1
|
Blöck J, Li H, Collado-Lara G, Kooiman K, Rix A, Chen J, Hark C, Radermacher H, Porte C, Kiessling F. The Compression-Dominated Ultrasound Response of Poly( n-butyl cyanoacrylate) Hard-Shelled Microbubbles Induces Significant Sonoporation and Sonopermeation Effects In Vitro. ACS APPLIED BIO MATERIALS 2025; 8:1240-1250. [PMID: 39900350 PMCID: PMC11836932 DOI: 10.1021/acsabm.4c01551] [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: 10/23/2024] [Revised: 12/23/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025]
Abstract
The process of locally increasing the permeability of cell membranes or cell layers is referred to as sonoporation or sonopermeation, respectively, and opens up perspectives for drug delivery in cancer treatment by facilitating enhanced local drug accumulation. These effects are mediated by ultrasound-activated microbubbles in close proximity to cells. Here, the selection of ultrasound settings according to the intended effect on the biological tissue remains a challenge, especially for broadly size-distributed microbubbles, which show a heterogeneous response to ultrasound. For this purpose, we have analyzed the general response of narrower size-distributed poly(n-butyl cyanoacrylate) hard-shelled microbubbles to ultrasound via ultra-high-speed imaging and evaluated their ability to stimulate sonoporation and sonopermeation in vitro compared to lipid soft-shelled microbubbles. Ultra-high-speed imaging of hard-shelled microbubbles revealed either a compression-dominated or compression-only response at peak negative acoustic pressures higher than 165 kPa and an onset of bursting at 500 kPa. The in vitro experiments demonstrated that the hard-shelled microbubbles induced significant sonoporation and sonopermeation effects, also when only compressing at 300 kPa peak neagtive pressure. Compared to soft-shelled microbubbles, the effects were less prominent, which was attributed to differences in their ultrasound responses and size distributions. This in vitro validation of hard-shelled microbubbles qualifies them for future in vivo applications, which would benefit from their narrow size distribution, thereby allowing more control of their therapeutic effect by suitably adjusting the ultrasound parameters.
Collapse
Affiliation(s)
- Julia Blöck
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| | - Hongchen Li
- Biomedical
Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam , The Netherlands
| | - Gonzalo Collado-Lara
- Biomedical
Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam , The Netherlands
| | - Klazina Kooiman
- Biomedical
Engineering, Department of Cardiology, Cardiovascular Institute, Erasmus MC University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam , The Netherlands
| | - Anne Rix
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| | - Junlin Chen
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| | - Christopher Hark
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| | - Harald Radermacher
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| | - Céline Porte
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| | - Fabian Kiessling
- Institute
for Experimental Molecular Imaging, RWTH
Aachen University Hospital, Forckenbeckstraße 55, 52070 Aachen, Germany
| |
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
|
Gong Z, Mao Y, Liu Y, Hu X, Zhang Y, Zhu L, Guo S, Ding Z, Zhang L. Sono-promoted piezocatalysis and low-dose drug penetration for personalized therapy via tumor organoids. J Colloid Interface Sci 2024; 675:192-206. [PMID: 38968636 DOI: 10.1016/j.jcis.2024.07.010] [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: 04/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Chemotherapy is a widely used cancer treatment, however, it can have notable side effects owing to the high-doses of drugs administered. Sonodynamic therapy (SDT) induced by sonosensitizers has emerged as a promising approach to treat cancer, however, there is limited research evaluating its therapeutic effects on human tumors. In this study, we introduced a dual therapy that combines low-dose chemotherapeutic drugs with enhanced sonodynamic therapy, utilizing barium titanate (BaTiO3, BTO) nanoparticles (NPs) as sonosensitizers to treat tumor organoids. We demonstrated that ultrasound could improve the cellular uptake of chemotherapy drugs, while the chemotherapeutic effect of the drugs made it easier for BTO NPs to enter tumor cells, and the dual therapy synergistically inhibited tumor cell viability. Moreover, different patient-derived tumor organoids exhibited different sensitivities to this therapy, highlighting the potential to evaluate individual responses to combination therapies prior to clinical intervention. Furthermore, this dual therapy exhibited therapeutic effects equivalent to those of high-dose chemotherapy drugs on drug-resistant tumor organoids and showed the potential to enhance the efficacy of killing drug-resistant tumors. In addition, the biosafety of the BTO NPs was successfully verified in live mice via oral administration. This evidence confirms the reliable and safe nature of the dual therapy approach, making it a feasible option for precise and personalized therapy in clinical applications.
Collapse
Affiliation(s)
- Zhiyi Gong
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Jiufengshan Laboratory, Wuhan 430206, China; Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Microelectronics, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Yiqian Mao
- Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Microelectronics, School of Physics and Technology, Wuhan University, Wuhan 430072, China
| | - Yichao Liu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiao Hu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yusen Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lili Zhu
- Hubei Jiufengshan Laboratory, Wuhan 430206, China
| | - Shishang Guo
- Hubei Yangtze Memory Laboratories, Wuhan 430205, China; Key Laboratory of Artificial Micro- and Nano-structures of Ministry of Education, School of Microelectronics, School of Physics and Technology, Wuhan University, Wuhan 430072, China.
| | - Zhao Ding
- Department of Colorectal and Anal Surgery, Hubei Key Laboratory of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Lingling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
4
|
Falatah HA, Lacerda Q, Wessner CE, Lo S, Wheatley MA, Liu JB, Eisenbrey JR. Influence of Phase Change Droplet Activation and Microbubble Cavitation on the Microenvironment of Hepatocellular Carcinoma. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1387-1394. [PMID: 38876912 PMCID: PMC11298311 DOI: 10.1016/j.ultrasmedbio.2024.05.015] [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: 03/15/2024] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVE Both microbubble ultrasound contrast agents and acoustic phase change droplets (APCD) have been explored in hepatocellular carcinoma (HCC). This work aimed to evaluate changes to the HCC microenvironment following either microbubble or APCD destruction in a syngeneic pre-clinical model. METHODS Mouse RIL-175 HCC tumors were grown in the right flank of 64 immunocompetent mice. Pre-treatment, photoacoustic volumetric tumor oxygenation, and power Doppler measurements were obtained using a Vevo 3100 system (VisualSonics, Toronto, Canada). The experimental groups received a 0.1 mL bolus injection of either Definity ultrasound contrast agent (Lantheus Medical Imaging) or APCD fabricated by condensing Definity. Following injection, ultrasound destruction was performed using flash-replenishment sequences on a Sequoia with a 10L4 probe (Siemens) for the duration of enhancement. Tumor oxygenation and power Doppler measurements were then repeated immediately post-ultrasound treatment. Twenty-four hours post-treatment, animals were euthanized, and tumors were harvested and stained for CD31, Cleaved Caspase 3 and CD45. RESULTS Imaging biomarkers demonstrated a significant reduction in percent vascularity following either microbubble or APCD destruction in the tumor microenvironment ( p < 0.022) but no significant changes in tumor oxygenation (p = 0.12). Similarly, immunohistochemistry data demonstrated a significant decrease in CD31 expression (p < 0.042) and an increase in apoptosis (p < 0.014) in tumors treated with destroyed microbubbles or APCD relative to controls. Finally, a significant increase in CD45 expression was observed in tumors treated with APCD (p = 0.046), indicating an increase in tumor immune response. CONCLUSION Ultrasound-triggered destruction of both microbubbles and APCD reduces vascularity, increases apoptosis, and may also increase immune response in this HCC model.
Collapse
Affiliation(s)
- Hebah A Falatah
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA; College of Applied Medical Sciences King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia; King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Quezia Lacerda
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Corinne E Wessner
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Standley Lo
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Margaret A Wheatley
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Ji-Bin Liu
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Armistead FJ, Batchelor DVB, Johnson BRG, Evans SD. QCM-D Investigations on Cholesterol-DNA Tethering of Liposomes to Microbubbles for Therapy. J Phys Chem B 2023; 127:2466-2474. [PMID: 36917458 PMCID: PMC10041634 DOI: 10.1021/acs.jpcb.2c07256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Lipid-shelled microbubbles (MBs) offer potential as theranostic agents, capable of providing both contrast enhancement in ultrasound imaging as well as a route for triggered drug release and improved localized drug delivery. A common motif in the design of such therapeutic vehicles is the attachment of the drug carrier, often in the form of liposomes, to the microbubble. Traditionally, such attachments have been based around biotin-streptavidin and maleimide-PDP chemistries. Comparatively, the use of DNA-lipid tethers offers potential advantage. First, their specificity permits the construction of more complex architectures that might include bespoke combinations of different drug-loaded liposomes and/or targeting groups, such as affimers or antibodies. Second, the use of dual-lipid tether strategies should increase the strength of the individual tethers tethering the liposomes to the bubbles. The ability of cholesterol-DNA (cDNA) tethers for conjugation of liposomes to supported lipid bilayers has previously been demonstrated. For in vivo applications, bubbles and liposomes often contain a proportion of polyethylene glycol (PEG) to promote stealth-like properties and increase lifetimes. However, the associated steric effects may hinder tethering of the drug payload. We show that while the presence of PEG reduced the tethering affinity, cDNA can still be used for the attachment of liposomes to a supported lipid bilayer (SLB) as measured via QCM-D. Importantly, we show, for the first time, that QCM-D can be used to study the tethering of microbubbles to SLBs using cDNA, signified by a decrease in the magnitude of the frequency shift compared to liposomes alone due to the reduced density of the MBs. We then replicate this tethering interaction in the bulk and observe attachment of liposomes to the shell of a central MB and hence formation of a model therapeutic microbubble.
Collapse
Affiliation(s)
- Fern J Armistead
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Damien V B Batchelor
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Benjamin R G Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Stephen D Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
6
|
Bourn MD, Mohajerani SZ, Mavria G, Ingram N, Coletta PL, Evans SD, Peyman SA. Tumour associated vasculature-on-a-chip for the evaluation of microbubble-mediated delivery of targeted liposomes. LAB ON A CHIP 2023; 23:1674-1693. [PMID: 36779251 PMCID: PMC10013341 DOI: 10.1039/d2lc00963c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
The vascular system is the primary route for the delivery of therapeutic drugs throughout the body and is an important barrier at the region of disease interest, such as a solid tumour. The development of complex 3D tumour cultures has progressed significantly in recent years however, the generation of perfusable vascularised tumour models still presents many challenges. This study presents a microfluidic-based vasculature system that can be induced to display properties of tumour-associated blood vessels without direct incorporation of tumour cells. Conditioning healthy endothelial-fibroblast cell vasculature co-cultures with media taken from tumour cell cultures was found to result in the formation of disorganised, tortuous networks which display characteristics consistent with those of tumour-associated vasculature. Integrin αvβ3, a cell adhesion receptor associated with angiogenesis, was found to be upregulated in vasculature co-cultures conditioned with tumour cell media (TCM) - consistent with the reported αvβ3 expression pattern in angiogenic tumour vasculature in vivo. Increased accumulation of liposomes (LSs) conjugated to antibodies against αvβ3 was observed in TCM networks compared to non-conditioned networks, indicating αvβ3 may be a potential target for the delivery of drugs specifically to tumour vasculature. Furthermore, the use of microbubbles (MBs) and ultrasound (US) to further enhance the delivery of LSs to TCM-conditioned vasculature was investigated. Quantification of fluorescent LS accumulation post-perfusion of the vascular network showed 3-fold increased accumulation with the use of MBs and US, suggesting that targeted LS delivery could be further improved with the use of locally administered MBs and US.
Collapse
Affiliation(s)
- Matthew D Bourn
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Safoura Zahed Mohajerani
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Georgia Mavria
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Nicola Ingram
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - P Louise Coletta
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Stephen D Evans
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
| | - Sally A Peyman
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| |
Collapse
|
7
|
Khodabakhshi Z, Hosseinkhah N, Ghadiri H. Pulsating Microbubble in a Micro-vessel and Mechanical Effect on Vessel Wall: A Simulation Study. J Biomed Phys Eng 2021; 11:629-640. [PMID: 34722408 PMCID: PMC8546166 DOI: 10.31661/jbpe.v0i0.1131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/18/2019] [Indexed: 11/21/2022]
Abstract
Background: Microbubbles are widely used in diagnostic ultrasound applications as contrast agents. Recently, many studies have shown that microbubbles have
good potential for the use in therapeutic applications such as drug and gene delivery and opening of blood- brain barrier locally and transiently.
When microbubbles are located inside an elastic microvessel and activated by ultrasound, they oscillate and induce mechanical stresses on the vessel wall.
However, the mechanical stresses have beneficial therapeutic effects, they may induce vessel damage if they are too high. Microstreaming-induced
shear stress is one of the most important wall stresses. Objective: The overall aim of this study is to simulate the interaction between confined bubble inside an elastic microvessel and ultrasound field
and investigate the effective parameters on microstreaming-induced shear stress. Material and Methods: In this Simulation study, we conducted a 2D finite element simulation to study confined microbubble dynamics, also we investigated both
acoustical and bubble material parameters on microbubble oscillation and wall stress. Results: Based on our results, for acoustic parameters in the range of therapeutic applications, the maximum shear stress was lower than 4 kPa.
Shear stress was approximately independent from shell viscosity whereas it decreased by increasing the shell stiffness.
Moreover, shear stress showed an increasing trend with acoustic pressure. Conclusion: Beside the acoustical parameters, bubble properties have important effects on bubble behavior so that the softer and larger bubbles are
more appropriate for therapeutic application as they can decrease the required frequency and acoustic pressure while inducing the same biological effects.
Collapse
Affiliation(s)
- Zahra Khodabakhshi
- MSc, Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Science, Tehran, Iran
- MSc, Research Center for Molecular and Cellular Imaging (RCMCI), Tehran, Iran
| | - Nazanin Hosseinkhah
- PhD, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Hossein Ghadiri
- PhD, Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Science, Tehran, Iran
- PhD, Research Center for Molecular and Cellular Imaging (RCMCI), Tehran, Iran
| |
Collapse
|
8
|
Ultrasound-Enabled Therapeutic Delivery and Regenerative Medicine: Physical and Biological Perspectives. ACS Biomater Sci Eng 2021; 7:4371-4387. [PMID: 34460238 DOI: 10.1021/acsbiomaterials.1c00276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of ultrasound in medicine and biological sciences is expanding rapidly beyond its use in conventional diagnostic imaging. Numerous studies have reported the effects of ultrasound on cellular and tissue physiology. Advances in instrumentation and electronics have enabled successful in vivo applications of therapeutic ultrasound. Despite path breaking advances in understanding the biophysical and biological mechanisms at both microscopic and macroscopic scales, there remain substantial gaps. With the progression of research in this area, it is important to take stock of the current understanding of the field and to highlight important areas for future work. We present herein key developments in the biological applications of ultrasound especially in the context of nanoparticle delivery, drug delivery, and regenerative medicine. We conclude with a brief perspective on the current promise, limitations, and future directions for interfacing ultrasound technology with biological systems, which could provide guidance for future investigations in this interdisciplinary area.
Collapse
|
9
|
Batchelor DV, Armistead FJ, Ingram N, Peyman SA, Mclaughlan JR, Coletta PL, Evans SD. Nanobubbles for therapeutic delivery: Production, stability and current prospects. Curr Opin Colloid Interface Sci 2021. [DOI: 10.1016/j.cocis.2021.101456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Grisanti G, Caprini D, Sinibaldi G, Scognamiglio C, Silvani G, Peruzzi G, Casciola CM. A Microfluidic Platform for Cavitation-Enhanced Drug Delivery. MICROMACHINES 2021; 12:mi12060658. [PMID: 34204968 PMCID: PMC8229805 DOI: 10.3390/mi12060658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/30/2021] [Accepted: 05/30/2021] [Indexed: 11/17/2022]
Abstract
An endothelial-lined blood vessel model is obtained in a PDMS (Polydimethylsiloxane) microfluidic system, where vascular endothelial cells are grown under physiological shear stress, allowing -like maturation. This experimental model is employed for enhanced drug delivery studies, aimed at characterising the increase in endothelial permeability upon microbubble-enhanced ultrasound-induced (USMB) cavitation. We developed a multi-step protocol to couple the optical and the acoustic set-ups, thanks to a 3D-printed insonation chamber, provided with direct optical access and a support for the US transducer. Cavitation-induced interendothelial gap opening is then analysed using a customised code that quantifies gap area and the relative statistics. We show that exposure to US in presence of microbubbles significantly increases endothelial permeability and that tissue integrity completely recovers within 45 min upon insonation. This protocol, along with the versatility of the microfluidic platform, allows to quantitatively characterise cavitation-induced events for its potential employment in clinics.
Collapse
Affiliation(s)
- Giulia Grisanti
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00186 Roma, Italy; (G.G.); (G.S.); (G.S.)
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Via Regina Elena 291, 00161 Roma, Italy; (D.C.); (C.S.)
| | - Davide Caprini
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Via Regina Elena 291, 00161 Roma, Italy; (D.C.); (C.S.)
| | - Giorgia Sinibaldi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00186 Roma, Italy; (G.G.); (G.S.); (G.S.)
| | - Chiara Scognamiglio
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Via Regina Elena 291, 00161 Roma, Italy; (D.C.); (C.S.)
| | - Giulia Silvani
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00186 Roma, Italy; (G.G.); (G.S.); (G.S.)
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Via Regina Elena 291, 00161 Roma, Italy; (D.C.); (C.S.)
- School of Biomedical Engineering, Faculty of Engineering & Information Technology, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Giovanna Peruzzi
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Via Regina Elena 291, 00161 Roma, Italy; (D.C.); (C.S.)
- Correspondence: (G.P.); (C.M.C.)
| | - Carlo Massimo Casciola
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00186 Roma, Italy; (G.G.); (G.S.); (G.S.)
- Center for Life Nano- & Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Via Regina Elena 291, 00161 Roma, Italy; (D.C.); (C.S.)
- Correspondence: (G.P.); (C.M.C.)
| |
Collapse
|
11
|
Pouliopoulos AN, Smith CAB, Bezer JH, El Ghamrawy A, Sujarittam K, Bouldin CJ, Morse SV, Tang MX, Choi JJ. Doppler Passive Acoustic Mapping. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:2692-2703. [PMID: 32746222 DOI: 10.1109/tuffc.2020.3011657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In therapeutic ultrasound using microbubbles, it is essential to drive the microbubbles into the correct type of activity and the correct location to produce the desired biological response. Although passive acoustic mapping (PAM) is capable of locating where microbubble activities are generated, it is well known that microbubbles rapidly move within the ultrasound beam. We propose a technique that can image microbubble movement by estimating their velocities within the focal volume. Microbubbles embedded within a wall-less channel of a tissue-mimicking material were sonicated using 1-MHz focused ultrasound. The acoustic emissions generated by the microbubbles were captured with a linear array (L7-4). PAM with robust Capon beamforming was used to localize the microbubble acoustic emissions. We spectrally analyzed the time trace of each position and isolated the higher harmonics. Microbubble velocity maps were constructed from the position-dependent Doppler shifts at different time points during sonication. Microbubbles moved primarily away from the transducer at velocities on the order of 1 m/s due to primary acoustic radiation forces, producing a time-dependent velocity distribution. We detected microbubble motion both away and toward the receiving array, revealing the influence of acoustic radiation forces and fluid motion due to the ultrasound exposure. High-speed optical images confirmed the acoustically measured microbubble velocities. Doppler PAM enables passive estimation of microbubble motion and may be useful in therapeutic applications, such as drug delivery across the blood-brain barrier, sonoporation, sonothrombolysis, and drug release.
Collapse
|
12
|
Presset A, Bonneau C, Kazuyoshi S, Nadal-Desbarats L, Mitsuyoshi T, Bouakaz A, Kudo N, Escoffre JM, Sasaki N. Endothelial Cells, First Target of Drug Delivery Using Microbubble-Assisted Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1565-1583. [PMID: 32331799 DOI: 10.1016/j.ultrasmedbio.2020.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 06/11/2023]
Abstract
Microbubble-assisted ultrasound has emerged as a promising method for local drug delivery. Microbubbles are intravenously injected and locally activated by ultrasound, thus increasing the permeability of vascular endothelium for facilitating extravasation and drug uptake into the treated tissue. Thereby, endothelial cells are the first target of the effects of ultrasound-driven microbubbles. In this review, the in vitro and in vivo bioeffects of this method on endothelial cells are described and discussed, including aspects on the permeabilization of biologic barriers (endothelial cell plasma membranes and endothelial barriers), the restoration of their integrity, the molecular and cellular mechanisms involved in both these processes, and the resulting intracellular and intercellular consequences. Finally, the influence of the acoustic settings, microbubble parameters, treatment schedules and flow parameters on these bioeffects are also reviewed.
Collapse
Affiliation(s)
- Antoine Presset
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | - Sasaoka Kazuyoshi
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | - Takigucho Mitsuyoshi
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Nobuki Kudo
- Laboratory of Biological Engineering, Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | | | - Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Department of Clinical Sciences; Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
13
|
Linz G, Djeljadini S, Steinbeck L, Köse G, Kiessling F, Wessling M. Cell barrier characterization in transwell inserts by electrical impedance spectroscopy. Biosens Bioelectron 2020; 165:112345. [PMID: 32513645 DOI: 10.1016/j.bios.2020.112345] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/12/2020] [Accepted: 05/31/2020] [Indexed: 11/19/2022]
Abstract
We describe an impedance-based method for cell barrier integrity testing. A four-electrode electrical impedance spectroscopy (EIS) setup can be realized by simply connecting a commercial chopstick-like electrode (STX-1) to a potentiostat allowing monitoring cell barriers cultivated in transwell inserts. Subsequent electric circuit modeling of the electrical impedance results the capacitive properties of the barrier next to the well-known transepithelial electrical resistance (TEER). The versatility of the new method was analyzed by the EIS analysis of a Caco-2 monolayer in response to (a) different membrane coating materials, (b) two different permeability enhancers ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid (EGTA) and saponin, and (c) sonoporation. For the different membrane coating materials, the TEERs of the standard and new protocol coincide and increase during cultivation, while the capacitance shows a distinct maximum for three different surface materials (no coating, Matrigel®, and collagen I). The permeability enhancers cause a decline in the TEER value, but only saponin alters the capacitance of the cell layer by two orders of magnitude. Hence, cell layer capacitance and TEER represent two independent properties characterizing the monolayer. The use of commercial chopstick-like electrodes to access the impedance of a barrier cultivated in transwell inserts enables remarkable insight into the behavior of the cellular barrier with no extra work for the researcher. This simple method could evolve into a standard protocol used in cell barrier research.
Collapse
Affiliation(s)
- Georg Linz
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany; RWTH Aachen University, Aachener Verfahrenstechnik-Chemical Process Engineering, Forckenbeckstrasse 51, 52074, Aachen, Germany
| | - Suzana Djeljadini
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany; RWTH Aachen University, Aachener Verfahrenstechnik-Chemical Process Engineering, Forckenbeckstrasse 51, 52074, Aachen, Germany
| | - Lea Steinbeck
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany; RWTH Aachen University, Aachener Verfahrenstechnik-Chemical Process Engineering, Forckenbeckstrasse 51, 52074, Aachen, Germany
| | - Gurbet Köse
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Matthias Wessling
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany; RWTH Aachen University, Aachener Verfahrenstechnik-Chemical Process Engineering, Forckenbeckstrasse 51, 52074, Aachen, Germany.
| |
Collapse
|
14
|
Ciancia S, Cafarelli A, Zahoranova A, Menciassi A, Ricotti L. Pulsatile Drug Delivery System Triggered by Acoustic Radiation Force. Front Bioeng Biotechnol 2020; 8:317. [PMID: 32411680 PMCID: PMC7202567 DOI: 10.3389/fbioe.2020.00317] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/24/2020] [Indexed: 11/13/2022] Open
Abstract
Since biological systems exhibit a circadian rhythm (24-hour cycle), they are susceptible to the timing of drug administration. Indeed, several disorders require a therapy that synchronizes with the onset of symptoms. A targeted therapy with spatially and temporally precise controlled drug release can guarantee a considerable gain in terms of efficacy and safety of the treatment compared to traditional pharmacological methods, especially for chronotherapeutic disorders. This paper presents a proof of concept of an innovative pulsatile drug delivery system remotely triggered by the acoustic radiation force of ultrasound. The device consists of a case, in which a drug-loaded gel can be embedded, and a sliding top that can be moved on demand by the application of an acoustic stimulus, thus enabling drug release. Results demonstrate for the first time that ultrasound acoustic radiation force (up to 0.1 N) can be used for an efficient pulsatile drug delivery (up to 20 μg of drug released for each shot).
Collapse
Affiliation(s)
- Sabrina Ciancia
- The BioRobotics Institute, Sant'Anna School of Advanced Studies, Pisa, Italy.,Departments of Excellence, Robotics & AI, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Andrea Cafarelli
- The BioRobotics Institute, Sant'Anna School of Advanced Studies, Pisa, Italy.,Departments of Excellence, Robotics & AI, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Anna Zahoranova
- Department for Biomaterials Research, Polymer Institute SAS, Bratislava, Slovakia
| | - Arianna Menciassi
- The BioRobotics Institute, Sant'Anna School of Advanced Studies, Pisa, Italy.,Departments of Excellence, Robotics & AI, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Sant'Anna School of Advanced Studies, Pisa, Italy.,Departments of Excellence, Robotics & AI, Sant'Anna School of Advanced Studies, Pisa, Italy
| |
Collapse
|
15
|
Mannaris C, Yang C, Carugo D, Owen J, Lee JY, Nwokeoha S, Seth A, Teo BM. Acoustically responsive polydopamine nanodroplets: A novel theranostic agent. ULTRASONICS SONOCHEMISTRY 2020; 60:104782. [PMID: 31539725 DOI: 10.1016/j.ultsonch.2019.104782] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/21/2019] [Accepted: 09/06/2019] [Indexed: 05/06/2023]
Abstract
Ultrasound-induced cavitation has been used as a tool of enhancing extravasation and tissue penetration of anticancer agents in tumours. Initiating cavitation in tissue however, requires high acoustic intensities that are neither safe nor easy to achieve with current clinical systems. The use of cavitation nuclei can however lower the acoustic intensities required to initiate cavitation and the resulting bio-effects in situ. Microbubbles, solid gas-trapping nanoparticles, and phase shift nanodroplets are some examples in a growing list of proposed cavitation nuclei. Besides the ability to lower the cavitation threshold, stability, long circulation times, biocompatibility and biodegradability, are some of the desirable characteristics that a clinically applicable cavitation agent should possess. In this study, we present a novel formulation of ultrasound-triggered phase transition sub-micrometer sized nanodroplets (~400 nm) stabilised with a biocompatible polymer, polydopamine (PDA). PDA offers some important benefits: (1) facile fabrication, as dopamine monomers are directly polymerised on the nanodroplets, (2) high polymer biocompatibility, and (3) ease of functionalisation with other molecules such as drugs or targeting species. We demonstrate that the acoustic intensities required to initiate inertial cavitation can all be achieved with existing clinical ultrasound systems. Cell viability and haemolysis studies show that nanodroplets are biocompatible. Our results demonstrate the great potential of PDA nanodroplets as an acoustically active nanodevice, which is highly valuable for biomedical applications including drug delivery and treatment monitoring.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK.
| | - Chuanxu Yang
- Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China
| | - Dario Carugo
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK; Mechatronics and Bioengineering Science Research Groups, Faculty of Engineering and the Environment, University of Southampton, Southampton, UK
| | - Joshua Owen
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Jeong Yu Lee
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Sandra Nwokeoha
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Anjali Seth
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Boon Mian Teo
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK; Key Laboratory of Colloid and Interface Chemistry, Ministry of Education, Shandong University, Jinan 250100, China; School of Chemistry, Clayton Campus, Monash University Victoria, 3800, Australia.
| |
Collapse
|
16
|
Silvani G, Scognamiglio C, Caprini D, Marino L, Chinappi M, Sinibaldi G, Peruzzi G, Kiani MF, Casciola CM. Reversible Cavitation-Induced Junctional Opening in an Artificial Endothelial Layer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1905375. [PMID: 31762158 DOI: 10.1002/smll.201905375] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Indexed: 06/10/2023]
Abstract
Targeting pharmaceuticals through the endothelial barrier is crucial for drug delivery. In this context, cavitation-assisted permeation shows promise for effective and reversible opening of intercellular junctions. A vessel-on-a-chip is exploited to investigate and quantify the effect of ultrasound-excited microbubbles-stable cavitation-on endothelial integrity. In the vessel-on-a-chip, the endothelial cells form a complete lumen under physiological shear stress, resulting in intercellular junctions that exhibit barrier functionality. Immunofluorescence microscopy is exploited to monitor vascular integrity following vascular endothelial cadherin staining. It is shown that microbubbles amplify the ultrasound effect, leading to the formation of interendothelial gaps that cause barrier permeabilization. The total gap area significantly increases with pressure amplitude compared to the control. Gap opening is fully reversible with gap area distribution returning to the control levels 45 min after insonication. The proposed integrated platform allows for precise and repeatable in vitro measurements of cavitation-enhanced endothelium permeability and shows potential for validating irradiation protocols for in vivo applications.
Collapse
Affiliation(s)
- Giulia Silvani
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Chiara Scognamiglio
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Davide Caprini
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Luca Marino
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
| | - Mauro Chinappi
- Department of Industrial Engineering, Università di Roma Tor Vergata, Via del Politecnico 1, 00133, Rome, Italy
| | - Giorgia Sinibaldi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Mohammad F Kiani
- Department of Mechanical Engineering, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Carlo M Casciola
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Via Eudossiana 18, 00184, Rome, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| |
Collapse
|
17
|
Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: Designing a new perspective in nanomedicine delivery. Theranostics 2019; 9:8091-8108. [PMID: 31754383 PMCID: PMC6857058 DOI: 10.7150/thno.37204] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
The enhanced permeability and retention (EPR) effect has underlain the predominant nanomedicine design philosophy for the past three decades. However, growing evidence suggests that it is over-represented in preclinical models, and agents designed solely using its principle of passive accumulation can only be applied to a narrow subset of clinical tumors. For this reason, strategies that can improve upon the EPR effect to facilitate nanomedicine delivery to otherwise non-responsive tumors are required for broad clinical translation. EPR-adaptive nanomedicine delivery comprises a class of chemical and physical techniques that modify tumor accessibility in an effort to increase agent delivery and therapeutic effect. In the present review, we overview the primary benefits and limitations of radiation, ultrasound, hyperthermia, and photodynamic therapy as physical strategies for EPR-adaptive delivery to EPR-insensitive tumor phenotypes, and we reflect upon changes in the preclinical research pathway that should be implemented in order to optimally validate and develop these delivery strategies.
Collapse
Affiliation(s)
- Alexander Dhaliwal
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- MD/PhD Program, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON Canada
| |
Collapse
|
18
|
Roovers S, Segers T, Lajoinie G, Deprez J, Versluis M, De Smedt SC, Lentacker I. The Role of Ultrasound-Driven Microbubble Dynamics in Drug Delivery: From Microbubble Fundamentals to Clinical Translation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10173-10191. [PMID: 30653325 DOI: 10.1021/acs.langmuir.8b03779] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In the last couple of decades, ultrasound-driven microbubbles have proven excellent candidates for local drug delivery applications. Besides being useful drug carriers, microbubbles have demonstrated the ability to enhance cell and tissue permeability and, as a consequence, drug uptake herein. Notwithstanding the large amount of evidence for their therapeutic efficacy, open issues remain. Because of the vast number of ultrasound- and microbubble-related parameters that can be altered and the variability in different models, the translation from basic research to (pre)clinical studies has been hindered. This review aims at connecting the knowledge gained from fundamental microbubble studies to the therapeutic efficacy seen in in vitro and in vivo studies, with an emphasis on a better understanding of the response of a microbubble upon exposure to ultrasound and its interaction with cells and tissues. More specifically, we address the acoustic settings and microbubble-related parameters (i.e., bubble size and physicochemistry of the bubble shell) that play a key role in microbubble-cell interactions and in the associated therapeutic outcome. Additionally, new techniques that may provide additional control over the treatment, such as monodisperse microbubble formulations, tunable ultrasound scanners, and cavitation detection techniques, are discussed. An in-depth understanding of the aspects presented in this work could eventually lead the way to more efficient and tailored microbubble-assisted ultrasound therapy in the future.
Collapse
Affiliation(s)
- Silke Roovers
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| | - Tim Segers
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center , University of Twente , P.O. Box 217, 7500 AE Enschede , The Netherlands
| | - Guillaume Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center , University of Twente , P.O. Box 217, 7500 AE Enschede , The Netherlands
| | - Joke Deprez
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| | - Michel Versluis
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center , University of Twente , P.O. Box 217, 7500 AE Enschede , The Netherlands
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| |
Collapse
|
19
|
Effect of drug-loaded microbubbles combined with ultrasound on the apoptosis of cancer cells and the expression of Bax and Bcl-2 in a rabbit VX2 liver tumor model. Biosci Rep 2019; 39:BSR20181144. [PMID: 30578377 PMCID: PMC6533209 DOI: 10.1042/bsr20181144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/02/2018] [Accepted: 12/17/2018] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate whether the use of drug-loaded microbubbles combined with ultrasound promotes the apoptosis of cancer cells by regulating B-cell lymphoma-2 (Bcl-2) and Bcl-2-associated X protein (Bax) expression. Adriamycin-loaded PLGA nanoparticles (ADM-NP) were fabricated using a modified emulsification process. Lipid microbubbles (NH2-MB) were prepared by mechanical vibration. The carboxyl groups of ADM-NP and NH2-MB underwent a condensation reaction after 48 h, and adriamycin-loaded PLGA nanoparticles microbubble complexes (ADM-NMC) were obtained. High-performance liquid chromatography demonstrated that the entrapment efficiency and drug loading of ADM-NMC were 85.32 ± 5.41% and 7.91 ± 0.27%, respectively. The VX2 liver cancer model was established in 30 New Zealand rabbits, which were subsequently divided into three groups (n=10): a control group that received 5 ml of saline, an ADM-NP group that received 5 ml of ADM-NP and an ADM-NMC group that received 5 ml of ADM-NMC. Rabbits in the ADM-NP and ADM-NMC groups underwent irradiation 120 s with low frequency ultrasound (1 MHz, 0.5 W/cm2) for 120 s following injection. The echogenicity of tumors markedly increased following ADM-NP and ADM-NMC treatment. Staining with hematoxylin and eosin demonstrated that the tumor shape became more normal in the ADM-NP and ADM-NMC groups compared with the control group. Immunohistochemical staining and Western blotting determined that the expression of Bax increased and the expression of Bcl-2 decreased following treatment with ADM-NP and ADM-NMC. Cancer cell apoptosis was detected by flow cytometry and it was determined that apoptosis significantly increased following treatment with ADM-NP and ADM-NMC (P<0.01). Therefore, the present study demonstrated that the use of drug-loaded microbubbles combined with ultrasound may enhance the efficiency of tumor inhibition. This may be due to the promotion of cancer cell apoptosis via regulation of Bax and Bcl-2 expression.
Collapse
|
20
|
Mannaris C, Bau L, Grundy M, Gray M, Lea-Banks H, Seth A, Teo B, Carlisle R, Stride E, Coussios CC. Microbubbles, Nanodroplets and Gas-Stabilizing Solid Particles for Ultrasound-Mediated Extravasation of Unencapsulated Drugs: An Exposure Parameter Optimization Study. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:954-967. [PMID: 30655109 DOI: 10.1016/j.ultrasmedbio.2018.10.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 06/09/2023]
Abstract
Ultrasound-induced cavitation has been proposed as a strategy to tackle the challenge of inadequate extravasation, penetration and distribution of therapeutics into tumours. Here, the ability of microbubbles, droplets and solid gas-trapping particles to facilitate mass transport and extravasation of a model therapeutic agent following ultrasound-induced cavitation is investigated. Significant extravasation and penetration depths on the order of millimetres are achieved with all three agents, including the range of pressures and frequencies achievable with existing clinical ultrasound systems. Deeper but highly directional extravasation was achieved with frequencies of 1.6 and 3.3 MHz compared with 0.5 MHz. Increased extravasation was observed with increasing pulse length and exposure time, while an inverse relationship is observed with pulse repetition frequency. No significant cell death or any haemolytic activity in human blood was observed at clinically relevant concentrations for any of the agents. Overall, solid gas-trapping nanoparticles were found to enable the most extensive extravasation for the lowest input acoustic energy, followed by microbubbles and then droplets. The ability of these agents to produce sustained inertial cavitation activity whilst being small enough to follow the drug out of the circulation and into diseased tissue, combined with a good safety profile and the possibility of real-time monitoring, offers considerable potential for enhanced drug delivery of unmodified drugs in oncological and other biomedical applications.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Megan Grundy
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Harriet Lea-Banks
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Anjali Seth
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Boon Teo
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Constantin C Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom.
| |
Collapse
|
21
|
Zhang J, Song L, Zhou S, Hu M, Jiao Y, Teng Y, Wang Y, Zhang X. Enhanced ultrasound imaging and anti-tumor in vivo properties of Span–polyethylene glycol with folic acid–carbon nanotube–paclitaxel multifunctional microbubbles. RSC Adv 2019; 9:35345-35355. [PMID: 35528086 PMCID: PMC9074749 DOI: 10.1039/c9ra06437k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 09/27/2019] [Indexed: 11/21/2022] Open
Abstract
With Span and polyethylene glycol (PEG) as the membrane material, the as-prepared folate–carbon nanotube–paclitaxel (FA–CNT–PTX) complex was added to the reaction system under sound vibration cavitation and Span–PEG with FA–CNT–PTX microbubbles was obtained.
Collapse
Affiliation(s)
- Jie Zhang
- Pharmacy College
- Jiamusi University
- Jiamusi 154007
- China
| | - Limei Song
- Pharmacy College
- Jiamusi University
- Jiamusi 154007
- China
| | - Shujing Zhou
- Pharmacy College
- Jiamusi University
- Jiamusi 154007
- China
| | - Ming Hu
- College of Materials Science & Engineering
- Jiamusi University
- Jiamusi 154007
- China
| | - Yufeng Jiao
- College of Materials Science & Engineering
- Jiamusi University
- Jiamusi 154007
- China
| | - Yang Teng
- Pharmacy College
- Jiamusi University
- Jiamusi 154007
- China
| | - Ying Wang
- Pharmacy College
- Jiamusi University
- Jiamusi 154007
- China
| | - Xiangyu Zhang
- Pharmacy College
- Jiamusi University
- Jiamusi 154007
- China
| |
Collapse
|
22
|
Mannaris C, Teo BM, Seth A, Bau L, Coussios C, Stride E. Gas-Stabilizing Gold Nanocones for Acoustically Mediated Drug Delivery. Adv Healthc Mater 2018; 7:e1800184. [PMID: 29696808 DOI: 10.1002/adhm.201800184] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/15/2018] [Indexed: 01/27/2023]
Abstract
The efficient penetration of drugs into tumors is a major challenge that remains unmet. Reported herein is a strategy to promote extravasation and enhanced penetration using inertial cavitation initiated by focused ultrasound and cone-shaped gold nanoparticles that entrap gas nanobubbles. The cones are capable of initiating inertial cavitation under pressures and frequencies achievable with existing clinical ultrasound systems and of promoting extravasation and delivery of a model large therapeutic molecule in an in vitro tissue mimicking flow phantom, achieving penetration depths in excess of 2 mm. Ease of functionalization and intrinsic imaging capabilities provide gold with significant advantages as a material for biomedical applications. The cones show neither cytotoxicity in Michigan Cancer Foundation (MCF)-7 cells nor hemolytic activity in human blood at clinically relevant concentrations and are found to be colloidally stable for at least 5 d at 37 °C and several months at 4 °C.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Boon M Teo
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
- Interdisciplinary Nanoscience Center (iNANO), The iNANO House, Aarhus University, Gustav Wieds Vej 14, DK-8000, Aarhus C, Denmark
- School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, VIC, 3800, Australia
| | - Anjali Seth
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Luca Bau
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
23
|
Eisenbrey JR, Shraim R, Liu JB, Li J, Stanczak M, Oeffinger B, Leeper DB, Keith SW, Jablonowski LJ, Forsberg F, O'Kane P, Wheatley MA. Sensitization of Hypoxic Tumors to Radiation Therapy Using Ultrasound-Sensitive Oxygen Microbubbles. Int J Radiat Oncol Biol Phys 2018; 101:88-96. [PMID: 29477294 PMCID: PMC5886808 DOI: 10.1016/j.ijrobp.2018.01.042] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/05/2017] [Accepted: 01/10/2018] [Indexed: 10/18/2022]
Abstract
PURPOSE Much of the volume of solid tumors typically exists in a chronically hypoxic microenvironment that has been shown to result in both chemotherapy and radiation therapy resistance. The purpose of this study was to use localized microbubble delivery to overcome hypoxia prior to therapy. MATERIALS AND METHODS In this study, surfactant-shelled oxygen microbubbles were fabricated and injected intravenously to locally elevate tumor oxygen levels when triggered by noninvasive ultrasound in mice with human breast cancer tumors. Changes in oxygen and sensitivity to radiation therapy were then measured. RESULTS In this work, we show that oxygen-filled microbubbles successfully and consistently increase breast tumor oxygenation levels in a murine model by 20 mmHg, significantly more than control injections of saline solution or untriggered oxygen microbubbles (P < .001). Using photoacoustic imaging, we also show that oxygen delivery is independent of hemoglobin transport, enabling oxygen delivery to avascular regions of the tumor. Finally, we show that overcoming hypoxia by this method immediately prior to radiation therapy nearly triples radiosensitivity. This improvement in radiosensitivity results in roughly 30 days of improved tumor control, providing statistically significant improvements in tumor growth and animal survival (P < .03). CONCLUSIONS Our findings demonstrate the potential advantages of ultrasound-triggered oxygen delivery to solid tumors and warrant future efforts into clinical translation of the microbubble platform.
Collapse
Affiliation(s)
- John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania.
| | - Rawan Shraim
- School of Biomedical Engineering and Health Sciences, Drexel University, Philadelphia, Pennsylvania
| | - Ji-Bin Liu
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jingzhi Li
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Maria Stanczak
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Brian Oeffinger
- School of Biomedical Engineering and Health Sciences, Drexel University, Philadelphia, Pennsylvania
| | - Dennis B Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Scott W Keith
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lauren J Jablonowski
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Patrick O'Kane
- Department of Radiology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Margaret A Wheatley
- School of Biomedical Engineering and Health Sciences, Drexel University, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Beekers I, van Rooij T, Verweij MD, Versluis M, de Jong N, Trietsch SJ, Kooiman K. Acoustic Characterization of a Vessel-on-a-Chip Microfluidic System for Ultrasound-Mediated Drug Delivery. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2018; 65:570-581. [PMID: 29610087 DOI: 10.1109/tuffc.2018.2803137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ultrasound in the presence of gas-filled microbubbles can be used to enhance local uptake of drugs and genes. To study the drug delivery potential and its underlying physical and biological mechanisms, an in vitro vessel model should ideally include 3-D cell culture, perfusion flow, and membrane-free soft boundaries. Here, we propose an organ-on-a-chip microfluidic platform to study ultrasound-mediated drug delivery: the OrganoPlate. The acoustic propagation into the OrganoPlate was determined to assess the feasibility of controlled microbubble actuation, which is required to study the microbubble-cell interaction for drug delivery. The pressure field in the OrganoPlate was characterized non-invasively by studying experimentally the well-known response of microbubbles and by simulating the acoustic wave propagation in the system. Microbubble dynamics in the OrganoPlate were recorded with the Brandaris 128 ultrahigh-speed camera (17 million frames/s) and a control experiment was performed in an OptiCell, an in vitro monolayer cell culture chamber that is conventionally used to study ultrasound-mediated drug delivery. When insonified at frequencies between 1 and 2 MHz, microbubbles in the OrganoPlate experienced larger oscillation amplitudes resulting from higher local pressures. Microbubbles responded similarly in both systems when insonified at frequencies between 2 and 4 MHz. Numerical simulations performed with a 3-D finite-element model of ultrasound propagation into the OrganoPlate and the OptiCell showed the same frequency-dependent behavior. The predictable and homogeneous pressure field in the OrganoPlate demonstrates its potential to develop an in vitro 3-D cell culture model, well suited to study ultrasound-mediated drug delivery.
Collapse
|
25
|
Peruzzi G, Sinibaldi G, Silvani G, Ruocco G, Casciola CM. Perspectives on cavitation enhanced endothelial layer permeability. Colloids Surf B Biointerfaces 2018; 168:83-93. [PMID: 29486912 DOI: 10.1016/j.colsurfb.2018.02.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 12/20/2022]
Abstract
Traditional drug delivery systems, where pharmaceutical agents are conveyed to the target tissue through the blood circulation, suffer of poor therapeutic efficiency and limited selectivity largely due to the low permeability of the highly specialised biological interface represented by the endothelial layer. Examples concern cancer therapeutics or degenerative disorders where drug delivery is inhibited by the blood-brain barrier (BBB). Microbubbles injected into the bloodstream undergo volume oscillations under localised ultrasound irradiation and possibly collapse near the site of interest, with no effect on the rest of the endothelium. The resulting mechanical action induces a transient increase of the inter-cellular spaces and facilitates drug extravasation. This approach, already pursed in in vivo animal models, is extremely expensive and time-consuming. On the other hand in vitro studies using different kinds of microfluidic networks are firmly established in the pharmaceutical industry for drug delivery testing. The combination of the in vitro approach with ultrasound used to control microbubbles oscillations is expected to provide crucial information for developing cavitation enhanced drug delivery protocols and for screening the properties of the biological interface in presence of healthy or diseased tissues. Purpose of the present review is providing the state of the art in this rapidly growing field where cavitation is exploited as a viable technology to transiently modify the permeability of the biological interface. After describing current in vivo studies, particular emphasis will be placed on illustrating characteristics of micro-devices, biological functionalisation, properties of the artificial endothelium and ultrasound irradiation techniques.
Collapse
Affiliation(s)
- Giovanna Peruzzi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giorgia Sinibaldi
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Italy
| | - Giulia Silvani
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy; Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy; Department of Physics, Sapienza University of Rome, Italy.
| | - Carlo Massimo Casciola
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy; Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Italy
| |
Collapse
|
26
|
Cao S, Zhou Q, Chen JL, Jiang N, Wang YJ, Deng Q, Hu B, Guo RQ. Enhanced effect of nuclear localization signal peptide during ultrasound‑targeted microbubble destruction‑mediated gene transfection. Mol Med Rep 2017; 16:565-572. [PMID: 28586046 PMCID: PMC5482142 DOI: 10.3892/mmr.2017.6661] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/03/2017] [Indexed: 11/06/2022] Open
Abstract
Ultrasound‑targeted microbubble destruction (UTMD) can promote the entry of plasmid DNA (pDNA) into the cell cytoplasm, by increasing the permeability of the cell membrane. But the transfection efficiency remains low due to inability of the pDNA to enter the nucleus. Various methods have been explored to improve the UTMD transfection efficiency, but with little success. In cells, the classic nuclear localization signal (cNLS) peptide is an amino acid sequence that signals proteins that are due for nuclear transport. The present study aimed to investigate whether binding of a cNLS peptide to the pDNA may improve the transfection efficiency of UTMD. Four experimental groups were analyzed: Control group (UTMD + pDNA), group with cNLS (UTMD + pDNA + cNLS), group with mutated NLS (mNLS; UTMD + pDNA + mNLS), and group with cNLS and the nuclear import blocker, wheat germ agglutinin (WGA; UTMD + pDNA + cNLS + WGA). The NLS was labeled by fluorescein isothiocyanate, whereas pDNA was labeled with Cy3. Different molar ratios were tested for the NLS and pDNA combination in order to achieve optimal binding of the two molecules. Human umbilical vein endothelial cells were then transfected using the optimum ultrasonic irradiation parameters and NLS/pDNA molar ratio. At 6 h post‑transfection, the rates of Cy3‑labeled pDNA inside the cells and their nuclei were detected by flow cytometry and laser confocal microscopy, and the cellular vs. nuclear uptake of pDNA was calculated. In order to further evaluate the effect of NLS on UTMD‑mediated gene transfection, the transfection efficiency and relative expression levels of mRNA and protein were detected at 48 h post‑transfection. The results demonstrated that the optimal molar ratio of NLS with pDNA was 104:1. The rates of pDNA successful entry into the cell and nucleus were significantly higher in the cNLS group compared with the control group. The transfection efficiency, and relative expression levels of mRNA and protein from the plasmid were significantly increased in the cNLS group compared with the control group. The mNLS group displayed no significant difference compared with the control group, while the WGA group exhibited significant inhibition in most indicators of transfection efficiency compared to the cNLS group. These results suggest that combining a cNLS peptide with pDNA during UTMD‑mediated transfection significantly improved transfection efficiency. Thus, a cNLS peptide may be an important mediator and a new strategy in enhancing the efficiency of UTMD‑mediated gene transfection.
Collapse
Affiliation(s)
- Sheng Cao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jin-Ling Chen
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Nan Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yi-Jia Wang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rui-Qiang Guo
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
27
|
Bubulis A, Garalienė V, Jurėnas V, Navickas J, Giedraitis S. Effect of Low-Intensity Cavitation on the Isolated Human Thoracic Artery In Vitro. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1040-1047. [PMID: 28196770 DOI: 10.1016/j.ultrasmedbio.2016.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 12/03/2016] [Accepted: 12/14/2016] [Indexed: 06/06/2023]
Abstract
Reported here are the results of an experimental study on the response to low-intensity cavitation induced by low-frequency (4-6 W/cm2, 20 kHz and 32.6 kHz) ultrasound of isolated human arterial samples taken during conventional myocardial revascularization operations. Studies have found that low-frequency ultrasound results in a significant (48%-54%) increase in isometric contraction force and does not depend on the number of exposures (10 or 20) or the time passed since the start of ultrasound (0, 10 and 20 min), but does depend on the frequency and location (internal or external) of the blood vessels for the application of ultrasound. Diltiazem (an inhibitor of slow calcium channels) and carbachol (an agonist of muscarinic receptors) used in a concentration-dependent manner did not modify the relaxation dynamics of smooth muscle affected by ultrasound. Thus, ultrasound conditioned to the augmentation of the isometric contraction force the smooth muscle of blood vessels and did not improve endothelial- and calcium channel blocker-dependent relaxation.
Collapse
|
28
|
Lelu S, Afadzi M, Berg S, Aslund AKO, Torp SH, Sattler W, de L Davies C. Primary Porcine Brain Endothelial Cells as In Vitro Model to Study Effects of Ultrasound and Microbubbles on Blood-Brain Barrier Function. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2017; 64:281-290. [PMID: 27529871 DOI: 10.1109/tuffc.2016.2597004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Focused ultrasound (FUS) in the presence of microbubbles transiently and reversibly opens the blood-brain barrier (BBB) in rodents and humans, thereby providing a time window for increased drug delivery into brain tissue. To get insight into the underlying mechanisms that govern ultrasound (US)-mediated opening of the BBB, in vitro models are a useful alternative. In this paper, we have utilized an in vitro BBB model that consists of primary porcine brain endothelial cells (PBECs). PBEC monolayers are grown on permeable membranes, which allow assessment of key features of BBB function as well as US treatment. This experimental model is characterized by low permeability for both small molecules and proteins, has a high transendothelial electrical resistance, and expresses tight junctions and efflux pumps. Here, we compare the effects of inertial and stable cavitation in the presence of SonoVue microbubbles on PBEC monolayers' electrical resistance and permeability properties. Our results point out the fragility of PBEC monolayers, which enhances results variability. In particular, we show that handling of the inserts, such as medium change and transfer to the US setup, modifies the cellular response, and immunostaining of the monolayers introduces damage and cell detachment within the US-exposed monolayers. Our results indicate that stable cavitation might have a more pronounced impact on cell permeability as compared with inertial cavitation in vitro. This paper might contribute to further development of experimental setups that are suitable to characterize the impact of FUS and microbubbles on BBB properties in vitro.
Collapse
|
29
|
Sennoga CA, Kanbar E, Auboire L, Dujardin PA, Fouan D, Escoffre JM, Bouakaz A. Microbubble-mediated ultrasound drug-delivery and therapeutic monitoring. Expert Opin Drug Deliv 2016; 14:1031-1043. [DOI: 10.1080/17425247.2017.1266328] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Charles A. Sennoga
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Emma Kanbar
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Laurent Auboire
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | | | - Damien Fouan
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Jean-Michel Escoffre
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| | - Ayache Bouakaz
- UMR Imagerie et Cerveau, Inserm U930, Université François Rabelais, Tours, France
| |
Collapse
|
30
|
Increase of intracellular cisplatin levels and radiosensitization by ultrasound in combination with microbubbles. J Control Release 2016; 238:157-165. [DOI: 10.1016/j.jconrel.2016.07.049] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/01/2023]
|
31
|
Chen C, Gu Y, Tu J, Guo X, Zhang D. Microbubble oscillating in a microvessel filled with viscous fluid: A finite element modeling study. ULTRASONICS 2016; 66:54-64. [PMID: 26651263 DOI: 10.1016/j.ultras.2015.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/04/2015] [Accepted: 11/08/2015] [Indexed: 06/05/2023]
Abstract
Understanding the dynamics of coated-microbubble oscillating in an elastic microvessel is important for effective and safe applications of ultrasound contrast agents (UCAs) in imaging and therapy. Numerical simulations are performed based on a two-dimensional (2D) asymmetric finite element model to investigate the influences of both acoustic driving parameters (e.g., pressure and frequency) and material properties (vessel size, microbubble shell visco-elastic parameters and fluid viscosity) on the dynamic interactions in the bubble-blood-vessel system. The results show that, the constrained effect of the blood vessel along the radial direction will induce the asymmetric bubble oscillation and vessel deformation, as well as shifting the bubble resonance frequency toward the higher frequency range. For a bubble (1.5-μm radius) activated by 1-MHz ultrasound pulses in a microvessel with a radius varying between 2 and 6.5 μm, up to 26.95 kPa shear stress could be generated on the vessel wall at a driving pressure of 0.2 MPa, which should be high enough to damage the vascular endothelial cells. The asymmetrical oscillation ratio of the bubble can be aggravated from 0.12% to 79.94% with the increasing acoustic driving pressure and blood viscosity, or the decreasing vessel size and microbubble shell visco-elastic properties. The maximum compression velocity on the bubble shell will be enhanced from 0.19 to 22.79 m/s by the increasing vessel size and acoustic pressure, or the decreasing microbubble shell visco-elasticity and blood viscosity. As the results, the peak values of microstreaming-induced shear stress on the vessel wall increases from 0.003 to 26.95 kPa and the deformation degree of vessel is raised from 1.01 to 1.49, due to the enhanced acoustic amplitude, or the decreasing vessel size, blood viscosity and microbubble shell visco-elasticity. Moreover, it also suggests that, among above impact parameters, microbubble resonance frequency and UCA shell elasticity might play more dominant roles in dynamic interactions of the bubble-blood-vessel system.
Collapse
Affiliation(s)
- Chuyi Chen
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Yuyang Gu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China.
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China.
| |
Collapse
|
32
|
Lajoinie G, De Cock I, Coussios CC, Lentacker I, Le Gac S, Stride E, Versluis M. In vitro methods to study bubble-cell interactions: Fundamentals and therapeutic applications. BIOMICROFLUIDICS 2016; 10:011501. [PMID: 26865903 PMCID: PMC4733084 DOI: 10.1063/1.4940429] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/05/2016] [Indexed: 05/08/2023]
Abstract
Besides their use as contrast agents for ultrasound imaging, microbubbles are increasingly studied for a wide range of therapeutic applications. In particular, their ability to enhance the uptake of drugs through the permeabilization of tissues and cell membranes shows great promise. In order to fully understand the numerous paths by which bubbles can interact with cells and the even larger number of possible biological responses from the cells, thorough and extensive work is necessary. In this review, we consider the range of experimental techniques implemented in in vitro studies with the aim of elucidating these microbubble-cell interactions. First of all, the variety of cell types and cell models available are discussed, emphasizing the need for more and more complex models replicating in vivo conditions together with experimental challenges associated with this increased complexity. Second, the different types of stabilized microbubbles and more recently developed droplets and particles are presented, followed by their acoustic or optical excitation methods. Finally, the techniques exploited to study the microbubble-cell interactions are reviewed. These techniques operate over a wide range of timescales, or even off-line, revealing particular aspects or subsequent effects of these interactions. Therefore, knowledge obtained from several techniques must be combined to elucidate the underlying processes.
Collapse
Affiliation(s)
- Guillaume Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Ine De Cock
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University , Ghent, Belgium
| | | | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Faculty of Pharmaceutical Sciences, Ghent University , Ghent, Belgium
| | - Séverine Le Gac
- MESA+ Institute for Nanotechnology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford , Oxford, United Kingdom
| | - Michel Versluis
- Physics of Fluids Group, MESA+ Institute for Nanotechnology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| |
Collapse
|
33
|
Nande R, Howard CM, Claudio PP. Ultrasound-mediated oncolytic virus delivery and uptake for increased therapeutic efficacy: state of art. Oncolytic Virother 2015; 4:193-205. [PMID: 27512682 PMCID: PMC4918399 DOI: 10.2147/ov.s66097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The field of ultrasound (US) has changed significantly from medical imaging and diagnosis to treatment strategies. US contrast agents or microbubbles (MB) are currently being used as potential carriers for chemodrugs, small molecules, nucleic acids, small interfering ribonucleic acid, proteins, adenoviruses, and oncolytic viruses. Oncolytic viruses can selectively replicate within and destroy a cancer cell, thus making them a powerful therapeutic in treating late-stage or metastatic cancer. These viruses have been shown to have robust activity in clinical trials when injected directly into tumor nodules. However limitations in oncolytic virus’ effectiveness and its delivery approach have warranted exploration of ultrasound-mediated delivery. Gene therapy bearing adenoviruses or oncolytic viruses can be coupled with MBs and injected intravenously. Following application of US energy to the target region, the MBs cavitate, and the resulting shock wave enhances drug, gene, or adenovirus uptake. Though the underlying mechanism is yet to be fully understood, there is evidence to suggest that mechanical pore formation of cellular membranes allows for the temporary uptake of drugs. This delivery method circumvents the limitations due to stimulation of the immune system that prevented intravenous administration of viruses. This review provides insight into this intriguing new frontier on the delivery of oncolytic viruses to tumor sites.
Collapse
Affiliation(s)
- Rounak Nande
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - Candace M Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pier Paolo Claudio
- Department of BioMolecular Sciences and National Center for Natural Products Research, School of Pharmacy, University of Mississippi, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
34
|
Abstract
The combination of microbubbles and ultrasound has emerged as a promising method for local drug delivery. Microbubbles can be locally activated by a targeted ultrasound beam, which can result in several bio-effects. For drug delivery, microbubble-assisted ultrasound is used to increase vascular- and plasma membrane permeability for facilitating drug extravasation and the cellular uptake of drugs in the treated region, respectively. In the case of drug-loaded microbubbles, these two mechanisms can be combined with local release of the drug following destruction of the microbubble. The use of microbubble-assisted ultrasound to deliver chemotherapeutic agents is also referred to as sonochemotherapy. In this review, the basic principles of sonochemotherapy are discussed, including aspects such as the type of (drug-loaded) microbubbles used, the routes of administration used in vivo, ultrasound devices and parameters, treatment schedules and safety issues. Finally, the clinical translation of sonochemotherapy is discussed, including the first clinical study using sonochemotherapy.
Collapse
Affiliation(s)
- Bart H A Lammertink
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Clemens Bos
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Roel Deckers
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Gert Storm
- Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, Netherlands ; Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente Enschede, Netherlands
| | - Chrit T W Moonen
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| | - Jean-Michel Escoffre
- Image Guided Therapy, Imaging Division, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
35
|
Keravnou CP, Mannaris C, Averkiou MA. Accurate measurement of microbubble response to ultrasound with a diagnostic ultrasound scanner. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2015; 62:176-184. [PMID: 25585401 DOI: 10.1109/tuffc.2014.006664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Ultrasound and microbubbles are often used to enhance drug delivery and the suggested mechanisms are extravasation and sonoporation. Drug delivery schemes with ultrasound and microbubbles at both low and high acoustic amplitudes have been suggested. A diagnostic ultrasound scanner may play a double role as both an imaging and a therapy device. It was not possible to accurately measure microbubble response with an ultrasound scanner for a large range of acoustic pressures and microbubble concentrations until now, mainly because of signal saturation issues. A method for continuously adjusting the receive gain of a scanner and limiting signal saturation was developed to accurately measure backscattered echoes from microbubbles for mechanical indexes (MIs) up to 2.1. The intensity of backscattered echoes from microbubbles increased quarticly with MI without reaching any limit. The signal intensity from microbubbles was found to be linear with concentration at both low and high MIs. However, at very high concentrations, acoustic shadowing occurs which limits the delivered acoustic pressure in deeper areas. The contrastto- tissue ratio was also measured and found to stay constant with MI. These results can be used to better guide drug delivery approaches and to also develop imaging techniques for therapy procedures.
Collapse
|
36
|
Parrales MA, Fernandez JM, Perez-Saborid M, Kopechek JA, Porter TM. Acoustic characterization of monodisperse lipid-coated microbubbles: relationship between size and shell viscoelastic properties. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2014; 136:1077. [PMID: 25190383 DOI: 10.1121/1.4890643] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The acoustic attenuation spectrum of lipid-coated microbubble suspensions was measured in order to characterize the linear acoustic behavior of ultrasound contrast agents. For that purpose, microbubbles samples were generated with a very narrow size distribution by using microfluidics techniques. A performance as good as optical characterization techniques of single microbubbles was achieved using this method. Compared to polydispersions (i.e., contrast agents used clinically), monodisperse contrast agents have a narrower attenuation spectrum, which presents a maximum peak at a frequency value corresponding to the average single bubble resonance frequency. The low polydispersity index of the samples made the estimation of the lipid viscoelastic properties more accurate since, as previously reported, the shell linear parameters may change with the equilibrium bubble radius. The results showed the great advantage of dealing with monodisperse populations rather than polydisperse populations for the acoustic characterization of ultrasound contrast agents.
Collapse
Affiliation(s)
- Miguel A Parrales
- Aerospace Engineering and Fluid Mechanics Department, University of Seville, Camino de los Descubrimientos s/n, 41092 Sevilla, Spain
| | - Juan M Fernandez
- Aerospace Engineering and Fluid Mechanics Department, University of Seville, Camino de los Descubrimientos s/n, 41092 Sevilla, Spain
| | - Miguel Perez-Saborid
- Aerospace Engineering and Fluid Mechanics Department, University of Seville, Camino de los Descubrimientos s/n, 41092 Sevilla, Spain
| | - Jonathan A Kopechek
- Mechanical Engineering Department, Boston University, 110 Cummington Street, Boston, Massachusetts 02215
| | - Tyrone M Porter
- Mechanical Engineering Department, Boston University, 110 Cummington Street, Boston, Massachusetts 02215
| |
Collapse
|
37
|
Abstract
Ultrasound-mediated gene delivery with microbubbles has emerged as an attractive nonviral vector system for site-specific and noninvasive gene therapy. Ultrasound promotes intracellular uptake of therapeutic agents, particularly in the presence of microbubbles, by increasing vascular and cell membrane permeability. Several preclinical studies have reported successful gene delivery into solid tumors with significant therapeutic effects using this novel approach. This review provides background information on gene therapy and ultrasound bioeffects and discusses the current progress and overall perspectives on the application of ultrasound and microbubble-mediated gene delivery in cancer.
Collapse
|
38
|
Kooiman K, Vos HJ, Versluis M, de Jong N. Acoustic behavior of microbubbles and implications for drug delivery. Adv Drug Deliv Rev 2014; 72:28-48. [PMID: 24667643 DOI: 10.1016/j.addr.2014.03.003] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 02/11/2014] [Accepted: 03/18/2014] [Indexed: 12/21/2022]
Abstract
Ultrasound contrast agents are valuable in diagnostic ultrasound imaging, and they increasingly show potential for drug delivery. This review focuses on the acoustic behavior of flexible-coated microbubbles and rigid-coated microcapsules and their contribution to enhanced drug delivery. Phenomena relevant to drug delivery, such as non-spherical oscillations, shear stress, microstreaming, and jetting will be reviewed from both a theoretical and experimental perspective. Further, the two systems for drug delivery, co-administration and the microbubble as drug carrier system, are reviewed in relation to the microbubble behavior. Finally, future prospects are discussed that need to be addressed for ultrasound contrast agents to move from a pre-clinical tool into a clinical setting.
Collapse
|
39
|
Schlesinger D, Benedict S, Diederich C, Gedroyc W, Klibanov A, Larner J. MR-guided focused ultrasound surgery, present and future. Med Phys 2014; 40:080901. [PMID: 23927296 DOI: 10.1118/1.4811136] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MR-guided focused ultrasound surgery (MRgFUS) is a quickly developing technology with potential applications across a spectrum of indications traditionally within the domain of radiation oncology. Especially for applications where focal treatment is the preferred technique (for example, radiosurgery), MRgFUS has the potential to be a disruptive technology that could shift traditional patterns of care. While currently cleared in the United States for the noninvasive treatment of uterine fibroids and bone metastases, a wide range of clinical trials are currently underway, and the number of publications describing advances in MRgFUS is increasing. However, for MRgFUS to make the transition from a research curiosity to a clinical standard of care, a variety of challenges, technical, financial, clinical, and practical, must be overcome. This installment of the Vision 20∕20 series examines the current status of MRgFUS, focusing on the hurdles the technology faces before it can cross over from a research technique to a standard fixture in the clinic. It then reviews current and near-term technical developments which may overcome these hurdles and allow MRgFUS to break through into clinical practice.
Collapse
Affiliation(s)
- David Schlesinger
- Department of Radiation Oncology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Oh JS, Kwon YS, Lee KH, Jeong W, Chung SK, Rhee K. Drug perfusion enhancement in tissue model by steady streaming induced by oscillating microbubbles. Comput Biol Med 2014; 44:37-43. [DOI: 10.1016/j.compbiomed.2013.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/11/2013] [Accepted: 10/19/2013] [Indexed: 12/20/2022]
|
42
|
Kooiman K, van der Steen AFW, de Jong N. Role of intracellular calcium and reactive oxygen species in microbubble-mediated alterations of endothelial layer permeability. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2013; 60:1811-5. [PMID: 24658714 DOI: 10.1109/tuffc.2013.2767] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Drugs will be delivered to diseased tissue more efficiently if the vascular endothelial permeability is increased. Ultrasound in combination with an ultrasound contrast agent is known to increase the permeability of the endothelial layer, but the mechanism is not known. The goal of this study was to elucidate whether intracellular calcium ions, [Ca(2+)]i, and reactive oxygen species (ROS) are part of the mechanism that leads to an increased endothelial layer permeability following ultrasound and microbubble treatment. Human umbilical vein endothelial cells (HUVECs) treated for 2 min with ultrasound-activated microbubbles (1 MHz, 210 kPa, 10 000 cycles, 20 Hz repetition rate) had an increased permeability that lasted up to 12 h. Recovery of permeability after 2 h was only found when HUVECs were preincubated with the [Ca(2+)]i chelator BAPTA-AM or the antioxidant butylated hydroxytoluene (BHT). This suggests that both [Ca(2+)]i and ROS play an important role in the mechanism of increased permeability following ultrasound in combination with microbubble treatment.
Collapse
|
43
|
Villanueva FS. Getting good vibes: the therapeutic power of microbubbles and ultrasound. JACC Cardiovasc Imaging 2013; 5:1263-6. [PMID: 23236977 DOI: 10.1016/j.jcmg.2012.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 09/25/2012] [Accepted: 09/27/2012] [Indexed: 10/27/2022]
|
44
|
Mullin LB, Phillips LC, Dayton PA. Nanoparticle delivery enhancement with acoustically activated microbubbles. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2013; 60:65-77. [PMID: 23287914 PMCID: PMC3822910 DOI: 10.1109/tuffc.2013.2538] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The application of microbubbles and ultrasound to deliver nanoparticle carriers for drug and gene delivery is an area that has expanded greatly in recent years. Under ultrasound exposure, microbubbles can enhance nanoparticle delivery by increasing cellular and vascular permeability. In this review, the underlying mechanisms of enhanced nanoparticle delivery with ultrasound and microbubbles and various proposed delivery techniques are discussed. Additionally, types of nanoparticles currently being investigated in preclinical studies, as well as the general limitations and benefits of a microbubble- based approach to nanoparticle delivery, are reviewed.
Collapse
Affiliation(s)
- Lee B Mullin
- Joint Department of Biomedical Engineering The University of North Carolina at Chapel Hill, and North Carolina State University
| | - Linsey C Phillips
- Joint Department of Biomedical Engineering The University of North Carolina at Chapel Hill, and North Carolina State University
| | - Paul A Dayton
- Joint Department of Biomedical Engineering The University of North Carolina at Chapel Hill, and North Carolina State University
- Author to whom correspondence should be addressed Paul A. Dayton Campus Box 7575, UNC Chapel Hill Chapel Hill, NC 27599
| |
Collapse
|
45
|
Faez T, Emmer M, Kooiman K, Versluis M, van der Steen A, de Jong N. 20 years of ultrasound contrast agent modeling. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2013; 60:7-20. [PMID: 23287909 DOI: 10.1109/tuffc.2013.2533] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The merits of ultrasound contrast agents (UCAs) were already known in the 1960s. It was, however, not until the 1990s that UCAs were clinically approved and marketed. In these years, it was realized that the UCAs are not just efficient ultrasound scatterers, but that their main constituent, the coated gas microbubble, acts as a nonlinear resonator and, as such, is capable of generating harmonic energy. Subharmonic, ultraharmonic, and higher harmonic frequencies of the transmitted ultrasound frequency have been reported. This opened up new prospects for their use and several detection strategies have been developed to exploit this harmonic energy to discriminate the contrast bubbles from surrounding tissue. This insight created a need for tools to study coated bubble behavior in an ultrasound field and the first models were developed. Since then, 20 years have elapsed, in which a broad range of UCAs and UCA models have been developed. Although the models have helped in understanding the responses of coated bubbles, the influence of the coating has not been fully elucidated to date and UCA models are still being improved. The aim of this review paper is to offer an overview in these developments and indicate future directions for research.
Collapse
Affiliation(s)
- Telli Faez
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
46
|
Luan Y, Faez T, Gelderblom E, Skachkov I, Geers B, Lentacker I, van der Steen T, Versluis M, de Jong N. Acoustical properties of individual liposome-loaded microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:2174-2185. [PMID: 23196203 DOI: 10.1016/j.ultrasmedbio.2012.07.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 05/24/2023]
Abstract
A comparison between phospholipid-coated microbubbles with and without liposomes attached to the microbubble surface was performed using the ultra-high-speed imaging camera (Brandaris 128). We investigated 73 liposome-loaded microbubbles (loaded microbubbles) and 41 microbubbles without liposome loading (unloaded microbubbles) with a diameter ranging from 3-10 μm at frequencies ranging from 0.6-3.8 MHz and acoustic pressures ranging from 5-100 kPa. The experimental data showed nearly the same shell elasticity for the loaded and unloaded bubbles, but the shell viscosity was higher for loaded bubbles compared with unloaded bubbles. For loaded bubbles, a higher pressure threshold for the bubble vibrations was noticed. In addition, an "expansion-only" behavior was observed for up to 69% of the investigated loaded bubbles, which mostly occurred at low acoustic pressures (≤30 kPa). Finally, fluorescence imaging showed heterogeneity of liposome distributions of the loaded bubbles.
Collapse
Affiliation(s)
- Ying Luan
- Biomedical Engineering Thoraxcentre, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tlaxca JL, Rychak JJ, Ernst PB, Konkalmatt PR, Shevchenko TI, Pizarro TT, Pizzaro TT, Rivera-Nieves J, Klibanov AL, Lawrence MB. Ultrasound-based molecular imaging and specific gene delivery to mesenteric vasculature by endothelial adhesion molecule targeted microbubbles in a mouse model of Crohn's disease. J Control Release 2012; 165:216-25. [PMID: 23142578 DOI: 10.1016/j.jconrel.2012.10.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 09/27/2012] [Accepted: 10/15/2012] [Indexed: 02/06/2023]
Abstract
Crohn's disease (CD) is a chronic inflammatory disorder of the gastrointestinal tract (GI) for which treatments with immunosuppressive drugs have significant side-effects. Consequently, there is a clinical need for site-specific and non-toxic delivery of therapeutic genes or drugs for CD and related disorders such as inflammatory bowel disease. The aim of this study was to validate a gene delivery platform based on ultrasound-activated lipid-shelled microbubbles (MBs) targeted to inflamed mesenteric endothelium in the CD-like TNFΔARE mouse model. MBs bearing luciferase plasmid were functionalized with antibodies to MAdCAM-1 (MB-M) or VCAM-1 (MB-V), biomarkers of gut endothelial cell inflammation and evaluated in an in vitro flow chamber assay with appropriate ligands to confirm targeting specificity. Following MB retro-orbital injection in TNFΔARE mice, the mean contrast intensity in the ileocecal region from accumulated MB-M and MB-V was 8.5-fold and 3.6-fold greater, respectively, compared to MB-C. Delivery of luciferase plasmid to the GI tract in TNFΔARE mice was achieved by insonating the endothelial cell-bound agents using a commercial sonoporator. Luciferase expression in the midgut was detected 48 h later by bioluminescence imaging and further confirmed by immunohistochemical staining. The liver, spleen, heart, and kidney had no detectable bioluminescence following insonation. Transfection of the microcirculation guided by a targeted, acoustically-activated platform such as an ultrasound contrast agent microbubble has the potential to be a minimally-invasive treatment strategy to ameliorate CD and other inflammatory conditions.
Collapse
Affiliation(s)
- José L Tlaxca
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Image-guided, targeted and triggered drug delivery to tumors using polymer-based microbubbles. J Control Release 2012; 163:75-81. [PMID: 22580225 DOI: 10.1016/j.jconrel.2012.05.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/29/2012] [Accepted: 05/02/2012] [Indexed: 11/21/2022]
Abstract
Microbubbles (MB) are routinely used contrast agents for functional and molecular ultrasound (US) imaging. In addition, they have been attracting more and more attention for drug delivery purposes, enabling e.g. US-mediated drug delivery across biological barriers and US-induced triggered drug release from the MB shell. The vast majority of efforts in this regard have thus far focused on phospholipid-based soft-shell MB, which are suboptimal for stably incorporating large amounts of drug molecules because of their relatively thin shell. Using poly(butyl cyanoacrylate) (PBCA)-based hard-shell MB, we show here that both hydrophilic (Rhodamine-B) and hydrophobic (Coumarin-6) model drugs can be efficiently and stably entrapped within the ~50 nm shell of PBCA MB. In addition, we demonstrate that model drug loading does not negatively affect the acoustic properties of the MB, and that functionalizing the surface of fluorophore-loaded MB with anti-VEGFR2 antibodies enables image-guided and targeted model drug delivery to tumor blood vessels. Finally, we show both in vitro and in vivo that disintegrating VEGFR2-targeted MB with high-mechanical index US pulses leads to high levels of model drug release. Consequently, these findings indicate that polymer-based MB are highly suitable systems for image-guided, targeted and triggered drug delivery to tumors and tumor blood vessels.
Collapse
|
49
|
Mannell H, Pircher J, Fochler F, Stampnik Y, Räthel T, Gleich B, Plank C, Mykhaylyk O, Dahmani C, Wörnle M, Ribeiro A, Pohl U, Krötz F. Site directed vascular gene delivery in vivo by ultrasonic destruction of magnetic nanoparticle coated microbubbles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 8:1309-18. [PMID: 22480917 DOI: 10.1016/j.nano.2012.03.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 03/14/2012] [Accepted: 03/24/2012] [Indexed: 12/26/2022]
Abstract
UNLABELLED Site specific vascular gene delivery for therapeutic implications is favorable because of reduction of possible side effects. Yet this technology faces numerous hurdles that result in low transfection rates because of suboptimal delivery. Combining ultrasonic microbubble technology with magnetic nanoparticle enhanced gene transfer could make it possible to use the systemic vasculature as the route of application and to magnetically trap these compounds at the target of interest. In this study we show that magnetic nanoparticle-coated microbubbles bind plasmid DNA and successfully deliver it to endothelial cells in vitro and more importantly transport their cargo through the vascular system and specifically deliver it to the vascular wall in vivo at sites where microbubbles are retained by magnetic force and burst by local ultrasound application. This resulted in a significant enhancement in site specific gene delivery compared with the conventional microbubble technique. Thus, this technology may have promising therapeutic potential. FROM THE CLINICAL EDITOR This work focuses on combining ultrasonic microbubble technology with magnetic nanoparticle enhanced gene transfer to enable targeted gene delivery via the systemic vasculature and magnetic trapping of these compounds at the target of interest.
Collapse
Affiliation(s)
- Hanna Mannell
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mannaris C, Averkiou MA. Investigation of microbubble response to long pulses used in ultrasound-enhanced drug delivery. ULTRASOUND IN MEDICINE & BIOLOGY 2012; 38:681-91. [PMID: 22341047 DOI: 10.1016/j.ultrasmedbio.2011.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 12/13/2011] [Accepted: 12/17/2011] [Indexed: 05/05/2023]
Abstract
In current drug delivery approaches, microbubbles and drugs can be co-administered while ultrasound is applied. The mechanism of microbubble interaction with ultrasound, the drug and the cells is not fully understood. The aim of this study was to investigate microbubble response to long ultrasonic pulses used in drug delivery approaches. Two different in vitro set-ups were considered: with the microbubbles diluted in an enclosure and with the microbubbles flowing in a capillary tube. Acoustic streaming, which influences the observed bubble response, was observed in "typical" drug delivery conditions in the first set-up. With the capillary set-up, streaming effects were avoided and accurate bubble responses were recorded. The diffraction pattern of the source greatly influences the bubble response and in different locations of the field different bubble responses are observed. At low nondestructive pressures, microbubbles can oscillate for thousands of cycles repeatedly. At high acoustic pressures (at 1 MHz), most bubble activity disappeared within about 100 μs despite the length of the pulse, mainly due to violent bubble destruction and subsequent accelerated diffusion.
Collapse
|