1
|
Min H, Bau L, Payne SJ, Stride EP. Behavior of Microbubbles on Air-Aqueous Interfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:23259-23267. [PMID: 39454083 PMCID: PMC11542178 DOI: 10.1021/acs.langmuir.4c02546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 10/27/2024]
Abstract
Animal-derived lung surfactants have saved millions of lives of preterm neonates with neonatal Respiratory Distress Syndrome (nRDS). However, a replacement for animal-derived lung surfactants has been sought for decades due to its high manufacturing cost, inaccessibility in low-income countries, and failure to show efficacy when nebulized. This study investigated the use of lipid-coated microbubbles as potential replacements for exogenous lung surfactants. Three different formulations of microbubbles (DPPC with/out PEG40-stearate and poractant alfa) were prepared, and their equilibrium and dynamic surface tensions were tested on a clean air-saline interface or a simulated air-lung fluid interface using a Langmuir-Blodgett trough. In dynamic surface measurements, microbubbles reduced the minimum surface tension compared with the equivalent composition lipid suspension: e.g., PEG-free microbubbles had a minimum surface tension of 4.3 mN/m while the corresponding lipid suspension and poractant alfa had 20.4 (p ≤ 0.0001) and 21.8 mN/m (p ≤ 0.0001), respectively. Two potential mechanisms for the reduction of surface tension were found: Fragmentation of the foams created by microbubble coalescence; and clustering of microbubbles in the aqueous subphase disrupting the interfacial phospholipid monolayer. The predominant mechanism appears to depend on the formulation and/or the environment. The use of microbubbles as a replacement for exogenous lung surfactant products thus shows promise and further work is needed to evaluate efficacy in vivo.
Collapse
Affiliation(s)
- Hyunhong
J Min
- Institute
of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX3 7LD, U.K.
| | - Luca Bau
- Institute
of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX3 7LD, U.K.
| | - Stephen J Payne
- Institute
of Applied Mechanics, National Taiwan University, Taipei 10617, Taiwan
| | - Eleanor P Stride
- Institute
of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX3 7LD, U.K.
| |
Collapse
|
2
|
Fuenteslópez CV, Gray M, Bahcevanci S, Martin A, Smith CAB, Coussios C, Cui Z, Ye H, Patrulea V. Mesenchymal stem cell cryopreservation with cavitation-mediated trehalose treatment. COMMUNICATIONS ENGINEERING 2024; 3:129. [PMID: 39251849 PMCID: PMC11385975 DOI: 10.1038/s44172-024-00265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Dimethylsulfoxide (DMSO) has conventionally been used for cell cryopreservation both in research and in clinical applications, but has long-term cytotoxic effects. Trehalose, a natural disaccharide, has been proposed as a non-toxic cryoprotectant. However, the lack of specific cell membrane transporter receptors inhibits transmembrane transport and severely limits its cryoprotective capability. This research presents a method to successfully deliver trehalose into mesenchymal stem cells (MSCs) using ultrasound in the presence of microbubbles. The optimised trehalose concentration was shown to be able to not only preserve membrane integrity and cell viability but also the multipotency of MSCs, which are essential for stem cell therapy. Confocal imaging revealed that rhodamine-labelled trehalose was transported into cells rather than simply attached to the membrane. Additionally, the membranes were successfully preserved in lyophilised cells. This study demonstrates that ultrasonication with microbubbles facilitated trehalose delivery, offering promising cryoprotective capability without the cytotoxicity associated with DMSO-based methods.
Collapse
Affiliation(s)
- Carla V Fuenteslópez
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Simge Bahcevanci
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Alexander Martin
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Cameron A B Smith
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| | - Viorica Patrulea
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
3
|
Centner CS, Belott CJ, Patel RK, Menze MA, Yaddanapudi K, Kopechek JA. Biomodulatory Effects of Molecular Delivery in Human T Cells Using 3D-Printed Acoustofluidic Devices. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00256-4. [PMID: 39107206 DOI: 10.1016/j.ultrasmedbio.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/28/2024] [Accepted: 06/21/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE Cell-based therapies have shown significant promise for treating many diseases, including cancer. Current cell therapy manufacturing processes primarily utilize viral transduction to insert genomic material into cells, which has limitations, including variable transduction efficiency and extended processing times. Non-viral transfection techniques are also limited by high variability or reduced molecular delivery efficiency. Novel 3D-printed acoustofluidic devices are in development to address these challenges by delivering biomolecules into cells within seconds via sonoporation. METHODS In this study, we assessed biological parameters that influence the ultrasound-mediated delivery of fluorescent molecules (i.e., calcein and 150 kDa FITC-Dextran) to human T cells using flow cytometry and confocal imaging. RESULTS Low cell plating densities (100,000 cells/mL) enhanced molecular delivery compared to higher cell plating densities (p < 0.001), even though cells were resuspended at equal concentrations for acoustofluidic processing. Additionally, cells in the S phase of the cell cycle had enhanced intracellular delivery compared to cells in the G2/M phase (p < 0.001) and G0/G1 phase (p < 0.01), while also maintaining higher viability compared to G0/G1 phase (p < 0.001). Furthermore, the calcium chelator (EGTA) decreased overall molecular delivery levels. Confocal imaging indicated that the actin cytoskeleton had important implications on plasma membrane recovery dynamics after sonoporation. In addition, confocal imaging indicates that acoustofluidic treatment can permeabilize the nuclear membrane, which could enable rapid intranuclear delivery of nucleic acids. CONCLUSIONS The results of this study demonstrate that a 3D-printed acoustofluidic device can enhance molecular delivery to human T cells, which may enable improved techniques for non-viral processing of cell therapies.
Collapse
Affiliation(s)
- Connor S Centner
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Clinton J Belott
- Department of Biology, University of Louisville, Louisville, KY, USA
| | - Riyakumari K Patel
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Michael A Menze
- Department of Biology, University of Louisville, Louisville, KY, USA
| | | | - Jonathan A Kopechek
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
4
|
Zhang Z, Cao W, Xing H, Guo S, Huang L, Wang L, Sui X, Lu K, Luo Y, Wang Y, Yang J. A mix & act liposomes of phospholipase A2-phosphatidylserine for acute brain detoxification by blood‒brain barrier selective-opening. Acta Pharm Sin B 2024; 14:1827-1844. [PMID: 38572103 PMCID: PMC10985032 DOI: 10.1016/j.apsb.2023.11.015] [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] [Received: 08/19/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 04/05/2024] Open
Abstract
In the treatment of central nervous system disease, the blood-brain barrier (BBB) is a major obstruction to drug delivery that must be overcome. In this study, we propose a brain-targeted delivery strategy based on selective opening of the BBB. This strategy allows some simple bare nanoparticles to enter the brain when mixed with special opening material; however, the BBB still maintains the ability to completely block molecules from passing through. Based on the screening of BBB opening and matrix delivery materials, we determined that phospholipase A2-catalyzed 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoserine liposomes can efficiently carry drugs into the brain immediately. At an effective dose, this delivery system is safe, especially with its effect on the BBB being reversible. This mix & act delivery system has a simple structure and rapid preparation, making it a strong potential candidate for drug delivery across the BBB.
Collapse
Affiliation(s)
- Zinan Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Wenbin Cao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Huanchun Xing
- Tianjin University of Science and Technology, Tianjin 300222, China
| | - Shuai Guo
- Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Lijuan Huang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Lin Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Xin Sui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Kui Lu
- Tianjin University of Science and Technology, Tianjin 300222, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| | - Jun Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institutes of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
5
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
6
|
Spatarelu CP, Jandhyala S, Luke GP. Dual-drug loaded ultrasound-responsive nanodroplets for on-demand combination chemotherapy. ULTRASONICS 2023; 133:107056. [PMID: 37269682 DOI: 10.1016/j.ultras.2023.107056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/13/2023] [Accepted: 05/23/2023] [Indexed: 06/05/2023]
Abstract
Phase-changing nanodroplets are nanometric sized constructs that can be vaporized via external stimuli, such as focused ultrasound, to generate gaseous bubbles that are visible in ultrasound. Their activation can also be leveraged to release their payload, creating a method for ultrasound-modulated localized drug delivery. Here, we develop a perfluoropentane core nanodroplet that can simultaneously load paclitaxel and doxorubicin, and release them in response to an acoustic trigger. A double emulsion method is used to incorporate the two drugs with different physio-chemical properties, which allows for a combinatorial chemotherapy regimen to be used. Their loading, release, and biological effects on a triple negative breast cancer mouse model are investigated. We show that activation enhances the drug-delivery effect and delays the tumor growth rate in vivo. Overall, the phase-changing nanodroplets are a useful platform to allow on-demand delivery of combinations of drugs.
Collapse
Affiliation(s)
- Catalina-Paula Spatarelu
- Thayer School of Engineering, Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, United States
| | - Sidhartha Jandhyala
- Thayer School of Engineering, Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, United States
| | - Geoffrey P Luke
- Thayer School of Engineering, Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, United States; Translational Engineering in Cancer Research Program, Dartmouth Cancer Center, 1 Medical Center Drive, Lebanon, NH 03766, United States.
| |
Collapse
|
7
|
Plazonic F, LuTheryn G, Hind C, Clifford M, Gray M, Stride E, Glynne-Jones P, Hill M, Sutton JM, Carugo D. Bactericidal Effect of Ultrasound-Responsive Microbubbles and Sub-inhibitory Gentamicin against Pseudomonas aeruginosa Biofilms on Substrates With Differing Acoustic Impedance. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1888-1898. [PMID: 35798625 DOI: 10.1016/j.ultrasmedbio.2022.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
The aim of this research was to explore the interaction between ultrasound-activated microbubbles (MBs) and Pseudomonas aeruginosa biofilms, specifically the effects of MB concentration, ultrasound exposure and substrate properties on bactericidal efficacy. Biofilms were grown using a Centre for Disease Control (CDC) bioreactor on polypropylene or stainless-steel coupons as acoustic analogues for soft and hard tissue, respectively. Biofilms were treated with different concentrations of phospholipid-shelled MBs (107-108 MB/mL), a sub-inhibitory concentration of gentamicin (4 µg/mL) and 1-MHz ultrasound with a continuous or pulsed (100-kHz pulse repetition frequency, 25% duty cycle, 0.5-MPa peak-to-peak pressure) wave. The effect of repeated ultrasound exposure with intervals of either 15- or 60-min was also investigated. With polypropylene coupons, the greatest bactericidal effect was achieved with 2 × 5 min of pulsed ultrasound separated by 60 min and a microbubble concentration of 5 × 107 MBs/mL. A 0.76 log (83%) additional reduction in the number of bacteria was achieved compared with the use of an antibiotic alone. With stainless-steel coupons, a 67% (0.46 log) reduction was obtained under the same exposure conditions, possibly due to enhancement of a standing wave field which inhibited MB penetration in the biofilm. These findings demonstrate the importance of treatment parameter selection in antimicrobial applications of MBs and ultrasound in different tissue environments.
Collapse
Affiliation(s)
- Filip Plazonic
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Gareth LuTheryn
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK; Department of Pharmaceutics, School of Pharmacy, University College London, London, UK; National Biofilms Innovation Centre, University of Southampton, Southampton, UK
| | - Charlotte Hind
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Melanie Clifford
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Michael Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Peter Glynne-Jones
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Martyn Hill
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - J Mark Sutton
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK; Institute of Pharmaceutical Science, King's College London, London, UK
| | - Dario Carugo
- Department of Pharmaceutics, School of Pharmacy, University College London, London, UK.
| |
Collapse
|
8
|
Ultrasound and Microbubbles Enhance Uptake of Doxorubicin in Murine Kidneys. Pharmaceutics 2021; 13:pharmaceutics13122038. [PMID: 34959319 PMCID: PMC8703523 DOI: 10.3390/pharmaceutics13122038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022] Open
Abstract
The use of ultrasound and microbubble-enhanced drug delivery, commonly referred to as sonoporation, has reached numerous clinical trials and has shown favourable results. Nevertheless, the microbubbles and acoustic path also pass through healthy tissues. To date, the majority of studies have focused on the impact to diseased tissues and rarely evaluated the impact on healthy and collateral tissue. The aim of this study was to test the effect and feasibility of low-intensity sonoporation on healthy kidneys in a mouse model. In our work here, we used a clinical diagnostic ultrasound system (GE Vivid E9) with a C1-5 ultrasound transducer combined with a software modification for 20-µs-long pulses to induce the ultrasound-guided drug delivery of doxorubicin (DOX) in mice kidneys in combination with SonoVue® and Sonazoid™ microbubbles. The acoustic output settings were within the commonly used diagnostic ranges. Sonoporation with SonoVue® resulted in a significant decrease in weight vs. DOX alone (p = 0.0004) in the first nine days, whilst all other comparisons were not significant. Ultrasound alone resulted in a 381% increase in DOX uptake vs. DOX alone (p = 0.0004), whilst SonoVue® (p = 0.0001) and Sonazoid™ (p < 0.0001) further increased the uptake nine days after treatment (419% and 493%, respectively). No long-standing damage was observed in the kidneys via histology. In future sonoporation and drug uptake studies, we therefore suggest including an “ultrasound alone” group to verify the actual contribution of the individual components of the procedure on the drug uptake and to perform collateral damage studies to ensure there is no negative impact of low-intensity sonoporation on healthy tissues.
Collapse
|
9
|
Ruan JL, Browning RJ, Yildiz YO, Bau L, Kamila S, Gray MD, Folkes L, Hampson A, McHale AP, Callan JF, Vojnovic B, Kiltie AE, Stride E. Evaluation of Loading Strategies to Improve Tumor Uptake of Gemcitabine in a Murine Orthotopic Bladder Cancer Model Using Ultrasound and Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1596-1615. [PMID: 33707089 DOI: 10.1016/j.ultrasmedbio.2021.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
In this study we compared three different microbubble-based approaches to the delivery of a widely used chemotherapy drug, gemcitabine: (i) co-administration of gemcitabine and microbubbles (Gem+MB); (ii) conjugates of microbubbles and gemcitabine-loaded liposomes (GemlipoMB); and (iii) microbubbles with gemcitabine directly bound to their surfaces (GembioMB). Both in vitro and in vivo investigations were carried out, respectively, in the RT112 bladder cancer cell line and in a murine orthotopic muscle-invasive bladder cancer model. The in vitro (in vivo) ultrasound exposure conditions were a 1 (1.1) MHz centre frequency, 0.07 (1.0) MPa peak negative pressure, 3000 (20,000) cycles and 100 (0.5) Hz pulse repetition frequency. Ultrasound exposure produced no significant increase in drug uptake either in vitro or in vivo compared with the drug-only control for co-administered gemcitabine and microbubbles. In vivo, GemlipoMB prolonged the plasma circulation time of gemcitabine, but only GembioMB produced a statistically significant increase in cleaved caspase 3 expression in the tumor, indicative of gemcitabine-induced apoptosis.
Collapse
Affiliation(s)
- Jia-Ling Ruan
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Richard J Browning
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Yesna O Yildiz
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Sukanta Kamila
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Michael D Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Lisa Folkes
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Alix Hampson
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anthony P McHale
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - John F Callan
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Borivoj Vojnovic
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anne E Kiltie
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
10
|
Vince O, Peeters S, Johanssen VA, Gray M, Smart S, Sibson NR, Stride E. Microbubbles Containing Lysolipid Enhance Ultrasound-Mediated Blood-Brain Barrier Breakdown In Vivo. Adv Healthc Mater 2021; 10:e2001343. [PMID: 33191662 DOI: 10.1002/adhm.202001343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/09/2020] [Indexed: 12/13/2022]
Abstract
Ultrasound and microbubbles (MBs) offer a noninvasive method of temporarily enhancing blood-brain barrier (BBB) permeability to therapeutics. To reduce off-target effects, it is desirable to minimize the ultrasound pressures required. It has been shown that a new formulation of MBs containing lysolipids (Lyso-MBs) can increase the cellular uptake of a model drug in vitro. The aim of this study is to investigate whether Lyso-MBs can also enhance BBB permeability in vivo. Female BALB/c mice are injected with either Lyso-MBs or control MBs and gadolinium-DTPA (Gd-DTPA) and exposed to ultrasound (500 kHz, 1 Hz pulse repetition frequency, 1 ms pulse length, peak-negative pressures 160-480 kPa) for 2 min. BBB permeabilization is measured via magnetic resonance imaging (7.0 T) of Gd-DTPA extravasation and subsequent histological examination of brain tissue to assess serum immunoglobulin G (IgG) extravasation (n = 8 per group). An approximately twofold enhancement in BBB permeability is produced by the Lyso-MBs at the highest ultrasound pressure compared with the control. These findings indicate that modifying the composition of phospholipid-shelled MBs has the potential to improve the efficiency of BBB opening, without increasing the ultrasound pressure amplitude required. This is particularly relevant for delivery of therapeutics deep within the brain.
Collapse
Affiliation(s)
- Oliver Vince
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sarah Peeters
- Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Vanessa A Johanssen
- Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sean Smart
- Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Nicola R Sibson
- Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
11
|
Shan Y, Ji Y, Wang X, He L, Li S. Predicting asymmetric phospholipid microstructures in solutions. RSC Adv 2020; 10:24521-24532. [PMID: 35516199 PMCID: PMC9055179 DOI: 10.1039/d0ra03732j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/18/2020] [Indexed: 01/29/2023] Open
Abstract
Asymmetric phospholipid microstructures, such as asymmetric phospholipid membranes, have potential applications in biological and medicinal processes. Here, we used the dissipative particle dynamics simulation method to predict the asymmetric phospholipid microstructures in aqueous solutions. The asymmetric phospholipid membranes, tubes and vesicles are determined and characterized by the chain density distributions and order parameters. The phase diagrams are constructed to evaluate the effects of the chain length on the asymmetric structure formations at equilibrium states, while the average radius of gyration and shape factors are calculated to analyze the asymmetric structure formations in the non-equilibrium processes. Meanwhile, we predicted the mechanical properties of the asymmetric membranes by analyzing the spatial distributions of the interface tensions and osmotic pressures in solutions.
Collapse
Affiliation(s)
- Yue Shan
- Department of Physics, Wenzhou University Wenzhou Zhejiang 325035 China
| | - Yongyun Ji
- Department of Physics, Wenzhou University Wenzhou Zhejiang 325035 China
| | - Xianghong Wang
- Department of Physics, Wenzhou University Wenzhou Zhejiang 325035 China
| | - Linli He
- Department of Physics, Wenzhou University Wenzhou Zhejiang 325035 China
| | - Shiben Li
- Department of Physics, Wenzhou University Wenzhou Zhejiang 325035 China
| |
Collapse
|
12
|
Zhu Q, Ding W, Li S, Li F, Hu Y, Ya S, Luo T, Gao D, Qiu B. On-Chip Sonoporation-Based Flow Cytometric Magnetic Labeling. ACS Biomater Sci Eng 2020; 6:3187-3196. [PMID: 33463290 DOI: 10.1021/acsbiomaterials.9b01986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tracing magnetically labeled cells with magnetic resonance imaging (MRI) is an emerging and promising approach to uncover in vivo behaviors of cells in cell therapy. Today, existing methods for the magnetic labeling of cells are cumbersome and time-consuming, which has greatly limited the progress of such studies on cell therapy. Thus, in this study, using the flow cytometric loading technology, we develop a sonoporation-based microfluidic chip (i.e., a microfluidic chip integrated with ultrasound; MCU), to achieve the safe, instant, convenient, and continuous magnetic labeling of cells. For the MCU we designed, a suitable group of operating conditions for safely and efficiently loading superparamagnetic iron oxide (SPIO) nanoparticles into DC2.4 cells was identified experimentally. Under the identified operating conditions, the DC2.4 cells could be labeled in approximately 2 min with high viability (94%) and a high labeling quantity of SPIO nanoparticles (19 pg of iron per cell). In addition, the proliferative functions of the cells were also well maintained after labeling. Furthermore, the in vivo imaging ability of the DC2.4 cells labeled using the MCU was verified by injecting the labeled cells into the leg muscle of the C57BL/6 mice. The results show that the excellent imaging outcome can be continuously achieved for 7 days at a density of 106 cells/mL. This work can provide insight for the design of magnetic cell labeling devices and promote the MRI-based study of cell therapies.
Collapse
Affiliation(s)
- Qianwei Zhu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shibo Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Fenfen Li
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yi Hu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shengnan Ya
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Tianzhi Luo
- Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Bensheng Qiu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China.,Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|