1
|
Fan CH, Huang E, Lo WC, Yeh CK. Ultrasound-cavitation-enhanced drug delivery via microbubble clustering induced by acoustic vortex tweezers. ULTRASONICS SONOCHEMISTRY 2025; 114:107273. [PMID: 39979196 PMCID: PMC12013124 DOI: 10.1016/j.ultsonch.2025.107273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
The application of acoustic vortex tweezers (AVT) in conjunction with ultrasound (US) cavitation pulses presents a promising noninvasive approach for the delivery of high concentrations of therapeutic agents. This methodology facilitates the aggregation of drug-loaded microbubbles (MBs) into clusters, which are subsequently destroyed to release their contents. Nevertheless, prior investigations have not thoroughly examined the resonance frequency and cavitation activity of MB clusters, critical factors that could enhance the efficiency of payload release. Theoretically, the resonance frequency of an MB cluster is expected to approximate that of a single large bubble of comparable size, thus being significantly lower than that of the individual MBs constituting the cluster. Accordingly, this study aims to optimize the release of payloads from AVT-trapped MB clusters, which measure 15 to 40 μm (mean radius: 24.7 μm) in size, by employing US at their resonance frequency of 100 kHz, henceforth referred to as "on-resonance US." In this investigation, MBs were loaded with the model drug DiI, resulting in the formation of DiI-MBs, which were then clustered utilizing AVT. On-resonance US excitation was subsequently applied to enhance the release of the drug payload. The dimensional characteristics of the DiI-MB clusters formed via 3-MHz AVT were measured to determine the range of resonance frequencies. Concurrent optical and acoustic analyses were conducted to evaluate the size, oscillation dynamics, and cavitation activity of the DiI-MB clusters in response to on-resonance US excitation. Additionally, the payload release from these clusters was quantitatively assessed. Our results indicate that significant oscillations of individual DiI-MB clusters commenced at a pressure of 44 kPa during 100 kHz US excitation. Further quantitative experiments demonstrated that the synergistic combination of AVT and 100-kHz US at 65 kPa significantly enhanced the payload release efficiency to 93 %. This efficiency surpassed that achieved with either method independently, with increases of 1.8-fold relative to AVT alone and 2.3-fold compared to 100-kHz US alone. The acoustic analyses revealed the onset of inertial cavitation at 44 kPa, which strongly correlated with payload release efficiency (R2 = 0.78). These findings underscore the potential of our proposed methodology in monitoring and enhancing the efficiency of drug release.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Elaine Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Wei-Chen Lo
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan; Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
2
|
Al Refaai KA, AlSawaftah NA, Abuwatfa W, Husseini GA. Drug Release via Ultrasound-Activated Nanocarriers for Cancer Treatment: A Review. Pharmaceutics 2024; 16:1383. [PMID: 39598507 PMCID: PMC11597164 DOI: 10.3390/pharmaceutics16111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Conventional cancer chemotherapy often struggles with safely and effectively delivering anticancer therapeutics to target tissues, frequently leading to dose-limiting toxicity and suboptimal therapeutic outcomes. This has created a need for novel therapies that offer greater efficacy, enhanced safety, and improved toxicological profiles. Nanocarriers are nanosized particles specifically designed to enhance the selectivity and effectiveness of chemotherapy drugs while reducing their toxicity. A subset of drug delivery systems utilizes stimuli-responsive nanocarriers, which enable on-demand drug release, prevent premature release, and offer spatial and temporal control over drug delivery. These stimuli can be internal (such as pH and enzymes) or external (such as ultrasound, magnetic fields, and light). This review focuses on the mechanics of ultrasound-induced drug delivery and the various nanocarriers used in conjunction with ultrasound. It will also provide a comprehensive overview of key aspects related to ultrasound-induced drug delivery, including ultrasound parameters and the biological effects of ultrasound waves.
Collapse
Affiliation(s)
- Khaled Armouch Al Refaai
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - Nour A. AlSawaftah
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
3
|
Singh D, Memari E, He S, Yusefi H, Helfield B. Cardiac gene delivery using ultrasound: State of the field. Mol Ther Methods Clin Dev 2024; 32:101277. [PMID: 38983873 PMCID: PMC11231612 DOI: 10.1016/j.omtm.2024.101277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Over the past two decades, there has been tremendous and exciting progress toward extending the use of medical ultrasound beyond a traditional imaging tool. Ultrasound contrast agents, typically used for improved visualization of blood flow, have been explored as novel non-viral gene delivery vectors for cardiovascular therapy. Given this adaptation to ultrasound contrast-enhancing agents, this presents as an image-guided and site-specific gene delivery technique with potential for multi-gene and repeatable delivery protocols-overcoming some of the limitations of alternative gene therapy approaches. In this review, we provide an overview of the studies to date that employ this technique toward cardiac gene therapy using cardiovascular disease animal models and summarize their key findings.
Collapse
Affiliation(s)
- Davindra Singh
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Elahe Memari
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Hossein Yusefi
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Brandon Helfield
- Department of Biology, Concordia University, Montreal, QC, Canada
- Department of Physics, Concordia University, Montreal, QC, Canada
| |
Collapse
|
4
|
Fuenteslópez CV, Gray M, Bahcevanci S, Martin A, Smith CAB, Coussios C, Cui Z, Ye H, Patrulea V. Mesenchymal stem cell cryopreservation with cavitation-mediated trehalose treatment. COMMUNICATIONS ENGINEERING 2024; 3:129. [PMID: 39251849 PMCID: PMC11385975 DOI: 10.1038/s44172-024-00265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Dimethylsulfoxide (DMSO) has conventionally been used for cell cryopreservation both in research and in clinical applications, but has long-term cytotoxic effects. Trehalose, a natural disaccharide, has been proposed as a non-toxic cryoprotectant. However, the lack of specific cell membrane transporter receptors inhibits transmembrane transport and severely limits its cryoprotective capability. This research presents a method to successfully deliver trehalose into mesenchymal stem cells (MSCs) using ultrasound in the presence of microbubbles. The optimised trehalose concentration was shown to be able to not only preserve membrane integrity and cell viability but also the multipotency of MSCs, which are essential for stem cell therapy. Confocal imaging revealed that rhodamine-labelled trehalose was transported into cells rather than simply attached to the membrane. Additionally, the membranes were successfully preserved in lyophilised cells. This study demonstrates that ultrasonication with microbubbles facilitated trehalose delivery, offering promising cryoprotective capability without the cytotoxicity associated with DMSO-based methods.
Collapse
Affiliation(s)
- Carla V Fuenteslópez
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Simge Bahcevanci
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Alexander Martin
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Cameron A B Smith
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| | - Viorica Patrulea
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
5
|
Memari E, Helfield B. Shear stress preconditioning and microbubble flow pattern modulate ultrasound-assisted plasma membrane permeabilization. Mater Today Bio 2024; 27:101128. [PMID: 38988819 PMCID: PMC11234154 DOI: 10.1016/j.mtbio.2024.101128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
The recent and exciting success of anti-inflammatory therapies for ischemic heart disease (e.g. atherosclerosis) is hindered by the lack of site-specific and targeted therapeutic deposition. Microbubble-mediated focused ultrasound, which uses circulating, lipid-encapsulated intravascular microbubbles to locally enhance endothelial permeability, offers an exciting approach. Atherosclerotic plaques preferentially develop in regions with disturbed blood flow, and microbubble-endothelial cell membrane interactions under such flow conditions are not well understood. Here, using an acoustically-coupled microscopy system, endothelial cells were sonicated (1 MHz, 20 cycle bursts, 1 ms PRI, 4 s duration, 300 kPa peak-negative pressure) under perfusion with Definity™ bubbles to examine microbubble-mediated endothelial permeabilization under a range of physiological conditions. Endothelial preconditioning under prolonged shear influenced physiology and the secretome, inducing increased expression of pro-angiogenesis analytes, decreasing levels of pro-inflammatory ones, and increasing the susceptibility of ultrasound therapy. Ultrasound treatment efficiency was positively correlated with concentrations of pro-angiogenic cytokines (e.g. VEGF-A, EGF, FGF-2), and negatively correlated with pro-inflammatory chemokines (e.g. MCP-1, GCP-2, SDF-1). Furthermore, ultrasound therapy under non-reversing pulsatile flow (∼4-8 dyne/cm2, 0.5-1 Hz) increased permeabilization up to 2.4-fold compared to shear-matched laminar flow, yet treatment under reversing oscillatory flow resulted in more heterogeneous modulation. This study provides insight into the role of vascular physiology, including endothelial biology, into the design of a localized ultrasound drug delivery system for ischemic heart disease.
Collapse
Affiliation(s)
- Elahe Memari
- Department of Physics, Concordia University, Montreal, H4B 1R6, Canada
| | - Brandon Helfield
- Department of Physics, Concordia University, Montreal, H4B 1R6, Canada
- Department of Biology, Concordia University, Montreal, H4B 1R6, Canada
| |
Collapse
|
6
|
Fletcher SMP, Zhang Y, Chisholm A, Martinez S, McDannold N. The impact of pulse repetition frequency on microbubble activity and drug delivery during focused ultrasound-mediated blood-brain barrier opening. Phys Med Biol 2024; 69:145002. [PMID: 38914104 DOI: 10.1088/1361-6560/ad5b47] [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: 04/09/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024]
Abstract
Objective.Pulsed focused ultrasound (FUS) can deliver therapeutics to the brain by using intravenous microbubbles (MBs) to open the blood-brain barrier (BBB). MB emissions indicate treatment outcomes, like BBB opening (harmonics) and damage (broadband). Typically, a pulse repetition frequency (PRF) of 1 Hz is used, but the effect of PRF on MBs is not fully understood. We investigated the effect of PRF on MB activity and tracer delivery.Approach.The effect of PRF (0.125, 0.25, 0.5, 1, and 2 Hz) on MB activity was monitored through harmonic and wideband emissions during FUS sonications of the rat brain at 274.3 kHz. BBB opening was quantified through fluorescence imaging to estimate the concentration of Trypan Blue (TB) dye following a 75-pulse FUS exposure for PRFs of 1 and 0.25 Hz.Main results.At a fixed acoustic pressure, the percentage change in maximum harmonic amplitude compared to the control (PRF = 1 Hz) decreased with increasing PRF, with a median change of 73.8% at 0.125 Hz and -38.3% at 2 Hz. There was no difference in the pressure threshold for broadband emissions between PRFs of 0.25 and 1 Hz. PRF = 0.25 Hz, led to a 68.2% increase in the mean concentration of TB measured after FUS, with a 53.9% increase in the mean harmonic sum, compared with PRF = 1 Hz. Harmonic emissions-based control at PRF = 0.25 Hz yielded similar TB delivery, with less damage at histology, compared with 1 Hz.Significance.For a fixed number of FUS pulses, reducing the PRF was shown to increase the magnitude of harmonic emissions and TB delivery, but not the threshold for broadband emissions. While further research is necessary to understand the mechanisms involved, these results may be useful to improve clinical safety margins and sensitivity to detecting small harmonic signals from cavitating MBs.
Collapse
Affiliation(s)
- Stecia-Marie P Fletcher
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America
- Department of Radiology, Harvard Medical School, Boston, MA, United States of America
| | - Yongzhi Zhang
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America
- Department of Radiology, Harvard Medical School, Boston, MA, United States of America
| | - Amanda Chisholm
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Sofia Martinez
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Nathan McDannold
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America
- Department of Radiology, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
7
|
Wang J, Luo T, Chen J, Liu Z, Wang J, Zhang X, Li H, Ma Y, Zhang F, Ju H, Wang W, Wang Y, Zhu Q. Enhancement of Tumor Perfusion and Antiangiogenic Therapy in Murine Models of Clear Cell Renal Cell Carcinoma Using Ultrasound-Stimulated Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:680-689. [PMID: 38311538 DOI: 10.1016/j.ultrasmedbio.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/06/2024]
Abstract
OBJECTIVE To explore the effect of ultrasound-stimulated microbubble cavitation (USMC) on enhancing antiangiogenic therapy in clear cell renal cell carcinoma. MATERIALS AND METHODS We explored the effects of USMC with different mechanical indices (MIs) on tumor perfusion, 36 786-O tumor-bearing nude mice were randomly assigned into four groups: (i) control group, (ii) USMC0.25 group (MI = 0.25), (iii) USMC1.4 group (MI = 1.4) (iv) US1.4 group (MI = 1.4). Tumor perfusion was assessed by contrast-enhanced ultrasound (CEUS) before the USMC treatment and 30 min, 4h and 6h after the USMC treatment, respectively. Then we evaluated vascular normalization(VN) induced by low-MI (0.25) USMC treatment, 12 tumor-bearing nude mice were randomly divided into two groups: (i) control group (ii) USMC0.25 group. USMC treatment was performed, and tumor microvascular imaging and blood perfusion were analyzed by MicroFlow imaging (MFI) and CEUS 30 min after each treatment. In combination therapy, a total of 144 tumor-bearing nude mice were randomly assigned to six groups (n = 24): (i) control group, (ii) USMC1.4 group, (iii) USMC0.25 group, (iv) bevacizumab(BEV) group, (v) USMC1.4 +BEV group, (vi) USMC0.25 +BEV group. BEV was injected on the 6th, 10th, 14th, and 18th d after the tumors were inoculated, while USMC treatment was performed 24 h before and after every BEV administration. We examined the effects of the combination therapy through a series of experiments. RESULTS Tumor blood perfusion enhanced by USMC with low MI (0.25)could last for more than 6h, inducing tumor VN and promoting drug delivery. Compared with other groups, USMC0.25+BEV combination therapy had the strongest inhibition on tumor growth, led to the longest survival time of the mice. CONCLUSION The optimized USMC is a promising therapeutic approach that can be combined with antiangiogenic therapy to combat tumor progression.
Collapse
Affiliation(s)
- Juan Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jianghong Chen
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Juan Wang
- Department of Pathology,The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiiazhuang, Hebei, China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yulin Ma
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fan Zhang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongjuan Ju
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wengang Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueheng Wang
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
8
|
Fan CH, Tsai CY, Lai CY, Liou YF, Lee JK, Yeh CK. Feasibility of in vitro calcification plaque disruption using ultrasound-induced microbubble inertial cavitation. ULTRASONICS 2024; 138:107238. [PMID: 38183758 DOI: 10.1016/j.ultras.2023.107238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
Percutaneous transluminal coronary angioplasty (PTCA) is a clinical method in which plaque-narrowed arteries are widened by inflating an intravascular balloon catheter. However, PTCA remains challenging to apply in calcified plaques since the high pressure required for achieving a therapeutic outcome can result in balloon rupture, vessel rupture, and intimal dissection. To address the problem with PTCA, we hypothesized that a calcified plaque can be disrupted by microbubbles (MBs) inertial cavitation induced by ultrasound (US). This study proposed a columnar US transducer with a novel design to generate inertial cavitation at the lesion site. Experiments were carried out using tubular calcification phantom to mimic calcified plaques. After different parameters of US + MBs treatment (four types of MBs concentration, five types of cycle number, and three types of insonication duration; n = 4 in each group), inflation experiments were performed to examine the efficacy of cavitation for a clinically used balloon catheter. Finally, micro-CT was used to investigate changes in the internal structure of the tubular plaster phantoms. The inflation threshold of the untreated tubular plaster phantoms was > 11 atm, and this was significantly reduced to 7.4 ± 0.7 atm (p = 5.2E-08) using US-induced MBs inertial cavitation at a treatment duration of 20 min with an acoustic pressure of 214 kPa, an MBs concentration of 4.0 × 108 MBs/mL, a cycle number of 100 cycles, and a pulse repetition frequency of 100 Hz. Moreover, micro-CT revealed internal damage in the tubular calcification phantom, demonstrating that US-induced MBs inertial cavitation can effectively disrupt calcified plaques and reduce the inflation threshold of PTCA. The ex vivo histopathology results showed that the endothelium of pig blood vessels remained intact after the treatment. In summary, the results show that US-induced MBs inertial cavitation can markedly reduce the inflation threshold in PTCA without damaging blood vessel endothelia, indicating the potential of the proposed treatment method.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan 701, Taiwan
| | - Chieh-Yu Tsai
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chun-Yen Lai
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ya-Fu Liou
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jen-Kuang Lee
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 10617, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
9
|
Ostasevicius V, Jurenas V, Venslauskas M, Kizauskiene L, Zigmantaite V, Stankevicius E, Bubulis A, Vezys J, Mikuckyte S. Low-frequency ultrasound for pulmonary hypertension therapy. Respir Res 2024; 25:70. [PMID: 38317182 PMCID: PMC10840147 DOI: 10.1186/s12931-024-02713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/28/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Currently, there are no reliable clinical tools that allow non-invasive therapeutic support for patients with pulmonary arterial hypertension. This study aims to propose a low-frequency ultrasound device for pulmonary hypertension therapy and to demonstrate its potential. METHODS A novel low-frequency ultrasound transducer has been developed. Due to its structural properties, it is excited by higher vibrational modes, which generate a signal capable of deeply penetrating biological tissues. A methodology for the artificial induction of pulmonary hypertension in sheep and for the assessment of lung physiological parameters such as blood oxygen concentration, pulse rate, and pulmonary blood pressure has been proposed. RESULTS The results showed that exposure of the lungs to low-frequency ultrasound changed physiological parameters such as blood oxygen concentration, pulse rate and blood pressure. These parameters are most closely related to indicators of pulmonary hypertension (PH). The ultrasound exposure increased blood oxygen concentration over a 7-min period, while pulse rate and pulmonary blood pressure decreased over the same period. In anaesthetised sheep exposed to low-frequency ultrasound, a 10% increase in SpO2, a 10% decrease in pulse rate and an approximate 13% decrease in blood pressure were observed within 7 min. CONCLUSIONS The research findings demonstrate the therapeutic efficiency of low-frequency ultrasound on hypertensive lungs, while also revealing insights into the physiological aspects of gas exchange within the pulmonary system.
Collapse
Affiliation(s)
- Vytautas Ostasevicius
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, 51424, Kaunas, Lithuania.
| | - Vytautas Jurenas
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, 51424, Kaunas, Lithuania
| | - Mantas Venslauskas
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, 51424, Kaunas, Lithuania
| | - Laura Kizauskiene
- Department of Computer Sciences, Kaunas University of Technology, Studentu Street 50, 51368, Kaunas, Lithuania
| | - Vilma Zigmantaite
- Biological Research Center Lithuanian, University of Health Sciences, Tilžės Street 18, 47181, Kaunas, Lithuania
- Laboratory of Membrane Biophysics, Cardiology Department, Lithuanian University of Health Sciences, Sukilėlių Street 15, 50103, Kaunas, Lithuania
| | - Edgaras Stankevicius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, A. Mickevicius Street 9, 44307, Kaunas, Lithuania
| | - Algimantas Bubulis
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, 51424, Kaunas, Lithuania
| | - Joris Vezys
- Department of Mechanical Engineering, Kaunas University of Technology, Kaunas University of Technology, Studentu Street 56, 51424, Kaunas, Lithuania
| | - Sandra Mikuckyte
- Institute of Mechatronics, Kaunas University of Technology, Studentu Street 56, 51424, Kaunas, Lithuania
| |
Collapse
|
10
|
Li B, Lin Y, Chen G, Cai M, Zhong H, Xiao Z, Lin M, Li T, Cai Y, Shuai X, Ren J. Anchoring Microbubbles on Cerebrovascular Endothelium as a New Strategy Enabling Low-Energy Ultrasound-Assisted Delivery of Varisized Agents Across Blood-Brain Barrier. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302134. [PMID: 37870165 PMCID: PMC10667842 DOI: 10.1002/advs.202302134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/20/2023] [Indexed: 10/24/2023]
Abstract
The protective blood-brain barrier (BBB) prevents most therapeutic agents from entering the brain. Currently, focused ultrasound (FUS) is mostly employed to create microbubbles that induce a cavitation effect to open the BBB. However, microbubbles pass quickly through brain microvessels, substantially limiting the cavitation effect. Here, we constructed a novel perfluoropropane-loaded microbubble, termed ApoER-Pep-MB, which possessed a siloxane bonds-crosslinked surface to increase the microbubble stability against turbulence in blood circulation and was decorated with binding peptide for apolipoprotein E receptor (ApoER-Pep). The microbubble with tailor-made micron size (2 µm) and negative surface charge (-30 mV) performed ApoER-mediated binding rather than internalization into brain capillary endothelial cells. Consequently, the microbubble accumulated on the brain microvessels, based on which even a low-energy ultrasound with less safety risk than FUS, herein diagnostic ultrasound (DUS), could create a strong cavitation effect to open the BBB. Evans Blue and immunofluorescence staining studies demonstrated that the DUS-triggered cavitation effect not only temporarily opened the BBB for 2 h but also caused negligible damage to the brain tissue. Therefore, various agents, ranging from small molecules to nanoscale objects, can be efficiently delivered to target regions of the brain, offering tremendous opportunities for the treatment of brain diseases.
Collapse
Affiliation(s)
- Bo Li
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Yuejun Lin
- Department of Medical UltrasonicThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Gengjia Chen
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Mingyue Cai
- Department of Minimally Invasive Interventional Radiologythe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Huihai Zhong
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Zecong Xiao
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Minzhao Lin
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Tan Li
- Department of Minimally Invasive Interventional Radiologythe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510260China
| | - Yujun Cai
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Xintao Shuai
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Jie Ren
- Department of Medical UltrasonicThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| |
Collapse
|
11
|
Kancheva M, Aronson L, Pattilachan T, Sautto F, Daines B, Thommes D, Shar A, Razavi M. Bubble-Based Drug Delivery Systems: Next-Generation Diagnosis to Therapy. J Funct Biomater 2023; 14:373. [PMID: 37504868 PMCID: PMC10382061 DOI: 10.3390/jfb14070373] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
Current radiologic and medication administration is systematic and has widespread side effects; however, the administration of microbubbles and nanobubbles (MNBs) has the possibility to provide therapeutic and diagnostic information without the same ramifications. Microbubbles (MBs), for instance, have been used for ultrasound (US) imaging due to their ability to remain in vessels when exposed to ultrasonic waves. On the other hand, nanobubbles (NBs) can be used for further therapeutic benefits, including chronic treatments for osteoporosis and cancer, gene delivery, and treatment for acute conditions, such as brain infections and urinary tract infections (UTIs). Clinical trials are also being conducted for different administrations and utilizations of MNBs. Overall, there are large horizons for the benefits of MNBs in radiology, general medicine, surgery, and many more medical applications. As such, this review aims to evaluate the most recent publications from 2016 to 2022 to report the current uses and innovations for MNBs.
Collapse
Affiliation(s)
- Mihaela Kancheva
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Lauren Aronson
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Tara Pattilachan
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Francesco Sautto
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Benjamin Daines
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Donald Thommes
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Angela Shar
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
12
|
Boltman T, Meyer M, Ekpo O. Diagnostic and Therapeutic Approaches for Glioblastoma and Neuroblastoma Cancers Using Chlorotoxin Nanoparticles. Cancers (Basel) 2023; 15:3388. [PMID: 37444498 DOI: 10.3390/cancers15133388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma multiforme (GB) and high-risk neuroblastoma (NB) are known to have poor therapeutic outcomes. As for most cancers, chemotherapy and radiotherapy are the current mainstay treatments for GB and NB. However, the known limitations of systemic toxicity, drug resistance, poor targeted delivery, and inability to access the blood-brain barrier (BBB), make these treatments less satisfactory. Other treatment options have been investigated in many studies in the literature, especially nutraceutical and naturopathic products, most of which have also been reported to be poorly effective against these cancer types. This necessitates the development of treatment strategies with the potential to cross the BBB and specifically target cancer cells. Compounds that target the endopeptidase, matrix metalloproteinase 2 (MMP-2), have been reported to offer therapeutic insights for GB and NB since MMP-2 is known to be over-expressed in these cancers and plays significant roles in such physiological processes as angiogenesis, metastasis, and cellular invasion. Chlorotoxin (CTX) is a promising 36-amino acid peptide isolated from the venom of the deathstalker scorpion, Leiurus quinquestriatus, demonstrating high selectivity and binding affinity to a broad-spectrum of cancers, especially GB and NB through specific molecular targets, including MMP-2. The favorable characteristics of nanoparticles (NPs) such as their small sizes, large surface area for active targeting, BBB permeability, etc. make CTX-functionalized NPs (CTX-NPs) promising diagnostic and therapeutic applications for addressing the many challenges associated with these cancers. CTX-NPs may function by improving diffusion through the BBB, enabling increased localization of chemotherapeutic and genotherapeutic drugs to diseased cells specifically, enhancing imaging modalities such as magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), optical imaging techniques, image-guided surgery, as well as improving the sensitization of radio-resistant cells to radiotherapy treatment. This review discusses the characteristics of GB and NB cancers, related treatment challenges as well as the potential of CTX and its functionalized NP formulations as targeting systems for diagnostic, therapeutic, and theranostic purposes. It also provides insights into the potential mechanisms through which CTX crosses the BBB to bind cancer cells and provides suggestions for the development and application of novel CTX-based formulations for the diagnosis and treatment of GB and NB in the future.
Collapse
Affiliation(s)
- Taahirah Boltman
- Department of Medical Biosciences, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Mervin Meyer
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Okobi Ekpo
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
13
|
Heo J, Park JH, Kim HJ, Pahk K, Pahk KJ. Sonothrombolysis with an acoustic net-assisted boiling histotripsy: A proof-of-concept study. ULTRASONICS SONOCHEMISTRY 2023; 96:106435. [PMID: 37178667 DOI: 10.1016/j.ultsonch.2023.106435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Whilst sonothrombolysis is a promising and noninvasive ultrasound technique for treating blood clots, bleeding caused by thrombolytic agents used for dissolving clots and potential obstruction of blood flow by detached clots (i.e., embolus) are the major limitations of the current approach. In the present study, a new sonothrombolysis method is proposed for treating embolus without the use of thrombolytic drugs. Our proposed method involves (a) generating a spatially localised acoustic radiation force in a blood vessel against the blood flow to trap moving blood clots (i.e., generation of an acoustic net), (b) producing acoustic cavitation to mechanically destroy the trapped embolus, and (c) acoustically monitoring the trapping and mechanical fractionation processes. Three different ultrasound transducers with different purposes were employed in the proposed method: (1) 1-MHz dual focused ultrasound (dFUS) transducers for capturing moving blood clots, (2) a 2-MHz High Intensity Focused Ultrasound (HIFU) source for fractionating blood clots and (3) a passive acoustic emission detector with broad bandwidth (10 kHz to 20 MHz) for receiving and analysing acoustic waves scattered from a trapped embolus and acoustic cavitation. To demonstrate the feasibility of the proposed method, in vitro experiments with an optically transparent blood vessel-mimicking phantom filled with a blood mimicking fluid and a blood clot (1.2 to 5 mm in diameter) were performed with varying the dFUS and HIFU exposure conditions under various flow conditions (from 1.77 to 6.19 cm/s). A high-speed camera was used to observe the production of acoustic fields, acoustic cavitation formation and blood clot fragmentation within a blood vessel by the proposed method. Numerical simulations of acoustic and temperature fields generated under a given exposure condition were also conducted to further interpret experimental results on the proposed sonothrombolysis. Our results clearly showed that fringe pattern-like acoustic pressure fields (fringe width of 1 mm) produced in a blood vessel by the dFUS captured an embolus (1.2 to 5 mm in diameter) at the flow velocity up to 6.19 cm/s. This was likely to be due to the greater magnitude of the dFUS-induced acoustic radiation force exerted on an embolus in the opposite direction to the flow in a blood vessel than that of the drag force produced by the flow. The acoustically trapped embolus was then mechanically destructed into small pieces of debris (18 to 60 μm sized residual fragments) by the HIFU-induced strong cavitation without damaging the blood vessel walls. We also observed that acoustic emissions emitted from a blood clot captured by the dFUS and cavitation produced by the HIFU were clearly distinguished in the frequency domain. Taken together, these results can suggest that our proposed sonothrombolysis method could be used as a promising tool for treating thrombosis and embolism through capturing and destroying blood clots effectively.
Collapse
Affiliation(s)
- Jeongmin Heo
- Bionics Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jun Hong Park
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Hyo Jun Kim
- LAAS-CNRS, University of Toulouse, CNRS, Toulouse, France
| | - Kisoo Pahk
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ki Joo Pahk
- Department of Biomedical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|
14
|
Lopez A, Osborn J, Irwin R, Khismatullin DB, Clement GT, Myers MR. Vessel Rupture Thresholds for Vessel-Bubble Interactions Using an Earthworm Vasculature Model. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1108-1117. [PMID: 36717284 DOI: 10.1016/j.ultrasmedbio.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 12/08/2022] [Accepted: 12/18/2022] [Indexed: 05/11/2023]
Abstract
OBJECTIVE Intravenous microbubble oscillation in the presence of ultrasound has the potential to yield a wide range of therapeutic benefits. However, the likelihood of vessel damage caused by mechanical effects has not been quantified as a function of the numerous important parameters in therapeutic ultrasound procedures. In this study, we examined the effects of microbubbles injected into the vasculature of the earthworm. It was found that the elastic properties of earthworm blood vessels are similar to those of arteries in older humans, and that earthworms are well suited to the large number of experiments necessary to investigate safety of procedures involving microbubble oscillation in sonicated vessels. METHODS Microbubbles were infused into earthworm vessels, and the rupture time during sonication was recorded as a function of ultrasound frequency, pulse repetition frequency and acoustic pressure. DISCUSSION A modified mechanical index (MMI) was defined that successfully captured the trends in rupture probability and rupture time for the different parameter values, creating a database of vessel rupture thresholds. In the absence of bubbles, the product of MMI squared and rupture time was approximately constant, indicating a possible radiation-force effect. CONCLUSION The MMI was an effective correlating parameter in the presence of bubbles, though the mathematical dependence is not yet apparent. The results of the study are expected to be valuable in designing more refined studies in vertebrate models, as well as informing computational models.
Collapse
Affiliation(s)
- Asis Lopez
- Bioinnovation Ph.D. Program, Biomedical Engineering Department, Tulane University, New Orleans, LA, USA; U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Jenna Osborn
- U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Rachael Irwin
- Biomedical Engineering Department, George Washington University, Washington, DC, USA
| | | | | | | |
Collapse
|
15
|
Mathon B, Navarro V, Lecas S, Roussel D, Charpier S, Carpentier A. Safety Profile of Low-Intensity Pulsed Ultrasound-Induced Blood-Brain Barrier Opening in Non-epileptic Mice and in a Mouse Model of Mesial Temporal Lobe Epilepsy. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1327-1336. [PMID: 36878831 DOI: 10.1016/j.ultrasmedbio.2023.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE It is unknown whether ultrasound-induced blood-brain barrier (BBB) disruption can promote epileptogenesis and how BBB integrity changes over time after sonication. METHODS To gain more insight into the safety profile of ultrasound (US)-induced BBB opening, we determined BBB permeability as well as histological modifications in C57BL/6 adult control mice and in the kainate (KA) model for mesial temporal lobe epilepsy in mice after sonication with low-intensity pulsed ultrasound (LIPU). Microglial and astroglial changes in ipsilateral hippocampus were examined at different time points following BBB disruption by respectively analyzing Iba1 and glial fibrillary acidic protein immunoreactivity. Using intracerebral EEG recordings, we further studied the possible electrophysiological repercussions of a repeated disrupted BBB for seizure generation in nine non-epileptic mice. RESULTS LIPU-induced BBB opening led to transient albumin extravasation and reversible mild astrogliosis, but not to microglial activation in the hippocampus of non-epileptic mice. In KA mice, the transient albumin extravasation into the hippocampus mediated by LIPU-induced BBB opening did not aggravate inflammatory processes and histologic changes that characterize the hippocampal sclerosis. Three LIPU-induced BBB opening did not induce epileptogenicity in non-epileptic mice implanted with depth EEG electrodes. CONCLUSION Our experiments in mice provide persuasive evidence of the safety of LIPU-induced BBB opening as a therapeutic modality for neurological diseases.
Collapse
Affiliation(s)
- Bertrand Mathon
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France; Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, Paris, France.
| | - Vincent Navarro
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France; Epileptology Unit, Department of Neurology, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, Paris, France
| | - Sarah Lecas
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Delphine Roussel
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Stéphane Charpier
- Paris Brain Institute, ICM, INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, Paris, France
| |
Collapse
|
16
|
Zhao X, Wright A, Goertz DE. An optical and acoustic investigation of microbubble cavitation in small channels under therapeutic ultrasound conditions. ULTRASONICS SONOCHEMISTRY 2023; 93:106291. [PMID: 36640460 PMCID: PMC9852793 DOI: 10.1016/j.ultsonch.2023.106291] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 06/04/2023]
Abstract
Therapeutic focused ultrasound in combination with encapsulated microbubbles is being widely investigated for its ability to elicit bioeffects in the microvasculature, such as transient permeabilization for drug delivery or at higher pressures to achieve 'antivascular' effects. While it is well established that the behaviors of microbubbles are altered when they are situated within sufficiently small vessels, there is a paucity of data examining how the bubble population dynamics and emissions change as a function of channel (vessel) diameter over a size range relevant to therapeutic ultrasound, particularly at pressures relevant to antivascular ultrasound. Here we use acoustic emissions detection and high-speed microscopy (10 kframes/s) to examine the behavior of a polydisperse clinically employed agent (Definity®) in wall-less channels as their diameters are scaled from 1200 to 15 µm. Pressures are varied from 0.1 to 3 MPa using either a 5 ms pulse or a sequence of 0.1 ms pulses spaced at 1 ms, both of which have been previously employed in an in vivo context. With increasing pressure, the 1200 µm channel - on the order of small arteries and veins - exhibited inertial cavitation, 1/2 subharmonics and 3/2 ultraharmonics, consistent with numerous previous reports. The 200 and 100 µm channels - in the size range of larger microvessels less affected by therapeutic focused ultrasound - exhibited a distinctly different behavior, having muted development of 1/2 subharmonics and 3/2 ultraharmonics and reduced persistence. These were associated with radiation forces displacing bubbles to the distal wall and inducing clusters that then rapidly dissipated along with emissions. As the diameter transitioned to 50 and then 15 µm - a size regime that is most relevant to therapeutic focused ultrasound - there was a higher threshold for the onset of inertial cavitation as well as subharmonics and ultraharmonics, which importantly had more complex orders that are not normally reported. Clusters also occurred in these channels (e.g. at 3 MPa, the mean lateral and axial sizes were 23 and 72 µm in the 15 µm channel; 50 and 90 µm in the 50 µm channel), however in this case they occupied the entire lumens and displaced the wall boundaries. Damage to the 15 µm channel was observed for both pulse types, but at a lower pressure for the long pulse. Experiments conducted with a 'nanobubble' (<0.45 µm) subpopulation of Definity followed broadly similar features to 'native' Definity, albeit at a higher pressure threshold for inertial cavitation. These results provide new insights into the behavior of microbubbles in small vessels at higher pressures and have implications for therapeutic focused ultrasound cavitation monitoring and control.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| | - Alex Wright
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada
| | - David E Goertz
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| |
Collapse
|
17
|
Yusefi H, Helfield B. The influence of inter-bubble spacing on the resonance response of ultrasound contrast agent microbubbles. ULTRASONICS SONOCHEMISTRY 2022; 90:106191. [PMID: 36223708 PMCID: PMC9563339 DOI: 10.1016/j.ultsonch.2022.106191] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 06/06/2023]
Abstract
Ultrasound-driven microbubbles, typically between 1 and 8 µm in diameter, are resonant scatterers that are employed as diagnostic contrast agents and emerging as potentiators of targeted therapies. Microbubbles are administered in populations whereby their radial dynamics - key to their effectiveness - are greatly affected by intrinsic (e.g. bubble size) and extrinsic (e.g. boundaries) factors. In this work, we aim to understand how two neighbouring microbubbles influence each other. We developed a finite element model of a system of two individual phospholipid-encapsulated microbubbles vibrating in proximity to each other to study the effect of inter-bubble distance on microbubble radial resonance response. For the case of two equal-sized and identical bubbles, each bubble exhibits a decrease between 7 and 10% in the frequency of maximum response (fMR) and an increase in amplitude of maximum response (AMR) by 9-11% as compared to its isolated response in free-space, depending on the bubble size examined. For a system of two unequal-sized microbubbles, the large bubble shows no significant change, however the smaller microbubble shows an increase in fMR by 7-11% and a significant decrease in AMR by 38-52%. Furthermore, in very close proximity the small bubble shows a secondary off-resonance peak at the corresponding fMR of its larger companion microbubble. Our work suggests that frequency-dependent microbubble response is greatly affected by the presence of another bubble, which has implications in both imaging and therapy applications. Furthermore, our work suggests a mechanism by which nanobubbles show significant off-resonance vibrations in the clinical frequency range, a behaviour that has been observed experimentally but heretofore unexplained.
Collapse
Affiliation(s)
- Hossein Yusefi
- Department of Physics, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Brandon Helfield
- Department of Physics, Concordia University, Montreal, Quebec H4B 1R6, Canada; Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada.
| |
Collapse
|
18
|
van Wamel A, Mühlenpfordt M, Hansen R, Healey A, Villanueva FS, Kotopoulis S, Davies CDL, Chen X. Ultrafast Microscopy Imaging of Acoustic Cluster Therapy Bubbles: Activation and Oscillation. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1840-1857. [PMID: 35773079 DOI: 10.1016/j.ultrasmedbio.2022.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/15/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Acoustic Cluster Therapy (ACT®) is a platform for improving drug delivery and has had promising pre-clinical results. A clinical trial is ongoing. ACT® is based on microclusters of microbubbles-microdroplets that, when sonicated, form a large ACT® bubble. The aim of this study was to obtain new knowledge on the dynamic formation and oscillations of ACT® bubbles by ultrafast optical imaging in a microchannel. The high-speed recordings revealed the microbubble-microdroplet fusion, and the gas in the microbubble acted as a vaporization seed for the microdroplet. Subsequently, the bubble grew by gas diffusion from the surrounding medium and became a large ACT® bubble with a diameter of 5-50 μm. A second ultrasound exposure at lower frequency caused the ACT® bubble to oscillate. The recorded oscillations were compared with simulations using the modified Rayleigh-Plesset equation. A term accounting for the physical boundary imposed by the microchannel wall was included. The recorded oscillation amplitudes were approximately 1-2 µm, hence similar to oscillations of smaller contrast agent microbubbles. These findings, together with our previously reported promising pre-clinical therapeutic results, suggest that these oscillations covering a large part of the vessel wall because of the large bubble volume can substantially improve therapeutic outcome.
Collapse
Affiliation(s)
- Annemieke van Wamel
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Spiros Kotopoulis
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
He J, Liu Z, Zhu X, Xia H, Gao H, Lu J. Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy. Pharmaceutics 2022; 14:1642. [PMID: 36015267 PMCID: PMC9414228 DOI: 10.3390/pharmaceutics14081642] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy has an essential role not only in advanced solid tumor therapy intervention but also in society's health at large. Chemoresistance, however, seriously restricts the efficiency and sensitivity of chemotherapeutic agents, representing a significant threat to patients' quality of life and life expectancy. How to reverse chemoresistance, improve efficacy sensitization response, and reduce adverse side effects need to be tackled urgently. Recently, studies on the effect of ultrasonic microbubble cavitation on enhanced tissue permeability and retention (EPR) have attracted the attention of researchers. Compared with the traditional targeted drug delivery regimen, the microbubble cavitation effect, which can be used to enhance the EPR effect, has the advantages of less trauma, low cost, and good sensitization effect, and has significant application prospects. This article reviews the research progress of ultrasound-mediated microbubble cavitation in the treatment of solid tumors and discusses its mechanism of action to provide new ideas for better treatment strategies.
Collapse
Affiliation(s)
- Jide He
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Haizhui Xia
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
20
|
Singh E, Banerjee R. In vivo efficacy & phantom imaging connote the theranostic potential of a drug-loaded lipid nanobubble. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Almasri F, Karshafian R. Synergistic enhancement of cell death by triple combination therapy of docetaxel, ultrasound and microbubbles, and radiotherapy on PC3 a prostate cancer cell line. Heliyon 2022; 8:e10213. [PMID: 36033334 PMCID: PMC9404355 DOI: 10.1016/j.heliyon.2022.e10213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/30/2022] [Accepted: 08/04/2022] [Indexed: 11/30/2022] Open
Abstract
The application of ultrasound and microbubbles (USMB) has been shown to enhance both chemotherapy and radiotherapy. This study investigated the potential of triple combination therapy comprised of USMB, docetaxel (Taxotere: TXT) chemotherapy and XRT to enhance treatment efficacy. Prostate cancer (PC3) cells in suspension were treated with various combinations of USMB, chemotherapy and radiotherapy. Cells were treated with ultrasound and microbubbles (500 kHz pulse center frequency, 580 kPa peak negative pressure, 10 μs pulse duration, 60 s insonation time and 2% Definity microbubbles (v/v)), XRT (2 Gy), and Taxotere (TXT) at concentrations ranging from 0.001 to 0.1 nM for 5- and 120-minutes duration. Following treatment, cell viability was assessed using a clonogenic assay. Therapeutic efficiency of the combined treatments depended on chemotherapy and microbubble exposure conditions. Under the exposure conditions of the study, the triple combination therapy synergistically enhanced clonogenic cell death compared to single and double combination therapy. Cell viability of ∼2% was achieved with the triple combination therapy corresponding to ∼29, ∼37, and ∼38 folds decrease compared to XRT (57%), USMB (74%) and TXT (76%) alone conditions, respectively. In addition, the triple combination therapy decreased cell viability by ∼29, ∼19- and ∼11 folds compared to TXT2hr + USMB (58%), TXT2hr + XRT (37%), and USMB + XRT (22%), respectively. The in vivo PC3 tumours showed that USMB significantly enhanced cell death through detection of apoptosis (TUNEL) with both TXT and TXT + XRT. The study demonstrated that the triple combination therapy can significantly enhance cell death in prostate cancer cells both in vitro and in vivo under relatively low chemotherapy and ionizing radiation doses.
Collapse
Affiliation(s)
- Firas Almasri
- Department of Physics, Ryerson University, Toronto, Ontario, Canada.,Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Hawally, Kuwait.,Centre for Education Studies, University of Warwick, Coventry, UK.,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, Canada
| | - Raffi Karshafian
- Department of Physics, Ryerson University, Toronto, Ontario, Canada.,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Díaz-Alejo JF, González Gómez I, Earl J. Ultrasounds in cancer therapy: A summary of their use and unexplored potential. Oncol Rev 2022; 16:531. [PMID: 35340884 PMCID: PMC8941342 DOI: 10.4081/oncol.2022.531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/17/2021] [Indexed: 11/26/2022] Open
Abstract
Ultrasounds (US) are a non-ionizing mechanical wave, with less adverse effects than conventional pharmacological or surgical treatments. Different biological effects are induced in tissues and cells by ultrasound actuation depending on acoustic parameters, such as the wave intensity, frequency and treatment dose. This non-ionizing radiation has considerable applications in biomedicine including surgery, medical imaging, physical therapy and cancer therapy. Depending on the wave intensity, US are applied as high-intensity ultrasounds (HIUS) and low-intensity pulsed ultrasounds (LIPUS), with different effects on cells and tissues. HIUS produce thermal and mechanical effects, resulting in a large localized temperature increase, leading to tissue ablation and even tumor necrosis. This can be achieved by focusing low intensity waves emitted from different electrically shifted transducers, known as high-intensity focused ultrasounds (HIFU). LIPUS have been used extensively as a therapeutic, surgical and diagnostic tool, with diverse biological effects observed in tissues and cultured cells. US represent a non-invasive treatment strategy that can be applied to selected areas of the body, with limited adverse effects. In fact, tumor ablation using HIFU has been used as a curative treatment in patients with an early-stage pancreatic tumor and is an effective palliative treatment in patients with advanced stage disease. However, the biological effects, dose standardization, benefit-risk ratio and safety are not fully understood. Thus, it is an emerging field that requires further research in order to reach its full potential.
Collapse
Affiliation(s)
- Jesús Frutos Díaz-Alejo
- Molecular Epidemiology and Predictive Tumor Markers Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid
- Faculty of Medicine and Health Sciences, University of Alcalá de Henares (UAH), Madrid
| | | | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid
- Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| |
Collapse
|
23
|
Sharma D, Hussein F, Law N, Farhat G, Tarapacki C, Sannachi L, Giles A, Czarnota GJ. Focused Ultrasound Stimulation of Microbubbles in Combination With Radiotherapy for Acute Damage of Breast Cancer Xenograft Model. Technol Cancer Res Treat 2022; 21:15330338221132925. [DOI: 10.1177/15330338221132925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective: Several studies have focused on the use of ultrasound-stimulated microbubbles (USMB) to induce vascular damage in order to enhance tumor response to radiation. Methods: In this study, power Doppler imaging was used along with immunohistochemistry to investigate the effects of combining radiation therapy (XRT) and USMB using an ultrasound-guided focused ultrasound (FUS) therapy system in a breast cancer xenograft model. Specifically, MDA-MB-231 breast cancer xenograft tumors were induced in severe combined immuno-deficient female mice. The mice were treated with FUS alone, ultrasound and microbubbles (FUS + MB) alone, 8 Gy XRT alone, or a combined treatment consisting of ultrasound, microbubbles, and XRT (FUS + MB + XRT). Power Doppler imaging was conducted before and 24 h after treatment, at which time mice were sacrificed and tumors assessed histologically. The immunohistochemical analysis included terminal deoxynucleotidyl transferase dUTP nick end labeling, hematoxylin and eosin, cluster of differentiation-31 (CD31), Ki-67, carbonic anhydrase (CA-9), and ceramide labeling. Results: Tumors receiving treatment of FUS + MB combined with XRT demonstrated significant increase in cell death (p = 0.0006) compared to control group. Furthermore, CD31 and Power Doppler analysis revealed reduced tumor vascularization with combined treatment indicating ( P < .0001) and ( P = .0001), respectively compared to the control group. Additionally, lesser number of proliferating cells with enhanced tumor hypoxia, and ceramide content were also reported in group receiving a treatment of FUS + MB + XRT. Conclusion: The study results demonstrate that the combination of USMB with XRT enhances treatment outcomes.
Collapse
Affiliation(s)
- Deepa Sharma
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Farah Hussein
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Niki Law
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Golnaz Farhat
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Lakshmanan Sannachi
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| | - Anoja Giles
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Gregory J. Czarnota
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Adams C, Jones RM, Yang SD, Kan WM, Leung K, Zhou Y, Lee KU, Huang Y, Hynynen K. Implementation of a Skull-Conformal Phased Array for Transcranial Focused Ultrasound Therapy. IEEE Trans Biomed Eng 2021; 68:3457-3468. [PMID: 33950835 PMCID: PMC11979958 DOI: 10.1109/tbme.2021.3077802] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To implement a skull-conformal phased array for ultrasound-guided transcranial focused ultrasound therapy with improved patient comfort. METHODS Using patient-specific computed tomography and MRI neuroimaging data, tightly-conforming helmet scaffolds were designed computationally. The helmet scaffolds were designed to hold reusable transducer modules at near-normal incidence in an optimal configuration for the treatment location(s) of interest. Numerical simulations of trans-skull ultrasound propagation were performed to evaluate different conformal array designs and to compare with hemispherical arrays similar to those employed clinically. A 4096-element phased array was constructed by 3D printing a helmet scaffold optimised for an ex vivo human skullcap, and its performance was evaluated via benchtop and in vivo experiments. RESULTS Acoustic field measurements confirmed the system's ability to focus through human skull bone using simulation-based transcranial aberration corrections. Preliminary in vivo testing demonstrated safe trans-human skull blood-brain barrier (BBB) opening in rodents. CONCLUSION Patient-specific conformal ultrasound phased arrays appear to be a feasible and safe approach for conducting transcranial BBB opening procedures. SIGNIFICANCE Skull-conformal phased arrays stand to improve patient comfort and have the potential to accelerate the adoption of transcranial FUS therapy by improving access to the technology.
Collapse
|
25
|
Feng S, Qiao W, Tang J, Yu Y, Gao S, Liu Z, Zhu X. Chemotherapy Augmentation Using Low-Intensity Ultrasound Combined with Microbubbles with Different Mechanical Indexes in a Pancreatic Cancer Model. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3221-3230. [PMID: 34362582 DOI: 10.1016/j.ultrasmedbio.2021.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
The aim of the study was to explore the optimal mechanical indexes (MIs) for low-intensity ultrasound (LIUS) combined with microbubbles to enhance tumor blood perfusion and improve drug concentration in pancreatic cancer-bearing nude mice. Fifty-four nude mice bearing bilateral pancreatic tumors on the hind legs were randomly divided into three groups (the MI was set at 0.3, 0.7 and 1.1 in groups A, B and C, respectively). Five nude mice in each group were intravenously injected with the fluorescent dye DiR iodide (DiIC18(7),1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide); for each mouse, one tumor was treated with LIUS combined with microbubbles, and the contralateral tumor was exposed to sham ultrasound. In vivo fluorescence imaging was performed to detect the enrichment of intratumoral DiR iodide. Twelve mice in each group were intravenously injected with doxorubicin (DOX) and underwent ultrasound therapy as described above. Tumor blood perfusion changes were quantitatively evaluated with pre- and post-treatment contrast-enhanced ultrasound (CEUS, MI = 0.08). One hour after the post-treatment CEUS, nude mice were sacrificed to determine the DOX concentration in tumor tissue; one mouse in each group was sacrificed after ultrasound treatment for tumor hematoxylin-eosin staining examination. CEUS quantitative analysis and in vivo fluorescence images confirmed that LIUS at MI = 0.3 combined with microbubbles was able to enhance tumor blood flow and increase regional fluorescence dye DiR iodide concentration. The DOX concentration on the therapeutic side was significantly higher than that on the control side after ultrasound-stimulated (MI = 0.3) microbubble cavitation (USMC) treatment (1.45 ± 0.53 μg/g vs. 1.07 ± 0.46 μg/g, t = -5.163, p = 0.001). However, in groups B and C, there were no significant differences in DOX concentration between the therapeutic and control sides (Z = -0.297, -0.357, p = 0.766, 0.721). No hemorrhage or other tissue damage was observed in hematoxylin-eosin-stained tumor specimens of both sides in all groups. LIUS at MI = 0.3 combined with microbubbles was able to enhance tumor blood perfusion and improve local drug concentration in nude mice bearing pancreatic cancer.
Collapse
Affiliation(s)
- Shuang Feng
- Department of Ultrasound, General Hospital of Southern Theatre Command, Guangzhou, China
| | - Wei Qiao
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Jiawei Tang
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yanlan Yu
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Shunji Gao
- Department of Ultrasound, General Hospital of Central Theatre Command, Wuhan, China
| | - Zheng Liu
- Department of Ultrasound, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiansheng Zhu
- Department of Ultrasound, General Hospital of Southern Theatre Command, Guangzhou, China.
| |
Collapse
|
26
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
27
|
Blood-brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses. Proc Natl Acad Sci U S A 2021; 118:2021915118. [PMID: 33906946 DOI: 10.1073/pnas.2021915118] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation.
Collapse
|
28
|
Lawanprasert A, Chau A, Sloand JN, Hannifin S, Medina SH. Tuning the Interfacial Properties of Fluorous Colloids Toward Ultrasound Programmable Bioactivity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:5989-5998. [PMID: 33522791 DOI: 10.1021/acsami.0c20352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Liquid-in-liquid emulsions are kinetically stable colloids that undergo liquid-to-gas phase transitions in response to thermal or acoustic stimuli. Perfluorocarbons (PFCs) are preferred species as their highly fluorinated nature imparts unique properties that are unparalleled by nonfluorinated counterparts. However, traditional methods to prepare PFC emulsions lack the ability to precisely tune the thermodynamic stability of the fluorous-water interphase and consequently control their vaporization behavior. Here, we report a privileged fluoroalkanoic acid that undergoes concentration-dependent assembly on the surfaces of fluorous droplets to modulate interfacial tension. This allows for the rational formulation of orthogonal PFC droplets that can be programmed to vaporize at specified ultrasound powers. We exploit this behavior in two exemplary biomedical settings by developing emulsions that aid ultrasound-mediated hemostasis and enable bioorthogonal delivery of molecular sensors to mammalian cells. Mechanistic insights gained from these studies can be generalized to tune the thermodynamic interfacial equilibria of PFC emulsions toward designing controllable tools for precision medicine.
Collapse
Affiliation(s)
- Atip Lawanprasert
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Alda Chau
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Janna N Sloand
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Sean Hannifin
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
- Immunology Graduate Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Scott H Medina
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
- Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
29
|
Bao Y, Chen J, Huang P, Tong W. Synergistic Effects of Acoustics-based Therapy and Immunotherapy in Cancer Treatment. BIO INTEGRATION 2021. [DOI: 10.15212/bioi-2021-0007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Cancer is an intractable disease and has ability to escape immunological recognition. Cancer immunotherapy to enhance the autogenous immune response to cancer tissue is reported to be the most promising method for cancer treatment. After the release of damage-associated molecular patterns, dendritic cells come mature and then recruit activated T cells to induce immune response. To trigger the release of cancer associated antigens, cancer acoustics-based therapy has various prominent advantages and has been reported in various research. In this review, we classified the acoustics-based therapy into sonopyrolysis-, sonoporation-, and sonoluminescence-based therapy. Then, detailed mechanisms of these therapies are discussed to show the status of cancer immunotherapy induced by acoustics-based therapy in quo. Finally, we express some future prospects in this research field and make some predictions of its development direction
Collapse
Affiliation(s)
- Yuheng Bao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
30
|
Abstract
Gas-filled microbubbles are currently in clinical use as blood pool contrast agents for ultrasound imaging. The goal of this review is to discuss the trends and issues related to these relatively unusual intravascular materials, which are not small molecules per se, not polymers, not even nanoparticles, but larger micrometer size structures, compressible, flexible, elastic, and deformable. The intent is to connect current research and initial studies from 2 to 3 decades ago, tied to gas exchange between the bubbles and surrounding biological medium, in the following areas of focus: (1) parameters of microbubble movement in relation to vasculature specifics; (2) gas uptake and loss from the bubbles in the vasculature; (3) adhesion of microbubbles to target receptors in the vasculature; and (4) microbubble interaction with the surrounding vessels and tissues during insonation.Microbubbles are generally safe and require orders of magnitude lower material doses than x-ray and magnetic resonance imaging contrast agents. Application of microbubbles will soon extend beyond blood pool contrast and tissue perfusion imaging. Microbubbles can probe molecular and cellular biomarkers of disease by targeted contrast ultrasound imaging. This approach is now in clinical trials, for example, with the aim to detect and delineate tumor nodes in prostate, breast, and ovarian cancer. Imaging of inflammation, ischemia-reperfusion injury, and ischemic memory is also feasible. More importantly, intravascular microbubbles can be used for local deposition of focused ultrasound energy to enhance drug and gene delivery to cells and tissues, across endothelial barrier, especially blood-brain barrier.Overall, microbubble behavior, stability and in vivo lifetime, bioeffects upon the action of ultrasound and resulting enhancement of drug and gene delivery, as well as targeted imaging are critically dependent on the events of gas exchange between the bubbles and surrounding media, as outlined in this review.
Collapse
Affiliation(s)
- Alexander L Klibanov
- From the Cardiovascular Division, Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine; and Departments of Biomedical Engineering, and Radiology, University of Virginia, Charlottesville, VA
| |
Collapse
|
31
|
Yang Y, Li Q, Guo X, Tu J, Zhang D. Mechanisms underlying sonoporation: Interaction between microbubbles and cells. ULTRASONICS SONOCHEMISTRY 2020; 67:105096. [PMID: 32278246 DOI: 10.1016/j.ultsonch.2020.105096] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 05/04/2023]
Abstract
The past several decades have witnessed great progress in "smart drug delivery", an advance technology that can deliver genes or drugs into specific locations of patients' body with enhanced delivery efficiency. Ultrasound-activated mechanical force induced by the interactions between microbubbles and cells, which can stimulate so-called "sonoporation" process, has been regarded as one of the most promising candidates to realize spatiotemporal-controllable drug delivery to selected regions. Both experimental and numerical studies were performed to get in-depth understanding on how the microbubbles interact with cells during sonoporation processes, under different impact parameters. The current work gives an overview of the general mechanism underlying microbubble-mediated sonoporation, and the possible impact factors (e.g., the properties of cavitation agents and cells, acoustical driving parameters and bubble/cell micro-environment) that could affect sonoporation outcomes. Finally, current progress and considerations of sonoporation in clinical applications are reviewed also.
Collapse
Affiliation(s)
- Yanye Yang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Qunying Li
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiasheng Guo
- 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.
| | - 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
|
Pellow C, Abenojar EC, Exner AA, Zheng G, Goertz DE. Concurrent visual and acoustic tracking of passive and active delivery of nanobubbles to tumors. Am J Cancer Res 2020; 10:11690-11706. [PMID: 33052241 PMCID: PMC7545999 DOI: 10.7150/thno.51316] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background: There has been growing interest in nanobubbles for their potential to extend bubble-mediated ultrasound approaches beyond that of their larger microbubble counterparts. In particular, the smaller scale of nanobubbles may enable them to access the tumor extravascular compartment for imaging and therapy in closer proximity to cancer cells. Compelling preliminary demonstrations of the imaging and therapeutic abilities of nanobubbles have thus emerged, with emphasis on their ability to extravasate. However, studies to date rely on indirect histologic evidence that cannot confirm whether the structures remain intact beyond the vasculature - leaving their extravascular potential largely untapped. Methods: Nanobubble acoustic scattering was assessed using a recently reported ultra-stable formulation at low concentration (106 mL-1) and frequency (1 MHz), over a range of pressures (100-1500 kPa) in a channel phantom. The pressure-dependent response was utilized as a basis for in vivo experiments where ultrasound transmitters and receivers were integrated into a window chamber for simultaneous intravital multiphoton microscopy and acoustic monitoring in tumor-affected microcirculation. Microscopy and acoustic data were utilized to assess passive and active delivery of nanobubbles and determine whether they remained intact beyond the vasculature. Results: Nanobubbles exhibit pressure-dependent nonlinear acoustic scattering. Nanobubbles are also found to have prolonged acoustic vascular pharmacokinetics, and passively extravasate intact into tumors. Ultrasound stimulation of nanobubbles is shown to actively enhance the delivery of both intact nanobubbles and shell material, increasing their spatial bioavailability deeper into the extravascular space. A range of acute vascular effects were also observed. Conclusion: This study presents the first direct evidence that nanobubbles passively and actively extravasate intact in tumor tissue, and is the first to directly capture acute vascular events from ultrasound-stimulation of nanobubbles. The insights gained here demonstrate an important step towards unlocking the potential of nanobubbles and extending ultrasound-based applications.
Collapse
|
33
|
Naude J, Méndez F, Yepes C, Navarrete M, Cienfuegos-Pelaes RF, Moumtadi F. Frequency response curves for a Mooney-Rivlin hyperelastic microbubble oscillating as a contrast agent in an acoustic pressure field. ULTRASONICS 2020; 107:106161. [PMID: 32402859 DOI: 10.1016/j.ultras.2020.106161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 06/11/2023]
Abstract
In this work, we have developed numerical simulations and weakly nonlinear analysis based on the multiple-scales perturbation technique for a coated microbubble that performs radial pulsations subject to an acoustic pressure disturbance in the far-field and whose encapsulated hyperelastic material obeys the Mooney-Rivlin equation. Departing from an elastic coating as a hyperelastic shell of finite thickness, we assume eventually that the shell is of very small thickness in comparison with the microbubble radius. Under this condition, we then perform weakly nonlinear analysis, to identify resonance conditions for small pressure disturbances of the acoustic field. In parallel and also for the limit of small thickness, we have carried out numerical simulations of the radial motion of the microbubble, identifying the onset of limit cycles via the construction of Poincare maps. Under both schemes, we have recognized the importance of two dimensionless hyperelastic parameters that dictate the main behavior of the oscillations: α∗ and β∗. Decreasing the values of these parameters, the resonance conditions are drastically amplified, which is an expected result because of the weak rigidity of the hyperelastic solid, prevails. In this manner, we suggest that moderate values for these previous parameters can be widely advisable when, in medical diagnostic applications, we are applying microbubbles as contrast agents. Therefore, we recommend widely the use of shell softens, because in this case the amplitude of radial pulsation is always amplified.
Collapse
Affiliation(s)
- J Naude
- Departamento de Termofluidos, Facultad de Ingeniería, UNAM, 04510 CDMX, Mexico
| | - F Méndez
- Departamento de Termofluidos, Facultad de Ingeniería, UNAM, 04510 CDMX, Mexico.
| | - C Yepes
- Departamento de Termofluidos, Facultad de Ingeniería, UNAM, 04510 CDMX, Mexico
| | - M Navarrete
- Polo Universitario de Tecnología Avanzada, UNAM, 66629 Apodaca N. L., Mexico
| | | | - F Moumtadi
- Departamento de Ingeniería Electrónica, Facultad de Ingeniería, UNAM, 04510 CDMX, Mexico
| |
Collapse
|
34
|
Liu DD, Ullah M, Concepcion W, Dahl JJ, Thakor AS. The role of ultrasound in enhancing mesenchymal stromal cell-based therapies. Stem Cells Transl Med 2020; 9:850-866. [PMID: 32157802 PMCID: PMC7381806 DOI: 10.1002/sctm.19-0391] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been a popular platform for cell-based therapy in regenerative medicine due to their propensity to home to damaged tissue and act as a repository of regenerative molecules that can promote tissue repair and exert immunomodulatory effects. Accordingly, a great deal of research has gone into optimizing MSC homing and increasing their secretion of therapeutic molecules. A variety of methods have been used to these ends, but one emerging technique gaining significant interest is the use of ultrasound. Sound waves exert mechanical pressure on cells, activating mechano-transduction pathways and altering gene expression. Ultrasound has been applied both to cultured MSCs to modulate self-renewal and differentiation, and to tissues-of-interest to make them a more attractive target for MSC homing. Here, we review the various applications of ultrasound to MSC-based therapies, including low-intensity pulsed ultrasound, pulsed focused ultrasound, and extracorporeal shockwave therapy, as well as the use of adjunctive therapies such as microbubbles. At a molecular level, it seems that ultrasound transiently generates a local gradient of cytokines, growth factors, and adhesion molecules that facilitate MSC homing. However, the molecular mechanisms underlying these methods are far from fully elucidated and may differ depending on the ultrasound parameters. We thus put forth minimal criteria for ultrasound parameter reporting, in order to ensure reproducibility of studies in the field. A deeper understanding of these mechanisms will enhance our ability to optimize this promising therapy to assist MSC-based approaches in regenerative medicine.
Collapse
Affiliation(s)
- Daniel D. Liu
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| | | | - Jeremy J. Dahl
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| | - Avnesh S. Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of RadiologyStanford UniversityPalo AltoCalifornia
| |
Collapse
|
35
|
Pellow C, O'Reilly MA, Hynynen K, Zheng G, Goertz DE. Simultaneous Intravital Optical and Acoustic Monitoring of Ultrasound-Triggered Nanobubble Generation and Extravasation. NANO LETTERS 2020; 20:4512-4519. [PMID: 32374617 DOI: 10.1021/acs.nanolett.0c01310] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ultrasound-activated nanobubbles are being widely investigated as contrast agents and therapeutic vehicles. Nanobubbles hold potential for accessing the tumor extravascular compartment, though this relies on clinically debated passive accumulation for which evidence to date is indirect. We recently reported ultrasound-triggered conversion of high payload porphyrin-encapsulated microbubbles to nanobubbles, with actively enhanced permeability for local delivery. This platform holds implications for optical/ultrasound-based imaging and therapeutics. While promising, it remains to be established how nanobubbles are generated and whether they extravasate intact. Here, insights into the conversion process are reported, complemented by novel simultaneous intravital and acoustic monitoring in tumor-affected functional circulation. The first direct acoustic evidence of extravascular intact nanobubbles are presented. These insights collectively advance this delivery platform and multimodal micro- and nanobubbles, extending their utility for imaging and therapeutics within and beyond the vasculature.
Collapse
Affiliation(s)
- Carly Pellow
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 2C1, Canada
| | - Meaghan A O'Reilly
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| | - Kullervo Hynynen
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| | - Gang Zheng
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 2C1, Canada
| | - David E Goertz
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| |
Collapse
|
36
|
Kooiman K, Roovers S, Langeveld SAG, Kleven RT, Dewitte H, O'Reilly MA, Escoffre JM, Bouakaz A, Verweij MD, Hynynen K, Lentacker I, Stride E, Holland CK. Ultrasound-Responsive Cavitation Nuclei for Therapy and Drug Delivery. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1296-1325. [PMID: 32165014 PMCID: PMC7189181 DOI: 10.1016/j.ultrasmedbio.2020.01.002] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 05/03/2023]
Abstract
Therapeutic ultrasound strategies that harness the mechanical activity of cavitation nuclei for beneficial tissue bio-effects are actively under development. The mechanical oscillations of circulating microbubbles, the most widely investigated cavitation nuclei, which may also encapsulate or shield a therapeutic agent in the bloodstream, trigger and promote localized uptake. Oscillating microbubbles can create stresses either on nearby tissue or in surrounding fluid to enhance drug penetration and efficacy in the brain, spinal cord, vasculature, immune system, biofilm or tumors. This review summarizes recent investigations that have elucidated interactions of ultrasound and cavitation nuclei with cells, the treatment of tumors, immunotherapy, the blood-brain and blood-spinal cord barriers, sonothrombolysis, cardiovascular drug delivery and sonobactericide. In particular, an overview of salient ultrasound features, drug delivery vehicles, therapeutic transport routes and pre-clinical and clinical studies is provided. Successful implementation of ultrasound and cavitation nuclei-mediated drug delivery has the potential to change the way drugs are administered systemically, resulting in more effective therapeutics and less-invasive treatments.
Collapse
Affiliation(s)
- Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Silke Roovers
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Simone A G Langeveld
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert T Kleven
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Medical School of the Vrije Universiteit Brussel, Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Martin D Verweij
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Christy K Holland
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA; Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
37
|
Goel L, Jiang X. Advances in Sonothrombolysis Techniques Using Piezoelectric Transducers. SENSORS 2020; 20:s20051288. [PMID: 32120902 PMCID: PMC7085655 DOI: 10.3390/s20051288] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
One of the great advancements in the applications of piezoelectric materials is the application for therapeutic medical ultrasound for sonothrombolysis. Sonothrombolysis is a promising ultrasound based technique to treat blood clots compared to conventional thrombolytic treatments or mechanical thrombectomy. Recent clinical trials using transcranial Doppler ultrasound, microbubble mediated sonothrombolysis, and catheter directed sonothrombolysis have shown promise. However, these conventional sonothrombolysis techniques still pose clinical safety limitations, preventing their application for standard of care. Recent advances in sonothrombolysis techniques including targeted and drug loaded microbubbles, phase change nanodroplets, high intensity focused ultrasound, histotripsy, and improved intravascular transducers, address some of the limitations of conventional sonothrombolysis treatments. Here, we review the strengths and limitations of these latest pre-clincial advancements for sonothrombolysis and their potential to improve clinical blood clot treatments.
Collapse
Affiliation(s)
- Leela Goel
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA;
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Raleigh, NC 27695-7910, USA
| | - Xiaoning Jiang
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA;
- Correspondence: ; Tel.: +1-919-515-5240
| |
Collapse
|
38
|
Yemane PT, Åslund AKO, Snipstad S, Bjørkøy A, Grendstad K, Berg S, Mørch Y, Torp SH, Hansen R, Davies CDL. Effect of Ultrasound on the Vasculature and Extravasation of Nanoscale Particles Imaged in Real Time. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:3028-3041. [PMID: 31474384 DOI: 10.1016/j.ultrasmedbio.2019.07.683] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Ultrasound and microbubbles have been found to improve the delivery of drugs and nanoparticles to tumor tissue. To obtain new knowledge on the influence of vascular parameters on extravasation and to elucidate the effect of acoustic pressure on extravasation and penetration of nanoscale particles into the extracellular matrix, real-time intravital multiphoton microscopy was performed during sonication of tumors growing in dorsal window chambers. The impact of vessel diameter, vessel structure and blood flow was characterized. Fluorescein isothiocyanate-dextran (2 MDa) was injected to visualize blood vessels. Mechanical indexes (MI) of 0.2-0.8 and in-house-made, nanoparticle-stabilized microbubbles or Sonovue were applied. The rate and extent of penetration into the extracellular matrix increased with increasing MI. However, to achieve extravasation, smaller vessels required MIs (0.8) higher than those of blood vessels with larger diameters. Ultrasound changed the blood flow rate and direction. Interestingly, the majority of extravasations occurred at vessel branching points.
Collapse
Affiliation(s)
- Petros T Yemane
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andreas K O Åslund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Stroke Unit, Department of Internal Medicine, St. Olav's Hospital, Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristin Grendstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Sverre H Torp
- Department of Pathology, St. Olav's Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Hansen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | | |
Collapse
|
39
|
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
|
40
|
Shin J, Kong C, Cho JS, Lee J, Koh CS, Yoon MS, Na YC, Chang WS, Chang JW. Focused ultrasound-mediated noninvasive blood-brain barrier modulation: preclinical examination of efficacy and safety in various sonication parameters. Neurosurg Focus 2019; 44:E15. [PMID: 29385915 DOI: 10.3171/2017.11.focus17627] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The application of pharmacological therapeutics in neurological disorders is limited by the ability of these agents to penetrate the blood-brain barrier (BBB). Focused ultrasound (FUS) has recently gained attention for its potential application as a method for locally opening the BBB and thereby facilitating drug delivery into the brain parenchyma. However, this method still requires optimization to maximize its safety and efficacy for clinical use. In the present study, the authors examined several sonication parameters of FUS influencing BBB opening in small animals. METHODS Changes in BBB permeability were observed during transcranial sonication using low-intensity FUS in 20 adult male Sprague-Dawley rats. The authors examined the effects of FUS sonication with different sonication parameters, varying acoustic pressure, center frequency, burst duration, microbubble (MB) type, MB dose, pulse repetition frequency (PRF), and total exposure time. The focal region of BBB opening was identified by Evans blue dye. Additionally, H & E staining was used to identify blood vessel damage. RESULTS Acoustic pressure amplitude and burst duration were closely associated with enhancement of BBB opening efficiency, but these parameters were also highly correlated with tissue damage in the sonicated region. In contrast, MB types, MB dose, total exposure time, and PRF had an influence on BBB opening without conspicuous tissue damage after FUS sonication. CONCLUSIONS The study aimed to identify these influential conditions and provide safety and efficacy values for further studies. Future work based on the current results is anticipated to facilitate the implementation of FUS sonication for drug delivery in various CNS disease states in the near future.
Collapse
Affiliation(s)
- Jaewoo Shin
- Department of Neurosurgery, Brain Research Institute.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul; and
| | - Chanho Kong
- Department of Neurosurgery, Brain Research Institute
| | - Jae Sung Cho
- Department of Neurosurgery, Brain Research Institute
| | - Jihyeon Lee
- Department of Neurosurgery, Brain Research Institute
| | - Chin Su Koh
- Department of Neurosurgery, Brain Research Institute
| | - Min-Sik Yoon
- Department of Neurosurgery, Brain Research Institute
| | - Young Cheol Na
- Department of Neurosurgery, Catholic Kwandong University College of Medicine, International St. Mary's Hospital, Incheon Metropolitan City, Korea
| | - Won Seok Chang
- Department of Neurosurgery, Brain Research Institute.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul; and
| | - Jin Woo Chang
- Department of Neurosurgery, Brain Research Institute.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul; and
| |
Collapse
|
41
|
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
|
42
|
Abstract
All solid tumours require a vascular supply in order to progress. Although the ability to induce angiogenesis (new blood vessel growth) has long been regarded as essential to this purpose, thus far, anti-angiogenic therapies have shown only modest efficacy in patients. Importantly, overshadowed by the literature on tumour angiogenesis is a long-standing, but continually emerging, body of research indicating that tumours can grow instead by hijacking pre-existing blood vessels of the surrounding nonmalignant tissue. This process, termed vessel co-option, is a frequently overlooked mechanism of tumour vascularization that can influence disease progression, metastasis and response to treatment. In this Review, we describe the evidence that tumours located at numerous anatomical sites can exploit vessel co-option. We also discuss the proposed molecular mechanisms involved and the multifaceted implications of vessel co-option for patient outcomes.
Collapse
Affiliation(s)
- Elizabeth A Kuczynski
- Bioscience, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals St Augustinus, University of Antwerp, Wilrijk-Antwerp, Belgium
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Francesco Pezzella
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Andrew R Reynolds
- Tumour Biology Team, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
- Oncology Translational Medicine Unit, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
43
|
Sinharay S, Tu TW, Kovacs ZI, Schreiber-Stainthorp W, Sundby M, Zhang X, Papadakis GZ, Reid WC, Frank JA, Hammoud DA. In vivo imaging of sterile microglial activation in rat brain after disrupting the blood-brain barrier with pulsed focused ultrasound: [18F]DPA-714 PET study. J Neuroinflammation 2019; 16:155. [PMID: 31345243 PMCID: PMC6657093 DOI: 10.1186/s12974-019-1543-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 12/26/2022] Open
Abstract
Background Magnetic resonance imaging (MRI)-guided pulsed focused ultrasound combined with the infusion of microbubbles (pFUS+MB) induces transient blood-brain barrier opening (BBBO) in targeted regions. pFUS+MB, through the facilitation of neurotherapeutics’ delivery, has been advocated as an adjuvant treatment for neurodegenerative diseases and malignancies. Sterile neuroinflammation has been recently described following pFUS+MB BBBO. In this study, we used PET imaging with [18F]-DPA714, a biomarker of translocator protein (TSPO), to assess for neuroinflammatory changes following single and multiple pFUS+MB sessions. Methods Three groups of Sprague-Dawley female rats received MRI-guided pFUS+MB (Optison™; 5–8 × 107 MB/rat) treatments to the left frontal cortex and right hippocampus. Group A rats were sonicated once. Group B rats were sonicated twice and group C rats were sonicated six times on weekly basis. Passive cavitation detection feedback (PCD) controlled the peak negative pressure during sonication. We performed T1-weighted scans immediately after sonication to assess efficiency of BBBO and T2*-weighted scans to evaluate for hypointense voxels. [18F]DPA-714 PET/CT scans were acquired after the BBB had closed, 24 h after sonication in group A and within an average of 10 days from the last sonication in groups B and C. Ratios of T1 enhancement, T2* values, and [18F]DPA-714 percent injected dose/cc (%ID/cc) values in the targeted areas to the contralateral brain were calculated. Histological assessment for microglial activation/astrocytosis was performed. Results In all groups, [18F]DPA-714 binding was increased at the sonicated compared to non-sonicated brain (%ID/cc ratios > 1). Immunohistopathology showed increased staining for microglial and astrocytic markers in the sonicated frontal cortex compared to contralateral brain and to a lesser extent in the sonicated hippocampus. Using MRI, we documented BBB disruption immediately after sonication with resolution of BBBO 24 h later. We found more T2* hypointense voxels with increasing number of sonications. In a longitudinal group of animals imaged after two and after six sonications, there was no cumulative increase of neuroinflammation on PET. Conclusion Using [18F]DPA-714 PET, we documented in vivo neuroinflammatory changes in association with pFUS+MB. Our protocol (utilizing PCD feedback to minimize damage) resulted in neuroinflammation visualized 24 h post one sonication. Our findings were supported by immunohistochemistry showing microglial activation and astrocytosis. Experimental sonication parameters intended for BBB disruption should be evaluated for neuroinflammatory sequelae prior to implementation in clinical trials. Electronic supplementary material The online version of this article (10.1186/s12974-019-1543-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanhita Sinharay
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.,University of Texas, MD Anderson Cancer Center, Houston, USA
| | - Tsang-Wei Tu
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Radiology, Howard University, Washington DC, USA
| | - Zsofia I Kovacs
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,Institute for Biomedical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - William Schreiber-Stainthorp
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Xiang Zhang
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Georgios Z Papadakis
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.,Department of Radiology, University of Crete and Department of Medical Imaging Heraklion University Hospital, Crete, Greece
| | - William C Reid
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Dima A Hammoud
- Hammoud Laboratory, Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health, 10 Center Drive, Building 10, Room 1C-368, Bethesda, MD, 20892, USA.
| |
Collapse
|
44
|
Choi KH, Kim JH. Therapeutic Applications of Ultrasound in Neurological Diseases. ACTA ACUST UNITED AC 2019. [DOI: 10.31728/jnn.2019.00046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
45
|
Helfield B. A Review of Phospholipid Encapsulated Ultrasound Contrast Agent Microbubble Physics. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:282-300. [PMID: 30413335 DOI: 10.1016/j.ultrasmedbio.2018.09.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/11/2018] [Accepted: 09/20/2018] [Indexed: 06/08/2023]
Abstract
Ultrasound contrast agent microbubbles have expanded the utility of biomedical ultrasound from anatomic imaging to the assessment of microvascular blood flow characteristics and ultrasound-assisted therapeutic applications. Central to their effectiveness in these applications is their resonant and non-linear oscillation behaviour. This article reviews the salient physics of an oscillating microbubble in an ultrasound field, with particular emphasis on phospholipid-coated agents. Both the theoretical underpinnings of bubble vibration and the experimental evidence of non-linear encapsulated bubble dynamics and scattering are discussed and placed within the context of current and emerging applications.
Collapse
Affiliation(s)
- Brandon Helfield
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
46
|
Cohen G, Burks SR, Frank JA. Chlorotoxin-A Multimodal Imaging Platform for Targeting Glioma Tumors. Toxins (Basel) 2018; 10:E496. [PMID: 30486274 PMCID: PMC6316809 DOI: 10.3390/toxins10120496] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022] Open
Abstract
Chlorotoxin (CTX) is a 36-amino-acid disulfide-containing peptide derived from the venom of the scorpion Leiurus quinquestriatus. CTX alters physiology in numerous ways. It interacts with voltage gated chloride channels, Annexin-2, and matrix metalloproteinase-2 (MMP-2). CTX-based bioconjugates have been widely subjected to phase I/II clinical trials and have shown substantial promise. Many studies have demonstrated that CTX preferentially binds to neuroectodermal tumors, such as glioblastoma, without cross-reactivity to normal brain cells. With its ability to penetrate the blood-brain-barrier (BBB) and its tyrosine residue allows covalent conjugation with functional moieties, CTX is an attractive platform to explore development of diagnostic and therapeutic agents for gliomas. In this review, we outline CTX structure and its molecular targets, summarize molecular variations of CTX developed for glioma imaging, and discuss future trends and perspectives for CTX conjugates as a theranostic agent.
Collapse
Affiliation(s)
- Gadi Cohen
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Opening the Blood-Brain Barrier and Improving the Efficacy of Temozolomide Treatments of Glioblastoma Using Pulsed, Focused Ultrasound with a Microbubble Contrast Agent. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6501508. [PMID: 30534564 PMCID: PMC6252217 DOI: 10.1155/2018/6501508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 11/17/2022]
Abstract
Objective To explore the effects of pulsed, focused, and microbubble contrast agent-enhanced ultrasonography (mCEUS) on blood-brain barrier (BBB) permeability and the efficacy temozolomide for glioblastoma. Methods Wistar rats (n = 30) were divided into three groups (n = 10 per group) to determine optimal CUES conditions for achieving BBB permeability, as assessed by ultrastructure transmission electron microscopy (TEM) and western blot assays for the tight junction protein claudin-5. Optimized mCEUS effects on BBB permeability were subsequently confirmed with Evans blue staining (2 groups of 10 rats). The glioma cell line 9L was injected into the brain striatum of Wistar rats. After temozolomide chemotherapy, we detected glial fibrillary acidic protein (GFAP) levels in serum by enzyme-linked immunosorbent assay (ELISA) and in brain tissue by western blot, immunocytochemistry, and real-time quantitative polymerase chain reaction (qPCR). Results BBB permeability was maximized with 1 ml/kg contrast agent mCEUS delivered via 10-min intermittent launches with a 400-ms interval. Evans blue staining confirmed BBB permeability following ultrasonic cavitation in the control group (P < 0.05). Following temozolomide chemotherapy, levels of the tumor marker GFAP were increased in the group with ultrasonic cavitation compared with the control group (P < 0.05). Conclusions When rats were treated by mCEUS with intermittent launches (interval, 400 ms) and injected with 1 mg/kg contrast agent, BBB permeability was increased and temozolomide BBB penetration was enhanced, therapeutic enhancement for glioblastoma.
Collapse
|
48
|
Santos MA, Wu SK, Li Z, Goertz DE, Hynynen K. Microbubble-assisted MRI-guided focused ultrasound for hyperthermia at reduced power levels. Int J Hyperthermia 2018; 35:599-611. [PMID: 30295119 DOI: 10.1080/02656736.2018.1514468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Ultrasound contrast agent microbubbles were combined with magnetic resonance imaging (MRI)-guided focused ultrasound (MRgFUS) as a means to achieve mild hyperthermia at reduced power levels. METHODS MRgFUS hyperthermia (42°C for 20 min) was evaluated in rabbit thigh muscle or Vx2 tumors using infusions of microbubbles (Definity, 20 µL/kg) or saline (sham) administered over 5 min. The impact of treatments on drug uptake was assessed with liposomal doxorubicin (Caelyx, 2.5 mg/kg). Applied power levels before and after the injection of microbubbles or saline were compared, and drug uptake was evaluated with fluorometry of tissues harvested 24 hr post-treatment. RESULTS MRgFUS hyperthermia in muscle and tumors resulted in accurate temperature control (mean =42.0°C, root mean square error (RMSE) = 0.3°C). The power dropped significantly following the injection of microbubbles in muscle and tumors compared to exposures without microbubbles (-21.9% ± 12.5% vs -5.9% ± 7.8%, p = .009 in muscle; -33.8% ± 9.9% vs -3.0% ± 7.2%, p < .001 in tumors). Cavitation monitoring indicated emission of subharmonic, ultraharmonic, and elevated levels of fourth to sixth harmonic frequencies following microbubble injection. The drug delivery was elevated significantly in muscle with the use of microbubble-assisted relative to conventional heating (0.5 ± 0.5 ng/mg vs 0.20 ± 0.04 ng/mg, p = .05), whereas in tumors similar levels were found (11 ± 3 ng/mg vs 16 ± 4 ng/mg, p = .13). CONCLUSIONS The finding that microbubbles reduce the applied power requirements for hyperthermia has considerable clinical implications. The elevated levels of drug found in muscle but not tumor tissue suggest a complex interplay between the heating effects of microbubbles with those of enhanced permeabilization and possible vascular damage.
Collapse
Affiliation(s)
- Marc A Santos
- a Physical Sciences Platform , Sunnybrook Research Institute , Toronto , Canada.,b Department of Medical Biophysics , University of Toronto , Toronto , Canada
| | - Sheng-Kai Wu
- a Physical Sciences Platform , Sunnybrook Research Institute , Toronto , Canada.,b Department of Medical Biophysics , University of Toronto , Toronto , Canada
| | - Zhe Li
- a Physical Sciences Platform , Sunnybrook Research Institute , Toronto , Canada
| | - David E Goertz
- a Physical Sciences Platform , Sunnybrook Research Institute , Toronto , Canada.,b Department of Medical Biophysics , University of Toronto , Toronto , Canada
| | - Kullervo Hynynen
- a Physical Sciences Platform , Sunnybrook Research Institute , Toronto , Canada.,b Department of Medical Biophysics , University of Toronto , Toronto , Canada.,c Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto , Canada
| |
Collapse
|
49
|
Kovacs ZI, Tu TW, Sundby M, Qureshi F, Lewis BK, Jikaria N, Burks SR, Frank JA. MRI and histological evaluation of pulsed focused ultrasound and microbubbles treatment effects in the brain. Theranostics 2018; 8:4837-4855. [PMID: 30279741 PMCID: PMC6160777 DOI: 10.7150/thno.24512] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
Magnetic resonance imaging (MRI)-guided pulsed focused ultrasound (pFUS) combined with microbubbles (MB) contrast agent infusion has been shown to transiently disrupt the blood-brain barrier (BBBD), increasing the delivery of neurotherapeutics to treat central nervous system (CNS) diseases. pFUS interaction with the intravascular MB results in acoustic cavitation forces passing through the neurovascular unit (NVU), inducing BBBD detected on contrast-enhanced MRI. Multiple pFUS+MB exposures in Alzheimer's disease (AD) models are being investigated as a method to clear amyloid plaques by activated microglia or infiltrating immune cells. Since it has been reported that pFUS+MB can induce a sterile inflammatory response (SIR) [1-5] in the rat, the goal of this study was to investigate the potential long-term effects of SIR in the brain following single and six weekly sonications by serial high-resolution MRI and pathology. Methods: Female Sprague Dawley rats weighing 217±16.6 g prior to sonication received bromo-deoxyuridine (BrdU) to tag proliferating cells in the brain. pFUS was performed at 548 kHz, ultrasound burst 10 ms and initial peak negative pressure of 0.3 MPa (in water) for 120 s coupled with a slow infusion of ~460 µL/kg (5-8×107 MB) that started 30 s before and 30 s during sonication. Nine 2 mm focal regions in the left cortex and four regions over the right hippocampus were treated with pFUS+MB. Serial high-resolution brain MRIs at 3 T and 9.4 T were obtained following a single or during the course of six weekly pFUS+MB resulting in BBBD in the left cortex and the right hippocampus. Animals were monitored over 7 to 13 weeks and imaging results were compared to histology. Results: Fewer than half of the rats receiving a single pFUS+MB exposure displayed hypointense voxels on T2*-weighted (w) MRI at week 7 or 13 in the cortex or hippocampus without differences compared to the contralateral side on histograms of T2* maps. Single sonicated rats had evidence of limited microglia activation on pathology compared to the contralateral hemisphere. Six weekly pFUS+MB treatments resulted in pathological changes on T2*w images with multiple hypointense regions, cortical atrophy, along with 50% of rats having persistent BBBD and astrogliosis by MRI. Pathologic analysis of the multiple sonicated animals demonstrated the presence of metallophagocytic Prussian blue-positive cells in the parenchyma with significantly (p<0.05) increased areas of activated astrocytes and microglia, and high numbers of systemic infiltrating CD68+ macrophages along with BrdU+ cells compared to contralateral brain. In addition, multiple treatments caused an increase in the number of hyperphosphorylated Tau (pTau)-positive neurons containing neurofibrillary tangles (NFT) in the sonicated cortex but not in the hippocampus when compared to contralateral brain, which was confirmed by Western blot (WB) (p<0.04). Conclusions: The repeated SIR following multiple pFUS+MB treatments could contribute to changes on MR imaging including persistent BBBD, cortical atrophy, and hypointense voxels on T2w and T2*w images consistent with pathological injury. Moreover, areas of astrogliosis, activated microglia, along with higher numbers of CD68+ infiltrating macrophages and BrdU+ cells were detected in multiple sonicated areas of the cortex and hippocampus. Elevations in pTau and NFT were detected in neurons of the multiple sonicated cortex. Minimal changes on MRI and histology were observed in single pFUS+MB-treated rats at 7 and 13 weeks post sonication. In comparison, animals that received 6 weekly sonications demonstrated evidence on MRI and histology of vascular damage, inflammation and neurodegeneration associated with the NVU commonly observed in trauma. Further investigation is recommended of the long-term effects of multiple pFUS+MB in clinical trials.
Collapse
|
50
|
Mauri G, Nicosia L, Xu Z, Di Pietro S, Monfardini L, Bonomo G, Varano GM, Prada F, Della Vigna P, Orsi F. Focused ultrasound: tumour ablation and its potential to enhance immunological therapy to cancer. Br J Radiol 2018; 91:20170641. [PMID: 29168922 PMCID: PMC5965486 DOI: 10.1259/bjr.20170641] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/16/2017] [Accepted: 11/16/2017] [Indexed: 12/27/2022] Open
Abstract
Various kinds of image-guided techniques have been successfully applied in the last years for the treatment of tumours, as alternative to surgical resection. High intensity focused ultrasound (HIFU) is a novel, totally non-invasive, image-guided technique that allows for achieving tissue destruction with the application of focused ultrasound at high intensity. This technique has been successfully applied for the treatment of a large variety of diseases, including oncological and non-oncological diseases. One of the most fascinating aspects of image-guided ablations, and particularly of HIFU, is the reported possibility of determining a sort of stimulation of the immune system, with an unexpected "systemic" response to treatments designed to be "local". In the present article the mechanisms of action of HIFU are described, and the main clinical applications of this technique are reported, with a particular focus on the immune-stimulation process that might originate from tumour ablations.
Collapse
Affiliation(s)
- Giovanni Mauri
- Deparmtent of interventional radiology, European istitute of oncology, Milan, Italy
| | - Luca Nicosia
- Postgraduate School of Radiology, Università degli Studi di Milano, Milan, Italy
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Salvatore Di Pietro
- Postgraduate School of Radiology, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Monfardini
- Department of Radiology and diagnotic imaging, Poliambulazna di Brescia, Brescia, Italy
| | - Guido Bonomo
- Deparmtent of interventional radiology, European istitute of oncology, Milan, Italy
| | | | | | - Paolo Della Vigna
- Deparmtent of interventional radiology, European istitute of oncology, Milan, Italy
| | - Franco Orsi
- Deparmtent of interventional radiology, European istitute of oncology, Milan, Italy
| |
Collapse
|