-based MB products, facilitating the potential translation of next-generation PBCA MB.
Collapse
Affiliation(s)
- Mirjavad Moosavifar
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Roman A. Barmin
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Elena Rama
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anne Rix
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Rustam A. Gumerov
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Thomas Lisson
- Chair for Medical EngineeringRuhr University Bochum44780BochumGermany
| | - Céline Bastard
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Stephan Rütten
- Electron Microscope FacilityRWTH Aachen University Hospital52074AachenGermany
| | - Noah Avraham‐Radermacher
- Institute of Technical and Macromolecular ChemistryRWTH Aachen University Hospital52074AachenGermany
| | - Jens Koehler
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Michael Pohl
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Vedangi Kulkarni
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Jasmin Baier
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Susanne Koletnik
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Rui Zhang
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Alessandro Motta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Marek Weiler
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Igor I. Potemkin
- DWI – Leibniz Institute for Interactive MaterialsRWTH Aachen University52074AachenGermany
| | - Georg Schmitz
- Chair for Medical EngineeringRuhr University Bochum44780BochumGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Roger M. Pallares
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| |
Collapse
|
12
|
Singh D, Memari E, He S, Yusefi H, Helfield B. Cardiac gene delivery using ultrasound: State of the field. Mol Ther Methods Clin Dev 2024; 32:101277. [PMID: 38983873 PMCID: PMC11231612 DOI: 10.1016/j.omtm.2024.101277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Over the past two decades, there has been tremendous and exciting progress toward extending the use of medical ultrasound beyond a traditional imaging tool. Ultrasound contrast agents, typically used for improved visualization of blood flow, have been explored as novel non-viral gene delivery vectors for cardiovascular therapy. Given this adaptation to ultrasound contrast-enhancing agents, this presents as an image-guided and site-specific gene delivery technique with potential for multi-gene and repeatable delivery protocols-overcoming some of the limitations of alternative gene therapy approaches. In this review, we provide an overview of the studies to date that employ this technique toward cardiac gene therapy using cardiovascular disease animal models and summarize their key findings.
Collapse
Affiliation(s)
- Davindra Singh
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Elahe Memari
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Hossein Yusefi
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Brandon Helfield
- Department of Biology, Concordia University, Montreal, QC, Canada
- Department of Physics, Concordia University, Montreal, QC, Canada
| |
Collapse
|
13
|
Tu B, Li Y, Wen W, Liu J. Bibliometric and visualized analysis of ultrasound combined with microbubble therapy technology from 2009 to 2023. Front Pharmacol 2024; 15:1418142. [PMID: 39119614 PMCID: PMC11306066 DOI: 10.3389/fphar.2024.1418142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
Background In recent years, with the rapid advancement of fundamental ultrasonography research, the application of ultrasound in disease treatment has progressively increased. An increasing body of research indicates that microbubbles serve not only as contrast agents but also in conjunction with ultrasound, enhancing cavitation effects and facilitating targeted drug delivery, thereby augmenting therapeutic efficacy. The objective of this study is to explore the current status and prevailing research trends in this field from 2009 to 2023 through bibliometric analysis and to forecast future developmental trajectories. Methods We selected the Science Citation Index Expanded (SCI-Expanded) from the Web of Science Core Collection (WOSCC) as our primary data source. On 19 January 2024, we conducted a comprehensive search encompassing all articles and reviews published between 2009 and 2023 and utilized the bibliometric online analysis platform, CiteSpace and VOSviewer software to analyze countries/regions, institutions, authors, keywords, and references, used Microsoft Excel 2021 to visualize the trends of the number of articles published by year. Results Between 1 January 2009, and 31 December 2023, 3,326 publications on ultrasound combined with microbubble therapy technology were included. There were a total of 2,846 articles (85.6%) and 480 reviews (14.4%) from 13,062 scholars in 68 countries/regions published in 782 journals. China and the United States emerged as the primary contributors in this domain. In terms of publication output and global institutional collaboration, the University of Toronto in Canada has made the most significant contribution to this field. Professor Kullervo Hynynen has achieved remarkable accomplishments in this area. Ultrasound in Medicine and Biology is at the core of the publishing of research on ultrasound combined with microbubble therapy technology. Keywords such as "sonodynamic therapy," "oxygen," "loaded microbubbles" and "Alzheimer's disease" indicate emerging trends in the field and hold the potential to evolve into significant areas of future investigation. Conclusion This study provides a summary of the key contributions of ultrasound combined with microbubble therapy to the field's development over the past 15 years and delves into the historical underpinnings and contemporary trends of ultrasound combined with microbubble therapy technology, providing valuable guidance for researchers.
Collapse
Affiliation(s)
- Bin Tu
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yan Li
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wen Wen
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jian Liu
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
14
|
Jaroszewski B, Jelonek K, Kasperczyk J. Drug Delivery Systems of Betulin and Its Derivatives: An Overview. Biomedicines 2024; 12:1168. [PMID: 38927375 PMCID: PMC11200571 DOI: 10.3390/biomedicines12061168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Natural origin products are regarded as promising for the development of new therapeutic therapies with improved effectiveness, biocompatibility, reduced side effects, and low cost of production. Betulin (BE) is very promising due to its wide range of pharmacological activities, including its anticancer, antioxidant, and antimicrobial properties. However, despite advancements in the use of triterpenes for clinical purposes, there are still some obstacles that hinder their full potential, such as their hydrophobicity, low solubility, and poor bioavailability. To address these concerns, new BE derivatives have been synthesized. Moreover, drug delivery systems have emerged as a promising solution to overcome the barriers faced in the clinical application of natural products. The aim of this manuscript is to summarize the recent achievements in the field of delivery systems of BE and its derivatives. This review also presents the BE derivatives mostly considered for medical applications. The electronic databases of scientific publications were searched for the most interesting achievements in the last ten years. Thus far, it is mostly nanoparticles (NPs) that have been considered for the delivery of betulin and its derivatives, including organic NPs (e.g., micelles, conjugates, liposomes, cyclodextrins, protein NPs), inorganic NPs (carbon nanotubes, gold NPs, silver), and complex/hybrid and miscellaneous nanoparticulate systems. However, there are also examples of microparticles, gel-based systems, suspensions, emulsions, and scaffolds, which seem promising for the delivery of BE and its derivatives.
Collapse
Affiliation(s)
- Bartosz Jaroszewski
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland;
| | - Katarzyna Jelonek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Curie-Skłodowska 34 St., 41-819 Zabrze, Poland
| | - Janusz Kasperczyk
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland;
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Curie-Skłodowska 34 St., 41-819 Zabrze, Poland
| |
Collapse
|
15
|
Chen J, Wang B, Wang Y, Radermacher H, Qi J, Momoh J, Lammers T, Shi Y, Rix A, Kiessling F. mRNA Sonotransfection of Tumors with Polymeric Microbubbles: Co-Formulation versus Co-Administration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306139. [PMID: 38342634 PMCID: PMC11022722 DOI: 10.1002/advs.202306139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/24/2024] [Indexed: 02/13/2024]
Abstract
Despite its high potential, non-viral gene therapy of cancer remains challenging due to inefficient nucleic acid delivery. Ultrasound (US) with microbubbles (MB) can open biological barriers and thus improve DNA and mRNA passage. Polymeric MB are an interesting alternative to clinically used lipid-coated MB because of their high stability, narrow size distribution, and easy functionalization. However, besides choosing the ideal MB, it remains unclear whether nanocarrier-encapsulated mRNA should be administered separately (co-administration) or conjugated to MB (co-formulation). Therefore, the impact of poly(n-butyl cyanoacrylate) MB co-administration with mRNA-DOTAP/DOPE lipoplexes or their co-formulation on the transfection of cancer cells in vitro and in vivo is analyzed. Sonotransfection improved mRNA delivery into 4T1 breast cancer cells in vitro with co-administration being more efficient than co-formulation. In vivo, the co-administration sonotransfection approach also resulted in higher transfection efficiency and reached deeper into the tumor tissue. On the contrary, co-formulation mainly promoted transfection of endothelial and perivascular cells. Furthermore, the co-formulation approach is much more dependent on the US trigger, resulting in significantly lower off-site transfection. Thus, the findings indicate that the choice of co-administration or co-formulation in sonotransfection should depend on the targeted cell population, tolerable off-site transfection, and the therapeutic purpose.
Collapse
Affiliation(s)
- Junlin Chen
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Bi Wang
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Yuchen Wang
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Harald Radermacher
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Jinwei Qi
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Jeffrey Momoh
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Yang Shi
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Anne Rix
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| |
Collapse
|
16
|
Gusliakova OI, Kurochkin MA, Barmin RA, Prikhozhdenko ES, Estifeeva TM, Rudakovskaya PG, Sindeeva OA, Galushka VV, Vavaev ES, Komlev AS, Lyubin EV, Fedyanin AA, Dey KK, Gorin DA. Magnetically navigated microbubbles coated with albumin/polyarginine and superparamagnetic iron oxide nanoparticles. BIOMATERIALS ADVANCES 2024; 158:213759. [PMID: 38227987 DOI: 10.1016/j.bioadv.2024.213759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
While microbubbles (MB) are routinely used for ultrasound (US) imaging, magnetic MB are increasingly explored as they can be guided to specific sites of interest by applied magnetic field gradient. This requires the MB shell composition tuning to prolong MB stability and provide functionalization capabilities with magnetic nanoparticles. Hence, we developed air-filled MB stabilized by a protein-polymer complex of bovine serum albumin (BSA) and poly-L-arginine (pArg) of different molecular weights, showing that pArg of moderate molecular weight distribution (15-70 kDa) enabled MB with greater stability and acoustic response while preserving MB narrow diameters and the relative viability of THP-1 cells after 48 h of incubation. After MB functionalization with superparamagnetic iron oxide nanoparticles (SPION), magnetic moment values provided by single MB confirmed the sufficient SPION deposition onto BSA + pArg MB shells. During MB magnetic navigation in a blood vessel mimicking phantom with magnetic tweezers and in a Petri dish with adherent mouse renal carcinoma cell line, we demonstrated the effectiveness of magnetic MB localization in the desired area by magnetic field gradient. Magnetic MB co-localization with cells was further exploited for effective doxorubicin delivery with drug-loaded MB. Taken together, these findings open new avenues in control over albumin MB properties and magnetic navigation of SPION-loaded MB, which can envisage their applications in diagnostic and therapeutic needs.
Collapse
Affiliation(s)
- Olga I Gusliakova
- Science Medical Center, Saratov State University, Saratov 410012, Russia; Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| | - Maxim A Kurochkin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Roman A Barmin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | | | - Tatyana M Estifeeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Polina G Rudakovskaya
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Olga A Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Victor V Galushka
- Education and Research Institute of Nanostructures and Biosystems, Saratov State University, Saratov 410012, Russia
| | - Evgeny S Vavaev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Aleksei S Komlev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Evgeny V Lyubin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Andrey A Fedyanin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Krishna Kanti Dey
- Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382055, India
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| |
Collapse
|
17
|
Dash N, Tamadapu G. Nonspherical oscillations of an encapsulated microbubble with interface energy under the acoustic field. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2024; 155:2445-2459. [PMID: 38573054 DOI: 10.1121/10.0025390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
Spherical instability in acoustically driven encapsulated microbubbles (EBs) suspended in a fluid can trigger nonspherical oscillations within them. We apply the interface energy model [N. Dash and G. Tamadapu, J. Fluid Mech. 932, A26 (2022b)] to investigate nonspherical oscillations of smaller radius microbubbles encapsulated with a viscoelastic shell membrane under acoustic field. Using the Lagrangian energy formulation, coupled governing equations for spherical and nonspherical modes are derived, incorporating interface energy effects, shell elasticity, and viscosity. Numerical simulations of governing equations revealed that the parametrically forced even mode excites even modes, while the odd modes excite both even and odd modes. The model demonstrates that finite amplitude nonspherical oscillations are identifiable in smaller radius EBs only when the interface parameters are introduced into the model; otherwise, they are not. Realizing that nonlinear mode coupling is responsible for saturation of instability resulting in stable nonspherical oscillations, we perform a steady-state and stability analysis using the slow-time equations obtained from Krylov-Bogoliubov perturbation method. Analytical expressions for modal amplitudes and stability thresholds are derived in terms of interface and material parameters. The stability curves are invaluable in determining the precise range of excitation pressure and frequency values required for the EB to exhibit finite amplitude nonspherical oscillations.
Collapse
Affiliation(s)
- Nehal Dash
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai 600036, India
| | - Ganesh Tamadapu
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
18
|
Nittayacharn P, Abenojar E, Cooley MB, Berg FM, Counil C, Sojahrood AJ, Khan MS, Yang C, Berndl E, Golczak M, Kolios MC, Exner AA. Efficient ultrasound-mediated drug delivery to orthotopic liver tumors - Direct comparison of doxorubicin-loaded nanobubbles and microbubbles. J Control Release 2024; 367:135-147. [PMID: 38237687 PMCID: PMC11700473 DOI: 10.1016/j.jconrel.2024.01.028] [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: 09/14/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Liver metastasis is a major obstacle in treating aggressive cancers, and current therapeutic options often prove insufficient. To overcome these challenges, there has been growing interest in ultrasound-mediated drug delivery using lipid-shelled microbubbles (MBs) and nanobubbles (NBs) as promising strategies for enhancing drug delivery to tumors. Our previous work demonstrated the potential of Doxorubicin-loaded C3F8 NBs (hDox-NB, 280 ± 123 nm) in improving cancer treatment in vitro using low-frequency unfocused therapeutic ultrasound (TUS). In this study, we investigated the pharmacokinetics and biodistribution of sonicated hDox-NBs in orthotopic rat liver tumors. We compared their delivery and therapeutic efficiency with size-isolated MBs (hDox-MB, 1104 ± 373 nm) made from identical shell material and core gas. Results showed a similar accumulation of hDox in tumors treated with hDox-MBs and unfocused therapeutic ultrasound (hDox-MB + TUS) and hDox-NB + TUS. However, significantly increased apoptotic cell death in the tumor and fewer off-target apoptotic cells in the normal liver were found upon the treatment with hDox-NB + TUS. The tumor-to-liver apoptotic ratio was elevated 9.4-fold following treatment with hDox-NB + TUS compared to hDox-MB + TUS, suggesting that the therapeutic efficacy and specificity are significantly increased when using hDox-NB + TUS. These findings highlight the potential of this approach as a viable treatment modality for liver tumors. By elucidating the behavior of drug-loaded bubbles in vivo, we aim to contribute to developing more effective liver cancer treatments that could ultimately improve patient outcomes and decrease off-target side effects.
Collapse
Affiliation(s)
- Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Puttamonthon, Nakorn Pathom, Thailand
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Felipe M Berg
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Claire Counil
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Amin Jafari Sojahrood
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Muhammad Saad Khan
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Celina Yang
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Elizabeth Berndl
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
19
|
Bouakaz A, Michel Escoffre J. From concept to early clinical trials: 30 years of microbubble-based ultrasound-mediated drug delivery research. Adv Drug Deliv Rev 2024; 206:115199. [PMID: 38325561 DOI: 10.1016/j.addr.2024.115199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Ultrasound mediated drug delivery, a promising therapeutic modality, has evolved remarkably over the past three decades. Initially designed to enhance contrast in ultrasound imaging, microbubbles have emerged as a main vector for drug delivery, offering targeted therapy with minimized side effects. This review addresses the historical progression of this technology, emphasizing the pivotal role microbubbles play in augmenting drug extravasation and targeted delivery. We explore the complex mechanisms behind this technology, from stable and inertial cavitation to diverse acoustic phenomena, and their applications in medical fields. While the potential of ultrasound mediated drug delivery is undeniable, there are still challenges to overcome. Balancing therapeutic efficacy and safety and establishing standardized procedures are essential areas requiring attention. A multidisciplinary approach, gathering collaborations between researchers, engineers, and clinicians, is important for exploiting the full potential of this technology. In summary, this review highlights the potential of using ultrasound mediated drug delivery in improving patient care across various medical conditions.
Collapse
Affiliation(s)
- Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | | |
Collapse
|
20
|
Wang P, Tan C, Ji X, Bai J, Yu ACH, Qin P. Sensitivity improvement of subharmonic-based pressure measurement using phospholipid-coated monodisperse microbubbles. ULTRASONICS SONOCHEMISTRY 2024; 104:106830. [PMID: 38432151 PMCID: PMC10920959 DOI: 10.1016/j.ultsonch.2024.106830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
The use of the subharmonic signal from microbubbles exposed to ultrasound is a promising safe and cost-effective approach for the non-invasive measurement of blood pressure. Achieving a high sensitivity of the subharmonic amplitude to the ambient overpressure is crucial for clinical applications. However, currently used microbubbles have a wide size distribution and diverse shell properties. This causes uncertainty in the response of the subharmonic amplitude to changes in ambient pressure, which limits the sensitivity. The aim of this study was to use monodisperse microbubbles to improve the sensitivity of subharmonic-based pressure measurements. With the same shell materials and gas core, we used a flow-focusing microfluidic chip and a mechanical agitation method to fabricate monodisperse (∼2.45-µm mean radius and 4.7 % polydisperse index) and polydisperse microbubbles (∼1.51-µm mean radius and 48.4 % polydisperse index), respectively. We varied the ultrasound parameters (i.e., the frequency, peak negative pressure (PNP) and pulse length), and found that there was an optimal excitation frequency (2.8 MHz) for achieving maximal subharmonic emission for monodisperse microbubbles, but not for polydisperse microbubbles. Three distinct regimes (occurrence, growth, and saturation) were identified in the response of the subharmonic amplitude to increasing PNP for both monodisperse and polydisperse microbubbles. For the polydisperse microbubbles, the subharmonic amplitude decreased either monotonically or non-monotonically with ambient overpressure, depending on the PNP. By contrast, for the monodisperse microbubbles, there was only a monotonic decrease at all PNPs. The maximum sensitivity (1.18 dB/kPa, R2 = 0.97) of the subharmonic amplitude to ambient overpressure for the monodisperse microbubbles was ∼6.5 times higher than that for the polydisperse microbubbles (0.18 dB/kPa, R2 = 0.88). These results show that monodisperse microbubbles can achieve a more consistent response of the subharmonic signal to changes in ambient overpressure and greatly improve the measurement sensitivity.
Collapse
Affiliation(s)
- Pengcheng Wang
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chunjie Tan
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiang Ji
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jingfeng Bai
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, N2L3G1, Canada
| | - Peng Qin
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
21
|
Fernandez JL, Snipstad S, Bjørkøy A, Davies CDL. Real-Time Multiphoton Intravital Microscopy of Drug Extravasation in Tumours during Acoustic Cluster Therapy. Cells 2024; 13:349. [PMID: 38391962 PMCID: PMC10887035 DOI: 10.3390/cells13040349] [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: 12/29/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Optimising drug delivery to tumours remains an obstacle to effective cancer treatment. A prerequisite for successful chemotherapy is that the drugs reach all tumour cells. The vascular network of tumours, extravasation across the capillary wall and penetration throughout the extracellular matrix limit the delivery of drugs. Ultrasound combined with microbubbles has been shown to improve the therapeutic response in preclinical and clinical studies. Most studies apply microbubbles designed as ultrasound contrast agents. Acoustic Cluster Therapy (ACT®) is a novel approach based on ultrasound-activated microbubbles, which have a diameter 5-10 times larger than regular contrast agent microbubbles. An advantage of using such large microbubbles is that they are in contact with a larger part of the capillary wall, and the oscillating microbubbles exert more effective biomechanical effects on the vessel wall. In accordance with this, ACT® has shown promising therapeutic results in combination with various drugs and drug-loaded nanoparticles. Knowledge of the mechanism and behaviour of drugs and microbubbles is needed to optimise ACT®. Real-time intravital microscopy (IVM) is a useful tool for such studies. This paper presents the experimental setup design for visualising ACT® microbubbles within the vasculature of tumours implanted in dorsal window (DW) chambers. It presents ultrasound setups, the integration and alignment of the ultrasound field with the optical system in live animal experiments, and the methodologies for visualisation and analysing the recordings. Dextran was used as a fluorescent marker to visualise the blood vessels and to trace drug extravasation and penetration into the extracellular matrix. The results reveal that the experimental setup successfully recorded the kinetics of extravasation and penetration distances into the extracellular matrix, offering a deeper understanding of ACT's mechanisms and potential in localised drug delivery.
Collapse
Affiliation(s)
- Jessica Lage Fernandez
- Department of Physics, Norwegian University of Science and Technology, 7034 Trondheim, Norway; (S.S.); (A.B.); (C.d.L.D.)
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, 7034 Trondheim, Norway; (S.S.); (A.B.); (C.d.L.D.)
- Cancer Clinic, St. Olavs Hospital, 7030 Trondheim, Norway
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, 7034 Trondheim, Norway; (S.S.); (A.B.); (C.d.L.D.)
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology, 7034 Trondheim, Norway; (S.S.); (A.B.); (C.d.L.D.)
| |
Collapse
|
22
|
Pellow C, Jafari Sojahrood A, Zhao X, Kolios MC, Exner AA, Goertz DE. Synchronous Intravital Imaging and Cavitation Monitoring of Antivascular Focused Ultrasound in Tumor Microvasculature Using Monodisperse Low Boiling Point Nanodroplets. ACS NANO 2024; 18:410-427. [PMID: 38147452 PMCID: PMC10786165 DOI: 10.1021/acsnano.3c07711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.
Collapse
Affiliation(s)
- Carly Pellow
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
| | - Amin Jafari Sojahrood
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Xiaoxiao Zhao
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| | - Michael C. Kolios
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Agata A. Exner
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - David E. Goertz
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| |
Collapse
|
23
|
Barmin RA, Dasgupta A, Rix A, Weiler M, Appold L, Rütten S, Padilla F, Kuehne AJC, Pich A, De Laporte L, Kiessling F, Pallares RM, Lammers T. Enhanced Stable Cavitation and Nonlinear Acoustic Properties of Poly(butyl cyanoacrylate) Polymeric Microbubbles after Bioconjugation. ACS Biomater Sci Eng 2024; 10:75-81. [PMID: 36315422 DOI: 10.1021/acsbiomaterials.2c01021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Microbubbles (MB) are used as ultrasound (US) contrast agents in clinical settings because of their ability to oscillate upon exposure to acoustic pulses and generate nonlinear responses with a stable cavitation profile. Polymeric MB have recently attracted increasing attention as molecular imaging probes and drug delivery agents based on their tailorable acoustic responses, high drug loading capacity, and surface functionalization capabilities. While many of these applications require MB to be functionalized with biological ligands, the impact of bioconjugation on polymeric MB cavitation and acoustic properties remains poorly understood. Hence, we here evaluated the effects of MB shell hydrolysis and subsequent streptavidin conjugation on the acoustic behavior of poly(butyl cyanoacrylate) (PBCA) MB. We show that upon biofunctionalization, MB display higher acoustic stability, stronger stable cavitation, and enhanced second harmonic generation. Furthermore, functionalized MB preserve the binding capabilities of streptavidin conjugated on their surface. These findings provide insights into the effects of bioconjugation chemistry on polymeric MB acoustic properties, and they contribute to improving the performance of polymer-based US imaging and theranostic agents.
Collapse
Affiliation(s)
- Roman A Barmin
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Marek Weiler
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Lia Appold
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Stephan Rütten
- Electron Microscope Facility, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Frederic Padilla
- Focused Ultrasound Foundation, Charlottesville, Virginia 22903, United States
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ-Lyon, Lyon F-69003, France
- Department of Radiology, University of Virginia, Charlottesville, Virginia 22903, United States
| | - Alexander J C Kuehne
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Andrij Pich
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University Hospital, Aachen 52074, Germany
- Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen 52074, Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus, 6167 RD Geleen, The Netherlands
| | - Laura De Laporte
- DWI - Leibniz Institute for Interactive Materials, RWTH Aachen University Hospital, Aachen 52074, Germany
- Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, Aachen 52074, Germany
- Institute of Applied Medical Engineering, Department of Advanced Materials for Biomedicine, RWTH Aachen University, Aachen 52074, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen 52074, Germany
| |
Collapse
|
24
|
Li P, Du P, Peng J, Zhao Z, Li H, Yu W, Wang S, Liu L. Pharmacokinetics and pharmacodynamics of perfluoropropane after intra-venous bolus injection of perflutren lipid microsphere injection (DEFINITY®) in healthy Chinese volunteers. BMC Pharmacol Toxicol 2024; 25:6. [PMID: 38167238 PMCID: PMC10763448 DOI: 10.1186/s40360-023-00729-z] [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: 08/23/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Definity is an ultrasound contrast agent consisting of phospholipids-encapsulated perfluoropropane (PFP), also known as perflutren, microspheres, which is initially designed to enhance echocardiographic ultrasound images. With no pharmacologic action, Definity can increase the backscatter of ultrasound resulting enhanced ultrasound signals. The objective of this study was to determine the pharmacokinetics (PKs), Pharmacodynamics (PDs) and safety of Definity in healthy male and female Chinese volunteers. METHODS A simple GC-MS method was developed and applied to simultaneously quantify PFP both in human whole blood and in expired air using Perfluorobutane (PFB) as internal standard. Meanwhile, the blood microbubble Doppler intensities were continuously monitored as companion PDs by a Doppler ultrasonography system using a non-imaging method. RESULTS After intravenous infusion of 10 µL/kg of PFP within 30 seconds, the mean AUClast of the pharmacokinetic analysis set was 0.000653 (uL/mL)*min, the average AUC∞ was 0.001051 (uL/mL)*min. The main coefficient of variation of parameters were within 30%. In this trial, the blood drug concentration of female subjects was lower than that of males. Female Cmax, AUClast and AUC∞ were lower than males', Tmax and t1/2 was close to males', Vss and CL were slightly higher than males'. The concentration of PFP in the expired air of the subject reached the maximum value 1-2 min after administration and the PFP accumulation curve in the expired air began to become flat at 9.5-11 min after administration. The PFP in the expired air at the last sampling point of most subjects was still measurable. The results of the analysis showed that female subjects had slightly more and faster PFP excretion via the lungs than males. The change of blood drug concentration in this trial was related to the change process of Doppler signal intensity. The trend of the two was close, but the peak time of blood drug concentration was slightly delayed compared with the peak time of the Doppler signal intensity. The results showed that female tmax-pd, t10 was earlier than male, and women have lower AUCpd than men. CONCLUSION The pharmacokinetics and pharmacodynamics of Definity in blood and expired air were systematically evaluated for the first time in this study. The PK/PD analysis results of this trial showed that the change of blood concentration was related to the change process of Doppler signal intensity, the trend of the two was close and expired air are the main excretion pathways of Definity. Definity was well tolerated by all subjects in the trial. TRIAL REGISTRATION This study was registered on 08 December 2017 at the Chinese Clinical Trial Registry (CTR20171087).
Collapse
Affiliation(s)
- Pengfei Li
- Department of Pharmacy, Beijing An Ding Hospital, Capital Medical University, No.5 Ankang Hutong, Xicheng District, Beijing, 100088, China.
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Ping Du
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Jun Peng
- Guoke Excellence (Beijing) Medicine Technology Research Co., Ltd, No.18 Zhonghe street, Daxing District, Beijing, 100176, China
| | - Zhixia Zhao
- Department of Pharmacy, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
| | - Huiling Li
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Weiyue Yu
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China
| | - Shumin Wang
- Phase I Clinical Trial Unit, Beijing Chao-Yang Hospital, Capital Medical University, No.8 Gongti South Road, Chaoyang District, Beijing, 100020, China.
| | - Lihong Liu
- Department of Pharmacy, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
25
|
Wang HC, Phan TN, Kao CL, Yeh CK, Lin YC. Genetically encoded mediators for sonogenetics and their applications in neuromodulation. Front Cell Neurosci 2023; 17:1326279. [PMID: 38188668 PMCID: PMC10766825 DOI: 10.3389/fncel.2023.1326279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 01/09/2024] Open
Abstract
Sonogenetics is an emerging approach that harnesses ultrasound for the manipulation of genetically modified cells. The great penetrability of ultrasound waves enables the non-invasive application of external stimuli to deep tissues, particularly advantageous for brain stimulation. Genetically encoded ultrasound mediators, a set of proteins that respond to ultrasound-induced bio-effects, play a critical role in determining the effectiveness and applications of sonogenetics. In this context, we will provide an overview of these ultrasound-responsive mediators, delve into the molecular mechanisms governing their response to ultrasound stimulation, and summarize their applications in neuromodulation.
Collapse
Affiliation(s)
- Hsien-Chu Wang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Thi-Nhan Phan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chi-Ling Kao
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
26
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
27
|
Barmin RA, Moosavifar M, Dasgupta A, Herrmann A, Kiessling F, Pallares RM, Lammers T. Polymeric materials for ultrasound imaging and therapy. Chem Sci 2023; 14:11941-11954. [PMID: 37969594 PMCID: PMC10631124 DOI: 10.1039/d3sc04339h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/11/2023] [Indexed: 11/17/2023] Open
Abstract
Ultrasound (US) is routinely used for diagnostic imaging and increasingly employed for therapeutic applications. Materials that act as cavitation nuclei can improve the resolution of US imaging, and facilitate therapeutic US procedures by promoting local drug delivery or allowing temporary biological barrier opening at moderate acoustic powers. Polymeric materials offer a high degree of control over physicochemical features concerning responsiveness to US, e.g. via tuning chain composition, length and rigidity. This level of control cannot be achieved by materials made of lipids or proteins. In this perspective, we present key engineered polymeric materials that respond to US, including microbubbles, gas-stabilizing nanocups, microcapsules and gas-releasing nanoparticles, and discuss their formulation aspects as well as their principles of US responsiveness. Focusing on microbubbles as the most common US-responsive polymeric materials, we further evaluate the available chemical toolbox to engineer polymer shell properties and enhance their performance in US imaging and US-mediated drug delivery. Additionally, we summarize emerging applications of polymeric microbubbles in molecular imaging, sonopermeation, and gas and drug delivery, based on refinement of MB shell properties. Altogether, this manuscript provides new perspectives on US-responsive polymeric designs, envisaging their current and future applications in US imaging and therapy.
Collapse
Affiliation(s)
- Roman A Barmin
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - MirJavad Moosavifar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Andreas Herrmann
- DWI - Leibniz Institute for Interactive Materials Aachen 52074 Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Aachen 52074 Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| |
Collapse
|
28
|
Siebenmorgen C, Poortinga A, van Rijn P. Sono-processes: Emerging systems and their applicability within the (bio-)medical field. ULTRASONICS SONOCHEMISTRY 2023; 100:106630. [PMID: 37826890 PMCID: PMC10582584 DOI: 10.1016/j.ultsonch.2023.106630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
Sonochemistry, although established in various fields, is still an emerging field finding new effects of ultrasound on chemical systems and are of particular interest for the biomedical field. This interdisciplinary area of research explores the use of acoustic waves with frequencies ranging from 20 kHz to 1 MHz to induce physical and chemical changes. By subjecting liquids to ultrasonic waves, sonochemistry has demonstrated the ability to accelerate reaction rates, alter chemical reaction pathways, and change physical properties of the system while operating under mild reaction conditions. It has found its way into diverse industries including food processing, pharmaceuticals, material science, and environmental remediation. This review provides an overview of the principles, advancements, and applications of sonochemistry with a particular focus on the domain of (bio-)medicine. Despite the numerous benefits sonochemistry has to offer, most of the research in the (bio-)medical field remains in the laboratory stage. Translation of these systems into clinical practice is complex as parameters used for medical ultrasound are limited and toxic side effects must be minimized in order to meet regulatory approval. However, directing attention towards the applicability of the system in clinical practice from the early stages of research holds significant potential to further amplify the role of sonochemistry in clinical applications.
Collapse
Affiliation(s)
- Clio Siebenmorgen
- University of Groningen, University Medical Center Groningen, Department of Biomedical Engineering-FB40, Deusinglaan 1, Groningen 9713 AV, The Netherlands.
| | - Albert Poortinga
- Technical University Eindhoven, Department of Mechanical Engineering, Gemini Zuid, de Zaale, Eindhoven 5600 MB, The Netherlands.
| | - Patrick van Rijn
- University of Groningen, University Medical Center Groningen, Department of Biomedical Engineering-FB40, Deusinglaan 1, Groningen 9713 AV, The Netherlands.
| |
Collapse
|
29
|
van Elburg B, Deprez J, van den Broek M, De Smedt SC, Versluis M, Lajoinie G, Lentacker I, Segers T. Dependence of sonoporation efficiency on microbubble size: An in vitro monodisperse microbubble study. J Control Release 2023; 363:747-755. [PMID: 37778466 DOI: 10.1016/j.jconrel.2023.09.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/24/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023]
Abstract
Sonoporation is the process where intracellular drug delivery is facilitated by ultrasound-driven microbubble oscillations. Several mechanisms have been proposed to relate microbubble dynamics to sonoporation including shear and normal stress. The present work aims to gain insight into the role of microbubble size on sonoporation and thereby into the relevant mechanism(s) of sonoporation. To this end, we measured the sonoporation efficiency while varying microbubble size using monodisperse microbubble suspensions. Sonoporation experiments were performed in vitro on cell monolayers using a single ultrasound pulse with a fixed frequency of 1 MHz while the acoustic pressure amplitude and pulse length were varied at 250, 500, and 750 kPa, and 10, 100, and 1000 cycles, respectively. Sonoporation efficiency was quantified using flow cytometry by measuring the FITC-dextran (4 kDa and 2 MDa) fluorescence intensity in 10,000 cells per experiment to average out inherent variations in the bioresponse. Using ultra-high-speed imaging at 10 million frames per second, we demonstrate that the bubble oscillation amplitude is nearly independent of the equilibrium bubble radius at acoustic pressure amplitudes that induce sonoporation (≥ 500 kPa). However, we show that sonoporation efficiency is strongly dependent on the equilibrium bubble size and that under all explored driving conditions most efficiently induced by bubbles with a radius of 4.7 μm. Polydisperse microbubbles with a typical ultrasound contrast agent size distribution perform almost an order of magnitude lower in terms of sonoporation efficiency than the 4.7-μm bubbles. We elucidate that for our system shear stress is highly unlikely the mechanism of action. By contrast, we show that sonoporation efficiency correlates well with an estimate of the bubble-induced normal stress.
Collapse
Affiliation(s)
- Benjamin van Elburg
- Physics of Fluids Group and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - Joke Deprez
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium
| | - Martin van den Broek
- BIOS / Lab on a Chip Group, Max-Planck Center Twente for Complex Fluid Dynamics, MESA+ Institute for Nanotechnology, University of Twente, Enschede, Netherlands
| | - Stefaan 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
| | - Michel Versluis
- Physics of Fluids Group and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - Guillaume Lajoinie
- Physics of Fluids Group and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Tim Segers
- BIOS / Lab on a Chip Group, Max-Planck Center Twente for Complex Fluid Dynamics, MESA+ Institute for Nanotechnology, University of Twente, Enschede, Netherlands.
| |
Collapse
|
30
|
Nittayacharn P, Abenojar E, Cooley M, Berg F, Counil C, Sojahrood AJ, Khan MS, Yang C, Berndl E, Golczak M, Kolios MC, Exner AA. Efficient ultrasound-mediated drug delivery to orthotopic liver tumors - Direct comparison of doxorubicin-loaded nanobubbles and microbubbles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555196. [PMID: 37732235 PMCID: PMC10508722 DOI: 10.1101/2023.09.01.555196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Liver metastasis is a major obstacle in treating aggressive cancers, and current therapeutic options often prove insufficient. To overcome these challenges, there has been growing interest in ultrasound-mediated drug delivery using lipid-shelled microbubbles (MBs) and nanobubbles (NBs) as promising strategies for enhancing drug delivery to tumors. Our previous work demonstrated the potential of Doxorubicin-loaded C3F8 NBs (hDox-NB, 280 ± 123 nm) in improving cancer treatment in vitro using low-frequency ultrasound. In this study, we investigated the pharmacokinetics and biodistribution of sonicated hDox-NBs in orthotopic rat liver tumors. We compared their delivery and therapeutic efficiency with size-isolated MBs (hDox-MB, 1104 ± 373 nm). Results showed a similar accumulation of hDox in tumors treated with hDox-MBs and unfocused therapeutic ultrasound (hDox-MB+TUS) and hDox-NB+TUS. However, significantly increased apoptotic cell death in the tumor and fewer off-target apoptotic cells in the normal liver were found upon the treatment with hDox-NB+TUS. The tumor-to-liver apoptotic ratio was elevated 9.4-fold following treatment with hDox-NB+TUS compared to hDox-MB+TUS, suggesting that the therapeutic efficacy and specificity are significantly increased when using hDox-NB+TUS. These findings highlight the potential of this approach as a viable treatment modality for liver tumors. By elucidating the behavior of drug-loaded bubbles in vivo, we aim to contribute to developing more effective liver cancer treatments that could ultimately improve patient outcomes and decrease off-target side effects.
Collapse
Affiliation(s)
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Michaela Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Felipe Berg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Claire Counil
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Celina Yang
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Elizabeth Berndl
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Michael C. Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Agata A. Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
31
|
Kobayashi R, Narita J, Nakaoka N, Krafft MP, Koyama D. Quantitative estimation of phospholipid molecules desorbed from a microbubble surface under ultrasound irradiation. Sci Rep 2023; 13:13693. [PMID: 37608058 PMCID: PMC10444774 DOI: 10.1038/s41598-023-40823-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/17/2023] [Indexed: 08/24/2023] Open
Abstract
Microbubbles have potential applications as drug and gene carriers, and drug release can be triggered by externally applied ultrasound irradiation while inside blood vessels. Desorption of molecules forming the microbubble shell can be observed under ultrasound irradiation of a single isolated microbubble, and the volume of desorbed molecules can be quantitatively estimated from the contact angle between the bubble and a glass plate. Microbubbles composed of a 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) shell and a poorly-soluble gas are created. When the microbubbles are exposed to a pulsed ultrasound, the contact angles increase dramatically; the percentage of DMPC molecules desorbed from the bubble surface reaches 70%. Vibration of a single bubble in the radial direction is measured using a laser Doppler vibrometer. The relationship between the vibrational characteristics and the amount of molecular desorption reveals that a larger vibrational amplitude of the bubble around the resonance size induces a larger amount of molecular desorption. These results support the possibility of controlling molecular desorption with pulsed ultrasound.
Collapse
Affiliation(s)
- Reina Kobayashi
- Faculty of Science and Engineering, Doshisha University, 1-3 TataraMiyakodani, Kyotanabe, Kyoto, 610-0321, Japan
| | - Jun Narita
- Faculty of Science and Engineering, Doshisha University, 1-3 TataraMiyakodani, Kyotanabe, Kyoto, 610-0321, Japan
| | - Natsumi Nakaoka
- Faculty of Science and Engineering, Doshisha University, 1-3 TataraMiyakodani, Kyotanabe, Kyoto, 610-0321, Japan
| | - Marie Pierre Krafft
- Institut Charles Sadron (CNRS), University of Strasbourg, 23 rue du Loess, 67034, Strasbourg, France
| | - Daisuke Koyama
- Faculty of Science and Engineering, Doshisha University, 1-3 TataraMiyakodani, Kyotanabe, Kyoto, 610-0321, Japan.
| |
Collapse
|
32
|
Nistor M, Rugina D, Diaconeasa Z, Socaciu C, Socaciu MA. Pentacyclic Triterpenoid Phytochemicals with Anticancer Activity: Updated Studies on Mechanisms and Targeted Delivery. Int J Mol Sci 2023; 24:12923. [PMID: 37629103 PMCID: PMC10455110 DOI: 10.3390/ijms241612923] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pentacyclic triterpenoids (TTs) represent a unique family of phytochemicals with interesting properties and pharmacological effects, with some representatives, such as betulinic acid (BA) and betulin (B), being mainly investigated as potential anticancer molecules. Considering the recent scientific and preclinical investigations, a review of their anticancer mechanisms, structure-related activity, and efficiency improved by their insertion in nanolipid vehicles for targeted delivery is presented. A systematic literature study about their effects on tumor cells in vitro and in vivo, as free molecules or encapsulated in liposomes or nanolipids, is discussed. A special approach is given to liposome-TTs and nanolipid-TTs complexes to be linked to microbubbles, known as contrast agents in ultrasonography. The production of such supramolecular conjugates to deliver the drugs to target cells via sonoporation represents a new scientific and applicative direction to improve TT efficiency, considering that they have limited availability as lipophilic molecules. Relevant and recent examples of in vitro and in vivo studies, as well as the challenges for the next steps towards the application of these complex delivery systems to tumor cells, are discussed, as are the challenges for the next steps towards the application of targeted delivery to tumor cells, opening new directions for innovative nanotechnological solutions.
Collapse
Affiliation(s)
- Madalina Nistor
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Dumitrita Rugina
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Zorita Diaconeasa
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Carmen Socaciu
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Mihai Adrian Socaciu
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
- Department of Radiology, Imaging & Nuclear Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400347 Cluj-Napoca, Romania
| |
Collapse
|
33
|
Liu Q, Gao Y, Cong H, Liao L. Ultrasound-assisted intravesical botulinum toxin A delivery attenuates acetic acid-induced bladder hyperactivity in rats. Front Pharmacol 2023; 14:1214145. [PMID: 37554988 PMCID: PMC10406439 DOI: 10.3389/fphar.2023.1214145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Background: Intradetrusor injection of botulinum toxin A (BTX-A) is an effective treatment for overactive bladder (OAB). However, the occurrence of adverse events associated with BTX-A injection therapy hinders its acceptance among patients and its clinical promotion. Intravesical instillation of BTX-A offers a promising alternative to injection therapy for treating OAB. Nevertheless, due to the presence of the bladder permeability barrier (BPB) and the high molecular weight of BTX-A, direct instillation is unable to penetrate the bladder urothelium. Purpose: This study aims to investigate the safety and feasibility of ultrasound-assisted intravesical delivery of BTX-A and its potential benefits in a rat model of bladder hyperactivity induced by acetic acid instillation. Methods: Hengli BTX-A and microbubbles (MB) were mixed and prepared as a novel complex. The size distribution and zeta potentials of the complex were measured. On day 1, rats' bladders were instilled with 1 mL of saline, BTX-A (20 U in 1 mL), MB, or MB-BTX-A (20 U in 1 mL) complex with or without ultrasound (US) exposure (1 MHz, 1.5 W/cm2, 50% duty cycle, sonication for 10 s with a 10-s pause for a total of 10 min). The instillations were maintained for 30 min. After 7 days, cystometry was performed by filling the bladder with saline and 0.3% acetic acid (AA). Bladders were collected, weighed, and processed for immunoblotting, enzyme-linked immunosorbent assay (ELISA), histologic, and immunofluorescence analyses. Expression and distribution of SNAP-25 and SNAP-23 were assessed using Western blot and immunofluorescence. Calcitonin gene-related peptide (CGRP) in the bladder was detected using ELISA. Results: Intercontraction intervals (ICI) decreased by 72.99%, 76.16%, and 73.96% in rats pretreated with saline, BTX-A, and US + MB, respectively. However, rats treated with US + MB + BTX-A showed a significantly reduced response to AA instillation (57.31% decrease in ICI) without affecting amplitude, baseline pressure, or threshold pressure. Rats treated with US + MB + BTX-A exhibited increased cleavage of SNAP-25 and CGRP expression compared to the control group. Conclusion: Ultrasound-assisted intravesical delivery of BTX-A, with the assistance of MB cavitation, led to cleavage of SNAP-25, inhibition of calcitonin gene-related peptide release from afferent nerve terminals, and amelioration of acetic acid-induced bladder hyperactivity. These results support ultrasound-assisted intravesical delivery as an efficient non-injection method for administering BTX-A.
Collapse
Affiliation(s)
- Qinggang Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yi Gao
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Huiling Cong
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Limin Liao
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Bourdin A, Ortoli M, Karadayi R, Przegralek L, Sennlaub F, Bodaghi B, Guillonneau X, Carpentier A, Touhami S. Efficacy and Safety of Low-Intensity Pulsed Ultrasound-Induced Blood-Retinal Barrier Opening in Mice. Pharmaceutics 2023; 15:1896. [PMID: 37514082 PMCID: PMC10384184 DOI: 10.3390/pharmaceutics15071896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Systemic drugs can treat various retinal pathologies such as retinal cancers; however, their ocular diffusion may be limited by the blood-retina barrier (BRB). Sonication corresponds to the use of ultrasound (US) to increase the permeability of cell barriers including in the BRB. The objective was to study the efficacy and safety of sonication using microbubble-assisted low-intensity pulsed US in inducing a transient opening of the BRB. The eyes of C57/BL6J mice were sonicated at different acoustic pressures (0.10 to 0.50 MPa). Efficacy analyses consisted of fluorescein angiography (FA) performed at different timepoints and the size of the leaked molecules was assessed using FITC-marked dextrans. Tolerance was assessed by fundus photographs, optical coherence tomography, immunohistochemistry, RT-qPCR, and electroretinograms. Sonication at 0.15 MPa was the most suitable pressure for transient BRB permeabilization without altering the morphology or function of the retina. It did not increase the expression of inflammation or apoptosis markers in the retina, retinal pigment epithelium, or choroid. The dextran assay suggested that drugs up to 150 kDa in size can cross the BRB. Microbubble-assisted sonication at an optimized acoustic pressure of 0.15 MPa provides a non-invasive method to transiently open the BRB, increasing the retinal diffusion of systemic drugs without inducing any noticeable side-effect.
Collapse
Affiliation(s)
- Alexandre Bourdin
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Manon Ortoli
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Remi Karadayi
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Lauriane Przegralek
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Florian Sennlaub
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Bahram Bodaghi
- Ophthalmology Department, Pitié Salpêtrière University Hospital, AP-HP, Sorbonne Université, 75013 Paris, France
| | - Xavier Guillonneau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Alexandre Carpentier
- Department of Neurosurgery, Pitié Salpêtrière University Hospital, AP-HP, Sorbonne Université, 75013 Paris, France
- NeurOn Brain Machine Interface Clinical Research Group, Pitié Salpêtrière University Hospital, AP-HP, Sorbonne Université, 75013 Paris, France
- ASTRL Advanced Surgical Technologies Research Laboratory, Sorbonne Université, 75013 Paris, France
| | - Sara Touhami
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
- Ophthalmology Department, Pitié Salpêtrière University Hospital, AP-HP, Sorbonne Université, 75013 Paris, France
| |
Collapse
|
35
|
Azami RH, Forsberg F, Eisenbrey JR, Sarkar K. Ambient Pressure Sensitivity of the Subharmonic Response of Coated Microbubbles: Effects of Acoustic Excitation Parameters. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1550-1560. [PMID: 37100673 PMCID: PMC10306329 DOI: 10.1016/j.ultrasmedbio.2023.02.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/20/2023] [Accepted: 02/27/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE The sensitivity of the acoustic response of microbubbles, specifically a strong correlation between their subharmonic response and the ambient pressure, has motivated development of a non-invasive subharmonic-aided pressure estimation (SHAPE) method. However, this correlation has previously been found to vary depending on the microbubble type, the acoustic excitation and the hydrostatic pressure range. In this study, the ambient pressure sensitivity of microbubble response was investigated. METHODS The fundamental, subharmonic, second harmonic and ultraharmonic responses from an in-house lipid-coated microbubble were measured for excitations with peak negative pressures (PNPs) of 50-700 kPa and frequencies of 2, 3 and 4 MHz in the ambient overpressure range 0-25 kPa (0-187 mmHg) in an in vitro setup. RESULTS The subharmonic response typically has three stages-occurrence, growth and saturation-with increasing excitation PNP. We find distinct decreasing and increasing variations of the subharmonic signal with overpressure that are closely related to the threshold of subharmonic generation in a lipid-shelled microbubble. Above the excitation threshold, that is, in the growth-saturation phase, subharmonic signals decreased linearly with slopes as high as -0.56 dB/kPa with ambient pressure increase; below the threshold excitation (at atmospheric pressure), increasing overpressure triggers subharmonic generation, indicating a lowering of subharmonic threshold, and therefore leads to an increase in subharmonic with overpressure, the maximum enhancement being ∼11 dB for 15 kPa overpressure at 2 MHz and 100 kPa PNP. CONCLUSION This study indicates the possible development of novel and improved SHAPE methodologies.
Collapse
Affiliation(s)
- Roozbeh H Azami
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kausik Sarkar
- Department of Mechanical and Aerospace Engineering, George Washington University, Washington, DC, USA.
| |
Collapse
|
36
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
37
|
López-Rodulfo IM, Villa-Martínez E, Rios A, Escalante B. Caveolin Delivered by Ultrasound-Mediated Microbubble Destruction Prevents Endothelial Cell Proliferation. Cell Mol Bioeng 2023; 16:219-229. [PMID: 37456788 PMCID: PMC10338419 DOI: 10.1007/s12195-023-00763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/29/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction The nitric oxide synthase (eNOS) is an important regulator of vascular homeostasis. eNOS is modulated by intracellular mechanisms that include protein-protein interaction with Caveolin-1 (Cav). Cav binds to and impairs eNOS activation reducing vascular permeability and angiogenesis. Blocking of eNOS by Cav has been proposed as therapeutic antiangiogenic approach. However, the efficient and controlled delivery of the peptide requires to be solved. Methods The effect of antennapedia (AP)-Cav loaded into microbubbles (MBs) and delivered by ultrasound-mediated microbubble destruction (UMMD) into brain endothelial cells (bEnd.3 cells) was evaluated on NO production using DAF2-DA, cell migration assessed by the wound healing assay, cell proliferation with BrdU, and ex-vivo angiogenesis in rat aortic rings. Results An enhanced inhibitory effect of AP-Cav was observed on cells treated with UMMD. MBs and ultrasound disruption delivery of AP-Cav increased acetylcholine-induced NO release, wound healing, cell proliferation, and angiogenesis inhibition on bEnd.3 cells, compared to free AP-Cav administration. Conclusion We demonstrated that the delivery of Cav via AP-Cav-loaded MBs and UMMD may be an administration method for Cav that would increase its therapeutic potential by enhancing efficacy and cellular specificity.
Collapse
Affiliation(s)
- Iván M. López-Rodulfo
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
- Present Address: Aarhus Universitet, Nordre Ringgade 1, 8000 Aarhus C, Denmark
| | - Elisa Villa-Martínez
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
| | - Amelia Rios
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
| | - Bruno Escalante
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad-Monterrey, Cinvestav Monterrey, Vía del Conocimiento 201, PIIT, Apodaca, N. L. 66600 México
| |
Collapse
|
38
|
Navarro-Becerra JA, Borden MA. Targeted Microbubbles for Drug, Gene, and Cell Delivery in Therapy and Immunotherapy. Pharmaceutics 2023; 15:1625. [PMID: 37376072 DOI: 10.3390/pharmaceutics15061625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Microbubbles are 1-10 μm diameter gas-filled acoustically-active particles, typically stabilized by a phospholipid monolayer shell. Microbubbles can be engineered through bioconjugation of a ligand, drug and/or cell. Since their inception a few decades ago, several targeted microbubble (tMB) formulations have been developed as ultrasound imaging probes and ultrasound-responsive carriers to promote the local delivery and uptake of a wide variety of drugs, genes, and cells in different therapeutic applications. The aim of this review is to summarize the state-of-the-art of current tMB formulations and their ultrasound-targeted delivery applications. We provide an overview of different carriers used to increase drug loading capacity and different targeting strategies that can be used to enhance local delivery, potentiate therapeutic efficacy, and minimize side effects. Additionally, future directions are proposed to improve the tMB performance in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, CO 80309, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
39
|
LuTheryn G, Ho EML, Choi V, Carugo D. Cationic Microbubbles for Non-Selective Binding of Cavitation Nuclei to Bacterial Biofilms. Pharmaceutics 2023; 15:pharmaceutics15051495. [PMID: 37242736 DOI: 10.3390/pharmaceutics15051495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/06/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The presence of multi-drug resistant biofilms in chronic, persistent infections is a major barrier to successful clinical outcomes of therapy. The production of an extracellular matrix is a characteristic of the biofilm phenotype, intrinsically linked to antimicrobial tolerance. The heterogeneity of the extracellular matrix makes it highly dynamic, with substantial differences in composition between biofilms, even in the same species. This variability poses a major challenge in targeting drug delivery systems to biofilms, as there are few elements both suitably conserved and widely expressed across multiple species. However, the presence of extracellular DNA within the extracellular matrix is ubiquitous across species, which alongside bacterial cell components, gives the biofilm its net negative charge. This research aims to develop a means of targeting biofilms to enhance drug delivery by developing a cationic gas-filled microbubble that non-selectively targets the negatively charged biofilm. Cationic and uncharged microbubbles loaded with different gases were formulated and tested to determine their stability, ability to bind to negatively charged artificial substrates, binding strength, and, subsequently, their ability to adhere to biofilms. It was shown that compared to their uncharged counterparts, cationic microbubbles facilitated a significant increase in the number of microbubbles that could both bind and sustain their interaction with biofilms. This work is the first to demonstrate the utility of charged microbubbles for the non-selective targeting of bacterial biofilms, which could be used to significantly enhance stimuli-mediated drug delivery to the bacterial biofilm.
Collapse
Affiliation(s)
- Gareth LuTheryn
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), The Botnar Research Centre, University of Oxford, Windmill Road, Oxford OX3 7HE, UK
- Faculty of Engineering and Physical Sciences, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Elaine M L Ho
- Faculty of Engineering and Physical Sciences, University of Southampton, University Road, Southampton SO17 1BJ, UK
- Artificial Intelligence and Informatics, The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0QX, UK
| | - Victor Choi
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Parks Road, Oxford OX1 3PJ, UK
| | - Dario Carugo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), The Botnar Research Centre, University of Oxford, Windmill Road, Oxford OX3 7HE, UK
| |
Collapse
|
40
|
Smith MR, Khan S, Curiel L. Comparing Phantom and Animal Metrics Applied in the Determination of Focused Ultrasound Stable and Inertial Cavitation Levels. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1118-1128. [PMID: 36732151 DOI: 10.1016/j.ultrasmedbio.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/30/2022] [Accepted: 12/19/2022] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The opening of the blood-brain barrier (BBB) to allow therapeutic drug passage can be achieved by inducing microbubble cavitation using focused ultrasound (FUS). This approach can be monitored through analysis of the received signal to distinguish between stable cavitation associated with safe BBB opening and inertial cavitation associated with blood vessel damage. In this study, FUS phantom and animal studies were used to evaluate the experimental conditions that generate several cross-consistent metrics having the potential to be combined for the reliable, automatic control of cavitation levels. METHODS Typical metrics for cavitation monitoring involve observing changes in the spectrum generated by applying the discrete Fourier transform (DFT) to the time domain signal detected using a hydrophone during FUS. A confocal hydrophone was used to capture emissions during a 10 ms FUS burst, sampled at 32 ns intervals, to produce 321,500 points and a high-resolution spectrum when transformed. The FUS spectra were analyzed to show the impact that equipment-transients and well-known DFT-related distortions had on the metrics used for cavitation control. A new approach, physical sparsification (PH-SP), was introduced to sharpen FUS spectral peaks and minimize the effect of these distortions. DISCUSSION It was demonstrated that the general spectral signal-to-noise ratio (SNR) could be improved by removing the initial noisy phantom hydrophone signal transient. Minor changes in the transient length digitally removed from the sampled values significantly changed the spectral bandwidths of all the harmonically related FUS signals. We evaluated signal processing techniques to minimize the impact these DFT-related distortions on area-under-the-curve (AUC) metric calculations, and we identified the advantages of using PH-SP and proposed new metrics when characterizing FUS spectral properties. The results show many second, third and sub-harmonic metrics provide cross-consistent evidence of changes between stable and inertial cavitation levels. Removing the first harmonic signal component with a hardware low-pass filter allowed the hydrophone gain to be boosted without introducing distortion, leading to an improved analysis of the sub-harmonic signal orders of magnitude smaller in intensity. Metrics that optimized the energy in the real component of the complex-valued PH-SP spectra provided a 32% increase in the sub-harmonic sensitivity compared to standard metrics. CONCLUSION A preliminary investigation of existing and proposed metrics showed that system noise could be large enough to mask the transition between stable and inertial cavitation. Strong narrowing of sub-harmonic peak shapes on applying physical sparsification (PH-SP) were seen in both phantom and animal studies. However, validating equivalent trends of the metrics with pressure were limited by the increased system noise level in the animal study combined with the natural variability between subjects studied. The combined use of hardware low-pass filters and physical sparsification to selectively removing distortions in the spectrum allowed the optimization of metrics for cavitation monitoring by improving the sub-harmonic sensitivity.
Collapse
Affiliation(s)
- Michael R Smith
- Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Alberta, Canada; Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, Canada.
| | - Sonia Khan
- Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Alberta, Canada
| | - Laura Curiel
- Department of Biometrical Engineering, Schulich School of Engineering, University of Calgary, Alberta, Canada
| |
Collapse
|
41
|
Mühlenpfordt M, Olsen EB, Kotopoulis S, Torp SH, Snipstad S, Davies CDL, Olsman M. Real-Time Intravital Imaging of Acoustic Cluster Therapy-Induced Vascular Effects in the Murine Brain. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1212-1226. [PMID: 36858913 DOI: 10.1016/j.ultrasmedbio.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The blood-brain barrier (BBB) is an obstacle for cerebral drug delivery. Controlled permeabilization of the barrier by external stimuli can facilitate the delivery of drugs to the brain. Acoustic Cluster Therapy (ACT®) is a promising strategy for transiently and locally increasing the permeability of the BBB to macromolecules and nanoparticles. However, the mechanism underlying the induced permeability change and subsequent enhanced accumulation of co-injected molecules requires further elucidation. METHODS In this study, the behavior of ACT® bubbles in microcapillaries in the murine brain was observed using real-time intravital multiphoton microscopy. For this purpose, cranial windows aligned with a ring transducer centered around an objective were mounted to the skull of mice. Dextrans labeled with 2 MDa fluorescein isothiocyanate (FITC) were injected to delineate the blood vessels and to visualize extravasation. DISCUSSION Activated ACT® bubbles were observed to alter the blood flow, inducing transient and local increases in the fluorescence intensity of 2 MDa FITC-dextran and subsequent extravasation in the form of vascular outpouchings. The observations indicate that ACT® induced a transient vascular leakage without causing substantial damage to the vessels in the brain. CONCLUSION The study gave novel insights into the mechanism underlying ACT®-induced enhanced BBB permeability which will be important considering treatment optimization for a safe and efficient clinical translation of ACT®.
Collapse
Affiliation(s)
- Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Exact Therapeutics AS, Oslo, Norway.
| | - Emma Bøe Olsen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Spiros Kotopoulis
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre H Torp
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Pathology, St. Olav's Hospital, Trondheim University Hospital Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | | | - Marieke Olsman
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
42
|
Chabouh G, van Elburg B, Versluis M, Segers T, Quilliet C, Coupier G. Buckling of lipidic ultrasound contrast agents under quasi-static load. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2023; 381:20220025. [PMID: 36774952 DOI: 10.1098/rsta.2022.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/10/2022] [Indexed: 06/18/2023]
Abstract
Collapse of lipidic ultrasound contrast agents under high-frequency compressive load has been historically interpreted by the vanishing of surface tension. By contrast, buckling of elastic shells is known to occur when costly compressible stress is released through bending. Through quasi-static compression experiments on lipidic shells, we analyse the buckling events in the framework of classical elastic buckling theory and deduce the mechanical characteristics of these shells. They are then compared with that obtained through acoustic characterization. This article is part of the theme issue 'Probing and dynamics of shock sensitive shells'.
Collapse
Affiliation(s)
- Georges Chabouh
- Université Grenoble Alpes, CNRS, LIPhy, Grenoble 38000, France
| | - Benjamin van Elburg
- Physics of Fluids Group, Technical Medical (TechMed) Center and MESA+ Institute for Nanotechnology, University of Twente, Enschede 7500 AE, The Netherlands
| | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Center and MESA+ Institute for Nanotechnology, University of Twente, Enschede 7500 AE, The Netherlands
| | - Tim Segers
- BIOS/Lab-on-a-Chip Group, Max Planck Center Twente for Complex Fluid Dynamics, MESA+ Institute for Nanotechnology, University of Twente, Enschede, The Netherlands
| | | | - Gwennou Coupier
- Université Grenoble Alpes, CNRS, LIPhy, Grenoble 38000, France
| |
Collapse
|
43
|
Ostasevicius V, Jurenas V, Mikuckyte S, Vezys J, Stankevicius E, Bubulis A, Venslauskas M, Kizauskiene L. Development of a Low-Frequency Piezoelectric Ultrasonic Transducer for Biological Tissue Sonication. SENSORS (BASEL, SWITZERLAND) 2023; 23:3608. [PMID: 37050668 PMCID: PMC10098853 DOI: 10.3390/s23073608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
The safety of ultrasound exposure is very important for a patient's well-being. High-frequency (1-10 MHz) ultrasound waves are highly absorbed by biological tissue and have limited therapeutic effects on internal organs. This article presents the results of the development and application of a low-frequency (20-100 kHz) ultrasonic transducer for sonication of biological tissues. Using the methodology of digital twins, consisting of virtual and physical twins, an ultrasonic transducer has been developed that emits a focused ultrasound signal that penetrates into deeper biological tissues. For this purpose, the ring-shaped end surface of this transducer is excited not only by the main longitudinal vibrational mode, which is typical of the flat end surface transducers used to date, but also by higher mode radial vibrations. The virtual twin simulation shows that the acoustic signal emitted by the ring-shaped transducer, which is excited by a higher vibrational mode, is concentrated into a narrower and more precise acoustic wave that penetrates deeper into the biological tissue and affects only the part of the body to be treated, but not the whole body.
Collapse
Affiliation(s)
- Vytautas Ostasevicius
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, LT-51424 Kaunas, Lithuania
| | - Vytautas Jurenas
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, LT-51424 Kaunas, Lithuania
| | - Sandra Mikuckyte
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, LT-51424 Kaunas, Lithuania
| | - Joris Vezys
- Department of Mechanical Engineering, Kaunas University of Technology, Studentu Street 50, LT-51368 Kaunas, Lithuania
| | - Edgaras Stankevicius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, A. Mickevicius Street 9, LT-44307 Kaunas, Lithuania
| | - Algimantas Bubulis
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, LT-51424 Kaunas, Lithuania
| | - Mantas Venslauskas
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, LT-51424 Kaunas, Lithuania
| | - Laura Kizauskiene
- Department of Computer Sciences, Kaunas University of Technology, Studentu Street 50, LT-51368 Kaunas, Lithuania
| |
Collapse
|
44
|
Shen L, Tian Z, Zhang J, Zhu H, Yang K, Li T, Rich J, Upreti N, Hao N, Pei Z, Jin G, Yang S, Liang Y, Chaohui W, Huang TJ. Acousto-dielectric tweezers for size-insensitive manipulation and biophysical characterization of single cells. Biosens Bioelectron 2023; 224:115061. [PMID: 36634509 DOI: 10.1016/j.bios.2023.115061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/03/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
The intrinsic biophysical properties of cells, such as mechanical, acoustic, and electrical properties, are valuable indicators of a cell's function and state. However, traditional single-cell biophysical characterization methods are hindered by limited measurable properties, time-consuming procedures, and complex system setups. This study presents acousto-dielectric tweezers that leverage the balance between controllable acoustophoretic and dielectrophoretic forces applied on cells through surface acoustic waves and alternating current electric fields, respectively. Particularly, the balanced acoustophoretic and dielectrophoretic forces can trap cells at equilibrium positions independent of the cell size to differentiate between various cell-intrinsic mechanical, acoustic, and electrical properties. Experimental results show our mechanism has the potential for applications in single-cell analysis, size-insensitive cell separation, and cell phenotyping, which are all primarily based on cells' intrinsic biophysical properties. Our results also show the measured equilibrium position of a cell can inversely determine multiple biophysical properties, including membrane capacitance, cytoplasm conductivity, and acoustic contrast factor. With these features, our acousto-dielectric tweezing mechanism is a valuable addition to the resources available for biophysical property-based biological and medical research.
Collapse
Affiliation(s)
- Liang Shen
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA; State Key Laboratory of Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Zhenhua Tian
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
| | - Jinxin Zhang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Haodong Zhu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Kaichun Yang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Teng Li
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Joseph Rich
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Neil Upreti
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Nanjing Hao
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Zhichao Pei
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Geonsoo Jin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Shujie Yang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA
| | - Yaosi Liang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27708, USA
| | - Wang Chaohui
- State Key Laboratory of Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China.
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
45
|
Lin F, Wo K, Fan X, Wang W, Zou J. Directional Transport of Underwater Bubbles on Solid Substrates: Principles and Applications. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10325-10340. [PMID: 36802468 DOI: 10.1021/acsami.2c21466] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The manipulation of underwater bubbles on substrates has received extensive research interest from both the scientific community and industry, including the chemical industry, machinery, biology, medicine, and other fields. Recent advances in "smart" substrates have enabled the bubbles to be transported on demand. Herein, the progress in the directional transport of underwater bubbles on various types of substrates is summarized, including planes, wires, and cones. The transport mechanism can be classified as buoyancy-driven, Laplace-pressure-difference-driven, and external-force-driven according to the driven force of the bubble. Moreover, the wide applications of directional bubble transport are reported, ranging from gas collection, microbubble reaction, bubble detection and classification, bubble switch, and bubble microrobots. Lastly, the advantages and challenges of various directional bubble transportation methods are discussed, and the current challenges and future prospects in this field are also discussed. This Review outlines the fundamental mechanisms of underwater bubble transportation on solid substrates and helps to understand the methods of optimizing bubble transportation performances.
Collapse
Affiliation(s)
- Fangye Lin
- Ningbo Research Institute, Zhejiang University, Ningbo 315048, China
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
- MedicalSystem Biotechnology Co., Ltd., Ningbo 315104, China
| | - Keyu Wo
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Xujun Fan
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Wei Wang
- Zhejiang University City College, Hangzhou 310015, China
| | - Jun Zou
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
46
|
Keshmiri A, Heitkam S, Bashkatov A, Eftekhari M, Eckert K, Keshavarzi B. Surfactant sorption on a single air bubble in an ultrasonic standing acoustic wave field. Colloids Surf A Physicochem Eng Asp 2023. [DOI: 10.1016/j.colsurfa.2023.131210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
47
|
Lai B, Ouyang X, Mao S, Cao J, Li H, Li S, Wang J. Target tumor therapy in human gastric cancer cells through the combination of docetaxel-loaded cationic lipid microbubbles and ultrasound-triggered microbubble destruction. Funct Integr Genomics 2023; 23:59. [PMID: 36757623 DOI: 10.1007/s10142-022-00952-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/18/2022] [Accepted: 12/25/2022] [Indexed: 02/10/2023]
Abstract
It is well accepted that ultrasound-induced microbubble (USMB) cavitation is a promising method for drug delivery. Ultrasound-targeted destruction of cytotoxic drug-loaded lipid microbubbles (LMs) is used to promote the treatment of cancer. This study aimed to investigate the antitumor effects from a combination of docetaxel-loaded cationic lipid microbubbles (DLLM+) and ultrasound (US)-triggered microbubble destruction (UTMD) on gastric cancer (GC). It was found that the functional dose of DOC in this study was 1 × 10-9 mol/L. We found that DLLM combined with the UTMD group showed greater growth inhibition of the cultured human gastric cancer cells (HGCCs) when compared with the other five groups by arresting the G2/M phase in the cell cycle. However, DLLM+ combined with UTMD showed a higher inhibition rate of tumor growth than DLLM combined with UTMD and that of the RC/CMV-p16 combined with UTMD in vitro and in vivo experiments. DLLM+ combined with UTMD significantly suppressed proliferation and promoted the apoptosis of HGCCs with more cells arrested in the G2/M phase. In addition, DLLM+ combined with UTMD suppressed the proliferation and induced apoptosis by arresting cells in the G2/M phase, which led to a great inhibition of GC progression. Thus, our results indicated that the combination of DLLM+ and UTMD might represent a novel and promising approach to chemotherapy for GC.
Collapse
Affiliation(s)
- Bin Lai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xi Ouyang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengxun Mao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiaqin Cao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglang Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Song Li
- Mudanjiang Medical College, Mudanjiang, China
| | - Jiwei Wang
- Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, No.1, Minde Road, Donghu District, Nanchang, China.
| |
Collapse
|
48
|
Zhao X, Wright A, Goertz DE. An optical and acoustic investigation of microbubble cavitation in small channels under therapeutic ultrasound conditions. ULTRASONICS SONOCHEMISTRY 2023; 93:106291. [PMID: 36640460 PMCID: PMC9852793 DOI: 10.1016/j.ultsonch.2023.106291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 06/04/2023]
Abstract
Therapeutic focused ultrasound in combination with encapsulated microbubbles is being widely investigated for its ability to elicit bioeffects in the microvasculature, such as transient permeabilization for drug delivery or at higher pressures to achieve 'antivascular' effects. While it is well established that the behaviors of microbubbles are altered when they are situated within sufficiently small vessels, there is a paucity of data examining how the bubble population dynamics and emissions change as a function of channel (vessel) diameter over a size range relevant to therapeutic ultrasound, particularly at pressures relevant to antivascular ultrasound. Here we use acoustic emissions detection and high-speed microscopy (10 kframes/s) to examine the behavior of a polydisperse clinically employed agent (Definity®) in wall-less channels as their diameters are scaled from 1200 to 15 µm. Pressures are varied from 0.1 to 3 MPa using either a 5 ms pulse or a sequence of 0.1 ms pulses spaced at 1 ms, both of which have been previously employed in an in vivo context. With increasing pressure, the 1200 µm channel - on the order of small arteries and veins - exhibited inertial cavitation, 1/2 subharmonics and 3/2 ultraharmonics, consistent with numerous previous reports. The 200 and 100 µm channels - in the size range of larger microvessels less affected by therapeutic focused ultrasound - exhibited a distinctly different behavior, having muted development of 1/2 subharmonics and 3/2 ultraharmonics and reduced persistence. These were associated with radiation forces displacing bubbles to the distal wall and inducing clusters that then rapidly dissipated along with emissions. As the diameter transitioned to 50 and then 15 µm - a size regime that is most relevant to therapeutic focused ultrasound - there was a higher threshold for the onset of inertial cavitation as well as subharmonics and ultraharmonics, which importantly had more complex orders that are not normally reported. Clusters also occurred in these channels (e.g. at 3 MPa, the mean lateral and axial sizes were 23 and 72 µm in the 15 µm channel; 50 and 90 µm in the 50 µm channel), however in this case they occupied the entire lumens and displaced the wall boundaries. Damage to the 15 µm channel was observed for both pulse types, but at a lower pressure for the long pulse. Experiments conducted with a 'nanobubble' (<0.45 µm) subpopulation of Definity followed broadly similar features to 'native' Definity, albeit at a higher pressure threshold for inertial cavitation. These results provide new insights into the behavior of microbubbles in small vessels at higher pressures and have implications for therapeutic focused ultrasound cavitation monitoring and control.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| | - Alex Wright
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada
| | - David E Goertz
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| |
Collapse
|
49
|
Yin X, Jiang LH. Extracellular vesicles: Targeting the heart. Front Cardiovasc Med 2023; 9:1041481. [PMID: 36704471 PMCID: PMC9871562 DOI: 10.3389/fcvm.2022.1041481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular diseases rank the highest incidence and mortality worldwide. As the most common type of cardiovascular disease, myocardial infarction causes high morbidity and mortality. Recent studies have revealed that extracellular vesicles, including exosomes, show great potential as a promising cell-free therapy for the treatment of myocardial infarction. However, low heart-targeting efficiency and short plasma half-life have hampered the clinical translation of extracellular vesicle therapy. Currently, four major types of strategies aiming at enhancing target efficiency have been developed, including modifying EV surface, suppressing non-target absorption, increasing the uptake efficiency of target cells, and utilizing a hydrogel patch. This presented review summarizes the current research aimed at EV heart targeting and discusses the challenges and opportunities in EV therapy, which will be beneficial for the development of effective heart-targeting strategies.
Collapse
Affiliation(s)
- Xin Yin
- Faculty of Life Sciences and Technology, Kunming University of Science and Technology, Kunming, China,Department of Ultrasound, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China,The First People’s Hospital of Yunnan, Kunming, Yunnan, China
| | - Li-Hong Jiang
- Department of Ultrasound, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China,The First People’s Hospital of Yunnan, Kunming, Yunnan, China,*Correspondence: Li-Hong Jiang,
| |
Collapse
|
50
|
Tabata H, Koyama D, Matsukawa M, Krafft MP, Yoshida K. Concentration-Dependent Viscoelasticity of Poloxamer-Shelled Microbubbles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:433-441. [PMID: 36580034 DOI: 10.1021/acs.langmuir.2c02690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The oscillation of shelled microbubbles during exposure to ultrasound is influenced by the mechanical properties of the shell components. The oscillation behavior of bubbles coated with various phospholipids and other amphiphiles has been studied. However, there have been few investigations of how the adsorption conditions of the shell molecules relate to the viscoelastic properties of the shell and influence the oscillation behavior of the bubbles. In the present study, we investigated the oscillation characteristics of microbubbles coated with a poloxamer surfactant, that is, Pluronic F-68, at several concentrations after the adsorption kinetics of the surfactant at the gas-water interface had reached equilibrium. The dilatational viscoelasticity of the shell during exposure to ultrasound was analyzed in the frequency domain from the attenuation characteristics of the acoustic pulses propagated in the bubble suspension. At Pluronic F-68 concentrations lower than 2.0 × 10-2 mol L-1, the attenuation characteristics typically exhibited a sharp peak. At concentrations higher than 2.0 × 10-2 mol L-1, the peak flattened. The dilatational elasticity and viscosity of the shell were estimated by fitting the theoretical model to the experimental values, which revealed that both the elasticity and viscosity increased markedly at approximately 2.0 × 10-2 mol L-1. This suggests that the adsorption properties of Pluronic F-68 strongly affect the oscillation characteristics of microbubbles of a size suitable for medical ultrasound diagnostics.
Collapse
Affiliation(s)
- Hiraku Tabata
- Faculty of Science and Engineering, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe, Kyoto610-0321, Japan
| | - Daisuke Koyama
- Faculty of Science and Engineering, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe, Kyoto610-0321, Japan
| | - Mami Matsukawa
- Faculty of Science and Engineering, Doshisha University, 1-3 Tataramiyakodani, Kyotanabe, Kyoto610-0321, Japan
| | - Marie Pierre Krafft
- Institut Charles Sadron (CNRS), University of Strasbourg, 23 rue du Loess, Strasbourg67034, France
| | - Kenji Yoshida
- Center for Frontier Medical Engineering, Chiba University, 1-33 Yayoicho, Inage-ku, Chiba263-8522, Japan
| |
Collapse
|