51
|
Rong L, Li N, Zhang Z. Emerging therapies for glioblastoma: current state and future directions. J Exp Clin Cancer Res 2022; 41:142. [PMID: 35428347 PMCID: PMC9013078 DOI: 10.1186/s13046-022-02349-7] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/26/2022] [Indexed: 04/15/2023] Open
Abstract
Glioblastoma (GBM) is the most common high-grade primary malignant brain tumor with an extremely poor prognosis. Given the poor survival with currently approved treatments for GBM, new therapeutic strategies are urgently needed. Advances in decades of investment in basic science of glioblastoma are rapidly translated into innovative clinical trials, utilizing improved genetic and epigenetic profiling of glioblastoma as well as the brain microenvironment and immune system interactions. Following these encouraging findings, immunotherapy including immune checkpoint blockade, chimeric antigen receptor T (CAR T) cell therapy, oncolytic virotherapy, and vaccine therapy have offered new hope for improving GBM outcomes; ongoing studies are using combinatorial therapies with the aim of minimizing adverse side-effects and augmenting antitumor immune responses. In addition, techniques to overcome the blood-brain barrier (BBB) for targeted delivery are being tested in clinical trials in patients with recurrent GBM. Here, we set forth the rationales for these promising therapies in treating GBM, review the potential novel agents, the current status of preclinical and clinical trials, and discuss the challenges and future perspectives in glioblastoma immuno-oncology.
Collapse
Affiliation(s)
- Liang Rong
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ni Li
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Zhang
- Key Laboratory of Brain, Cognition and Education Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China.
| |
Collapse
|
52
|
Chien CY, Yang Y, Gong Y, Yue Y, Chen H. Blood-Brain Barrier Opening by Individualized Closed-Loop Feedback Control of Focused Ultrasound. BME FRONTIERS 2022; 2022:9867230. [PMID: 37850162 PMCID: PMC10521637 DOI: 10.34133/2022/9867230] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/01/2022] [Indexed: 10/19/2023] Open
Abstract
Objective and Impact Statement. To develop an approach for individualized closed-loop feedback control of microbubble cavitation to achieve safe and effective focused ultrasound in combination with microbubble-induced blood-brain barrier opening (FUS-BBBO). Introduction. FUS-BBBO is a promising strategy for noninvasive and localized brain drug delivery with a growing number of clinical studies currently ongoing. Real-time cavitation monitoring and feedback control are critical to achieving safe and effective FUS-BBBO. However, feedback control algorithms used in the past were either open-loop or without consideration of baseline cavitation level difference among subjects. Methods. This study performed feedback-controlled FUS-BBBO by defining the target cavitation level based on the baseline stable cavitation level of an individual subject with "dummy" FUS sonication. The dummy FUS sonication applied FUS with a low acoustic pressure for a short duration in the presence of microbubbles to define the baseline stable cavitation level that took into consideration of individual differences in the detected cavitation emissions. FUS-BBBO was then achieved through two sonication phases: ramping-up phase to reach the target cavitation level and maintaining phase to control the stable cavitation level at the target cavitation level. Results. Evaluations performed in wild-type mice demonstrated that this approach achieved effective and safe trans-BBB delivery of a model drug. The drug delivery efficiency increased as the target cavitation level increased from 0.5 dB to 2 dB without causing vascular damage. Increasing the target cavitation level to 3 dB and 4 dB increased the probability of tissue damage. Conclusions. Safe and effective brain drug delivery was achieved using the individualized closed-loop feedback-controlled FUS-BBBO.
Collapse
Affiliation(s)
- Chih-Yen Chien
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Yan Gong
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, Missouri 63108, USA
| |
Collapse
|
53
|
Translation of focused ultrasound for blood-brain barrier opening in glioma. J Control Release 2022; 345:443-463. [PMID: 35337938 DOI: 10.1016/j.jconrel.2022.03.035] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022]
Abstract
Survival outcomes for patients with glioblastoma multiforme (GBM) have remained poor for the past 15 years, reflecting a clear challenge in the development of more effective treatment strategies. The efficacy of systemic therapies for GBM is greatly limited by the presence of the blood-brain barrier (BBB), which prevents drug penetration and accumulation in regions of infiltrative tumour, as represented in a consistent portion of GBM lesions. Focused ultrasound (FUS) - a technique that uses low-frequency ultrasound waves to induce targeted temporary disruption of the BBB - promises to improve survival outcomes by enhancing drug delivery and accumulation to infiltrating tumour regions. In this review we discuss the current state of preclinical investigations using FUS to enhance delivery of systemic therapies to intracranial neoplasms. We highlight critical methodological inconsistencies that are hampering clinical translation of FUS and we provide guiding principles for future preclinical studies. Particularly, we focus our attention on the importance of the selection of clinically relevant animal models and to the standardization of methods for FUS delivery, which will be paramount to the successful clinical translation of this promising technology for treatment in GBM patients. We also discuss how preclinical FUS research can benefit the development of GBM immunotherapies.
Collapse
|
54
|
Lechpammer M, Rao R, Shah S, Mirheydari M, Bhattacharya D, Koehler A, Toukam DK, Haworth KJ, Pomeranz Krummel D, Sengupta S. Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers. Cancers (Basel) 2022; 14:1627. [PMID: 35406398 PMCID: PMC8997081 DOI: 10.3390/cancers14071627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, or glioblastoma multiforme (GBM, WHO Grade IV), is a highly aggressive adult glioma. Despite extensive efforts to improve treatment, the current standard-of-care (SOC) regimen, which consists of maximal resection, radiotherapy, and temozolomide (TMZ), achieves only a 12-15 month survival. The clinical improvements achieved through immunotherapy in several extracranial solid tumors, including non-small-cell lung cancer, melanoma, and non-Hodgkin lymphoma, inspired investigations to pursue various immunotherapeutic interventions in adult glioblastoma patients. Despite some encouraging reports from preclinical and early-stage clinical trials, none of the tested agents have been convincing in Phase III clinical trials. One, but not the only, factor that is accountable for the slow progress is the blood-brain barrier, which prevents most antitumor drugs from reaching the target in appreciable amounts. Herein, we review the current state of immunotherapy in glioblastoma and discuss the significant challenges that prevent advancement. We also provide thoughts on steps that may be taken to remediate these challenges, including the application of ultrasound technologies.
Collapse
Affiliation(s)
- Mirna Lechpammer
- Foundation Medicine, Inc., Cambridge, MA 02141, USA;
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rohan Rao
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Sanjit Shah
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Mona Mirheydari
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Abigail Koehler
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Donatien Kamdem Toukam
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Kevin J. Haworth
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| |
Collapse
|
55
|
Athanassiadis AG, Ma Z, Moreno-Gomez N, Melde K, Choi E, Goyal R, Fischer P. Ultrasound-Responsive Systems as Components for Smart Materials. Chem Rev 2022; 122:5165-5208. [PMID: 34767350 PMCID: PMC8915171 DOI: 10.1021/acs.chemrev.1c00622] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Indexed: 02/06/2023]
Abstract
Smart materials can respond to stimuli and adapt their responses based on external cues from their environments. Such behavior requires a way to transport energy efficiently and then convert it for use in applications such as actuation, sensing, or signaling. Ultrasound can carry energy safely and with low losses through complex and opaque media. It can be localized to small regions of space and couple to systems over a wide range of time scales. However, the same characteristics that allow ultrasound to propagate efficiently through materials make it difficult to convert acoustic energy into other useful forms. Recent work across diverse fields has begun to address this challenge, demonstrating ultrasonic effects that provide control over physical and chemical systems with surprisingly high specificity. Here, we review recent progress in ultrasound-matter interactions, focusing on effects that can be incorporated as components in smart materials. These techniques build on fundamental phenomena such as cavitation, microstreaming, scattering, and acoustic radiation forces to enable capabilities such as actuation, sensing, payload delivery, and the initiation of chemical or biological processes. The diversity of emerging techniques holds great promise for a wide range of smart capabilities supported by ultrasound and poses interesting questions for further investigations.
Collapse
Affiliation(s)
- Athanasios G. Athanassiadis
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Zhichao Ma
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Nicolas Moreno-Gomez
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
- Institute
of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Kai Melde
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Eunjin Choi
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
- Institute
of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Rahul Goyal
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Peer Fischer
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
- Institute
of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| |
Collapse
|
56
|
Schoen S, Dash P, Arvanitis CD. Experimental Demonstration of Trans-Skull Volumetric Passive Acoustic Mapping With the Heterogeneous Angular Spectrum Approach. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:534-542. [PMID: 34748486 PMCID: PMC10243207 DOI: 10.1109/tuffc.2021.3125670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Real-time, 3-D, passive acoustic mapping (PAM) of microbubble dynamics during transcranial focused ultrasound (FUS) is essential for optimal treatment outcomes. The angular spectrum approach (ASA) potentially offers a very efficient method to perform PAM, as it can reconstruct specific frequency bands pertinent to microbubble dynamics and may be extended to correct aberrations caused by the skull. Here, we experimentally assess the abilities of heterogeneous ASA (HASA) to perform trans-skull PAM. Our experimental investigations demonstrate that the 3-D PAMs of a known 1-MHz source, constructed with HASA through an ex vivo human skull segment, reduced both the localization error (from 4.7 ± 2.3 to 2.3 ± 1.6 mm) and the number, size, and energy of spurious lobes caused by aberration, with the modest additional computational expense. While further improvements in the localization errors are expected with arrays with denser elements and larger aperture, our analysis revealed that experimental constraints associated with the array pitch and aperture (here, 1.8 mm and 2.5 cm, respectively) can be ameliorated by interpolation and peak finding techniques. Beyond the array characteristics, our analysis also indicated that errors in the registration (translation and rotation of ±5 mm and ±5°, respectively) of the skull segment to the array can lead to peak localization errors of the order of a few wavelengths. Interestingly, errors in the spatially dependent speed of sound in the skull (±20%) caused only subwavelength errors in the reconstructions, suggesting that registration is the most important determinant of point source localization accuracy. Collectively, our findings show that HASA can address source localization problems through the skull efficiently and accurately under realistic conditions, thereby creating unique opportunities for imaging and controlling the microbubble dynamics in the brain.
Collapse
|
57
|
Tu J, Yu ACH. Ultrasound-Mediated Drug Delivery: Sonoporation Mechanisms, Biophysics, and Critical Factors. BME FRONTIERS 2022; 2022:9807347. [PMID: 37850169 PMCID: PMC10521752 DOI: 10.34133/2022/9807347] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/31/2021] [Indexed: 10/19/2023] Open
Abstract
Sonoporation, or the use of ultrasound in the presence of cavitation nuclei to induce plasma membrane perforation, is well considered as an emerging physical approach to facilitate the delivery of drugs and genes to living cells. Nevertheless, this emerging drug delivery paradigm has not yet reached widespread clinical use, because the efficiency of sonoporation is often deemed to be mediocre due to the lack of detailed understanding of the pertinent scientific mechanisms. Here, we summarize the current observational evidence available on the notion of sonoporation, and we discuss the prevailing understanding of the physical and biological processes related to sonoporation. To facilitate systematic understanding, we also present how the extent of sonoporation is dependent on a multitude of factors related to acoustic excitation parameters (ultrasound frequency, pressure, cavitation dose, exposure time), microbubble parameters (size, concentration, bubble-to-cell distance, shell composition), and cellular properties (cell type, cell cycle, biochemical contents). By adopting a science-backed approach to the realization of sonoporation, ultrasound-mediated drug delivery can be more controllably achieved to viably enhance drug uptake into living cells with high sonoporation efficiency. This drug delivery approach, when coupled with concurrent advances in ultrasound imaging, has potential to become an effective therapeutic paradigm.
Collapse
Affiliation(s)
- Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, China
| | - Alfred C. H. Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
58
|
Thombre R, Mess G, Kempski Leadingham KM, Kapoor S, Hersh A, Acord M, Kaovasia T, Theodore N, Tyler B, Manbachi A. Towards standardization of the parameters for opening the blood-brain barrier with focused ultrasound to treat glioblastoma multiforme: A systematic review of the devices, animal models, and therapeutic compounds used in rodent tumor models. Front Oncol 2022; 12:1072780. [PMID: 36873300 PMCID: PMC9978816 DOI: 10.3389/fonc.2022.1072780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/20/2022] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a deadly and aggressive malignant brain cancer that is highly resistant to treatments. A particular challenge of treatment is caused by the blood-brain barrier (BBB), the relatively impermeable vasculature of the brain. The BBB prevents large molecules from entering the brain parenchyma. This protective characteristic of the BBB, however, also limits the delivery of therapeutic drugs for the treatment of brain tumors. To address this limitation, focused ultrasound (FUS) has been safely utilized to create transient openings in the BBB, allowing various high molecular weight drugs access to the brain. We performed a systematic review summarizing current research on treatment of GBMs using FUS-mediated BBB openings in in vivo mouse and rat models. The studies gathered here highlight how the treatment paradigm can allow for increased brain and tumor perfusion of drugs including chemotherapeutics, immunotherapeutics, gene therapeutics, nanoparticles, and more. Given the promising results detailed here, the aim of this review is to detail the commonly used parameters for FUS to open the BBB in rodent GBM models.
Collapse
Affiliation(s)
- Rasika Thombre
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Griffin Mess
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Kelley M Kempski Leadingham
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Shivani Kapoor
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Andrew Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Molly Acord
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Tarana Kaovasia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amir Manbachi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,HEPIUS Innovation Laboratory, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Electrical Engineering and Computer Science, Johns Hopkins University, Baltimore, MD, United States.,Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, United States.,Department of Anesthesiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
59
|
Abstract
The blood-brain barrier (BBB) is the major barrier for brain drug delivery and limits the treatment options for central nervous system diseases. To circumvent the BBB, we introduced the focused ultrasound-mediated intranasal brain drug delivery (FUSIN) technique. FUSIN utilizes the nasal route for direct nose-to-brain drug administration, bypassing the BBB and minimizing systemic exposure. It also uses the transcranial application of ultrasound energy focused at a targeted brain region to induce microbubble cavitation, which enhances the transport of intranasally administered agents at the FUS-targeted brain location. FUSIN is unique in that it can achieve noninvasive and localized brain drug delivery with minimized systemic toxicity. The goal of this chapter is to provide a detailed protocol for FUSIN delivery to the mouse brain.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA.
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
60
|
Tan C, Yan B, Han T, Yu ACH, Qin P. Improving temporal stability of stable cavitation activity of circulating microbubbles using a closed-loop controller based on pulse-length regulation. ULTRASONICS SONOCHEMISTRY 2022; 82:105882. [PMID: 34969003 PMCID: PMC8855699 DOI: 10.1016/j.ultsonch.2021.105882] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/30/2021] [Accepted: 12/18/2021] [Indexed: 05/16/2023]
Abstract
Stable cavitation (SC) has shown great potential for novel therapeutic applications. The spatiotemporal distribution of the SC activity of microbubbles circulating in a target region is not only correlated with the uniformity of treatment, but also with some undesirable effects. Therefore, it is important to achieve controllable and desirable SC activity in target regions for improved therapeutic efficiency and biosafety. This study proposes a closed-loop feedback controller based on pulse length (PL) regulation to improve the temporal stability of SC activity. Microbubbles circulating in a physiological flowing phantom were exposed to a 1 MHz focused transducer. The SC signals produced were initially received by another 7.5 MHz plane transducer, followed by high-speed signal acquisition and real-time processing. Based on the real-time-measured SC intensity excited by the current acoustic pulse, the proposed closed-loop feedback controller used three proportional coefficients to regulate the peak negative pressure (PNP) and PL of the next acoustic pulse during the acceleration and stable stages, respectively. The results show that the rise time and the temporal stability of the SC intensity of the microbubbles circulating in these two stages were improved significantly by the optimized proportional coefficients used in the proposed controller. Importantly, when compared with the traditional closed-loop feedback controller based on PNP regulation, the proposed closed-loop feedback controller based on PL regulation reduced the probability of a transition between stable and inertial cavitation, thus avoiding the risk of disadvantageous bioeffects in practical applications. These results demonstrate the effectiveness of the proposed PL-based closed-loop feedback controller and provide a feasible strategy for realization of controllable cavitation activity in applications.
Collapse
Affiliation(s)
- Chunjie Tan
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bo Yan
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tao Han
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON N2L3G1, Canada
| | - Peng Qin
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
61
|
Ndemazie NB, Inkoom A, Morfaw EF, Smith T, Aghimien M, Ebesoh D, Agyare E. Multi-disciplinary Approach for Drug and Gene Delivery Systems to the Brain. AAPS PharmSciTech 2021; 23:11. [PMID: 34862567 PMCID: PMC8817187 DOI: 10.1208/s12249-021-02144-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Drug delivery into the brain has for long been a huge challenge as the blood–brain barrier (BBB) offers great resistance to entry of foreign substances (with drugs inclusive) into the brain. This barrier in healthy individuals is protective to the brain, disallowing noxious substances present in the blood to get to the brain while allowing for the exchange of small molecules into the brain by diffusion. However, BBB is disrupted under certain disease conditions, such as cerebrovascular diseases including acute ischemic stroke and intracerebral hemorrhage, and neurodegenerative disorders including multiple sclerosis (MS), Alzheimer’s disease (AD), Parkinson’s disease (PD), and cancers. This review aims to provide a broad overview of present-day strategies for brain drug delivery, emphasizing novel delivery systems. Hopefully, this review would inspire scientists and researchers in the field of drug delivery across BBB to uncover new techniques and strategies to optimize drug delivery to the brain. Considering the anatomy, physiology, and pathophysiological functioning of the BBB in health and disease conditions, this review is focused on the controversies drawn from conclusions of recently published studies on issues such as the penetrability of nanoparticles into the brain, and whether active targeted drug delivery into the brain could be achieved with the use of nanoparticles. We also extended the review to cover novel non-nanoparticle strategies such as using viral and peptide vectors and other non-invasive techniques to enhance brain uptake of drugs.
Collapse
|
62
|
Maesawa S, Nakatsubo D, Tsugawa T, Kato S, Shibata M, Takai S, Torii J, Ishizaki T, Wakabayashi T, Saito R. Techniques, Indications, and Outcomes in Magnetic Resonance-guided Focused Ultrasound Thalamotomy for Tremor. Neurol Med Chir (Tokyo) 2021; 61:629-639. [PMID: 34470990 PMCID: PMC8592814 DOI: 10.2176/nmc.ra.2021-0187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Magnetic resonance (MR)-guided focused ultrasound surgery (MRgFUS) is the latest minimally invasive stereotactic procedure, and thalamotomy using this novel modality has demonstrated its effectiveness and safety, especially for patients with essential tremor (ET) and Parkinson's disease (PD). In Japan, the application of MRgFUS to treat ET and PD has recently been covered by health insurance. Technically, the transducer with 1024 elements emits ultrasound beams, which are then focused on the target with a phase control, resulting in optimal ablation by thermal coagulation. The technical advantages of MRgFUS are continuous intraoperative monitoring of clinical symptoms and MR images and fine adjustment of the target by the steering function. Postoperative tremor control is compatible with other modalities, although long-term follow-up is necessary. The adverse effects are usually transient and acceptable. Prognostic factors for good tremor control include high temperature and large lesion size. A high skull density ratio is a factor to achieve high temperature and large lesioning, but it may not be necessary and sufficient for clinical outcomes. For patients with advanced symptoms such as bilateral tremor or head/neck tremor, deep brain stimulation may be recommended because of the adjustability of stimulation and the possibility of bilateral treatment. Patients have high expectations of MRgFUS because of its non-invasiveness. To perform this treatment safely and effectively, physicians need to understand the technological aspects, the physiological principles. To choose the appropriate modality, physicians also should recognize the clinical advantages and disadvantages of MRgFUS compared to other modalities.
Collapse
Affiliation(s)
- Satoshi Maesawa
- Brain and Mind Research Center, Nagoya University
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
| | - Daisuke Nakatsubo
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
- Radiosurgery and Focused Ultrasound Surgery Center, Nagoya Kyoritsu Hospital
| | - Takahiko Tsugawa
- Radiosurgery and Focused Ultrasound Surgery Center, Nagoya Kyoritsu Hospital
| | - Sachiko Kato
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
- Radiosurgery and Focused Ultrasound Surgery Center, Nagoya Kyoritsu Hospital
| | - Masashi Shibata
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
- Radiosurgery and Focused Ultrasound Surgery Center, Nagoya Kyoritsu Hospital
| | - Sou Takai
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
| | - Jun Torii
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
| | - Tomotaka Ishizaki
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
- Department of Neurosurgery, Kainan Hospital
| | - Toshihiko Wakabayashi
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
- Radiosurgery and Focused Ultrasound Surgery Center, Nagoya Kyoritsu Hospital
| | - Ryuta Saito
- Department of Neurosurgery, Nagoya University Graduate School of Medicine
| |
Collapse
|
63
|
Hu Z, Xu L, Chien CY, Yang Y, Gong Y, Ye D, Pacia CP, Chen H. 3-D Transcranial Microbubble Cavitation Localization by Four Sensors. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:3336-3346. [PMID: 34166187 PMCID: PMC8808337 DOI: 10.1109/tuffc.2021.3091950] [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] [Indexed: 06/08/2023]
Abstract
Cavitation is the fundamental physical mechanism of various focused ultrasound (FUS)-mediated therapies in the brain. Accurately knowing the three-dimensional (3-D) location of cavitation in real-time can improve the targeting accuracy and avoid off-target tissue damage. Existing techniques for 3-D passive transcranial cavitation detection require the use of expensive and complicated hemispherical phased arrays with 128 or 256 elements. The objective of this study was to investigate the feasibility of using four sensors for transcranial 3-D localization of cavitation. Differential microbubble cavitation detection combined with the time difference of arrival algorithm was developed for the localization using the four sensors. Numerical simulation using k-Wave toolbox was performed to validate the proposed method for transcranial cavitation source localization. The sensors with a center frequency of 2.25 MHz and a 6 dB bandwidth of 1.39 MHz were used to locate cavitation generated by FUS (500 kHz) sonication of microbubbles that were injected into a tube positioned inside an ex vivo human skullcap. Cavitation emissions from the microbubbles were detected transcranially using the four sensors. Both simulation and experimental studies found that the proposed method achieved accurate 3-D cavitation localization. When the cavitation source was located within 30 mm from the geometric center of the sensor network, the accuracy of the localization method with the skull was measured to be 1.9±1.0 mm, which was not significantly different from that without the skull (1.7 ± 0.5 mm). The accuracy decreased as the cavitation source was away from the geometric center of the sensor network. It also decreased as the pulse length increased. Its accuracy was not significantly affected by the sensor position relative to the skull. In summary, four sensors combined with the proposed localization algorithm offer a simple approach for 3-D transcranial cavitation localization.
Collapse
|
64
|
Roy J, Pandey V, Gupta I, Shekhar H. Antibacterial Sonodynamic Therapy: Current Status and Future Perspectives. ACS Biomater Sci Eng 2021; 7:5326-5338. [PMID: 34714638 DOI: 10.1021/acsbiomaterials.1c00587] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Multidrug-resistant bacteria have emerged in both community and hospital settings, partly due to the misuse of antibiotics. The inventory of viable antibiotics is rapidly declining, and efforts toward discovering newer antibiotics are not yielding the desired outcomes. Therefore, alternate antibacterial therapies based on physical mechanisms such as light and ultrasound are being explored. Sonodynamic therapy (SDT) is an emerging therapeutic approach that involves exposing target tissues to a nontoxic sensitizing chemical and low-intensity ultrasound. SDT can enable site-specific cytotoxicity by producing reactive oxygen species (ROS) in response to ultrasound, which can be harnessed for treating bacterial infections. This approach can potentially be used for both superficial and deep-seated microbial infections. The majority of the sonosensitizers reported are nonpolar, exhibiting limited bioavailability and a high clearance rate in the body. Therefore, targeted delivery agents such as nanoparticle composites, liposomes, and microbubbles are being investigated. This article reviews recent developments in antibacterial sonodynamic therapy, emphasizing biophysical and chemical mechanisms, novel delivery agents, ultrasound exposure and image guidance strategies, and the challenges in the pathway to clinical translation.
Collapse
Affiliation(s)
- Jayishnu Roy
- Discipline of Biological Engineering, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Vijayalakshmi Pandey
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Iti Gupta
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Himanshu Shekhar
- Discipline of Electrical Engineering, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
65
|
Lea-Banks H, Hynynen K. Sub-millimetre precision of drug delivery in the brain from ultrasound-triggered nanodroplets. J Control Release 2021; 338:731-741. [PMID: 34530050 DOI: 10.1016/j.jconrel.2021.09.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/17/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022]
Abstract
Drug-loaded nanoscale cavitation agents, called nanodroplets, are an attractive solution to enhance and localize drug delivery, offering increased stability and prolonged half-life in circulation compared to microbubbles. However, the spatial precision with which drug can be released and delivered into brain tissue from such agents has not been directly mapped. Decafluorobutane lipid-shell droplets (206 +/- 6 nm) were loaded with a fluorescent blood-brain barrier (BBB)-penetrating dye (Nile Blue) and vaporized with ultrasound (1.66 MHz, 10 ms pulse length, 1 Hz pulse repetition frequency), generating transient echogenic microbubbles and delivering the encapsulated dye. The distribution and intensity of released fluorophore was mapped in a tissue-mimicking phantom, and in the brain of rats (Sprague Dawley, N = 4, n = 16). The release and distribution of dye was found to be pressure-dependent (0.2-3.5 MPa) and to occur only above the vaporization threshold of the nanodroplets (1.5 +/- 0.25 MPa in vitro, 2.4 +/- 0.05 MPa in vivo). Dye delivery was achieved with sub-millimetre spatial precision, covering an area of 0.4 to 1.5 mm in diameter, determined by the sonication pressure. The distribution and intensity of dye released at depth in the brain followed the axial pressure profile of the ultrasound beam. Nile Blue (354 Da, LogP 2.7) was compared to Nile Red (318 Da, LogP 3.8) and Quantum Dots (CdSe/ZnS, 5 nm diameter) to visualize the role of molecule size and lipophilicity in crossing the intact BBB following triggered release. Acoustic emissions were shown to predict the successful delivery of the BBB-penetrating dye and the extent of the distribution, demonstrating the theranostic capabilities of nanoscale droplets to precisely localize drug delivery in the brain.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
66
|
Omata D, Munakata L, Maruyama K, Suzuki R. Enhanced Vascular Permeability by Microbubbles and Ultrasound in Drug Delivery. Biol Pharm Bull 2021; 44:1391-1398. [PMID: 34602547 DOI: 10.1248/bpb.b21-00453] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ultrasound and microbubbles, an ultrasound contrast agent, have recently increased attention to developing novel drug delivery systems. Ultrasound exposure can induce mechanical effects derived from microbubbles behaviors such as an expansion, contraction, and collapse depending on ultrasound conditions. These mechanical effects induce several biological effects, including enhancement of vascular permeability. For drug delivery, one promising approach is enhancing vascular permeability using ultrasound and microbubbles, resulting in improved drug transport to targeted tissues. This approach is applied to several tissues and drugs to cure diseases. This review describes the enhancement of vascular permeability by ultrasound and microbubbles and its therapeutic application, including our recent study. We also discuss the current situation of the field and its potential future perspectives.
Collapse
Affiliation(s)
- Daiki Omata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University
| | - Kazuo Maruyama
- Laboratory of Theranostics, Faculty of Pharma-Science, Teikyo University.,Advanced Comprehensive Research Organization (ACRO), Teikyo University
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University.,Advanced Comprehensive Research Organization (ACRO), Teikyo University
| |
Collapse
|
67
|
Byrne J, Huang HW, McRae JC, Babaee S, Soltani A, Becker SL, Traverso G. Devices for drug delivery in the gastrointestinal tract: A review of systems physically interacting with the mucosa for enhanced delivery. Adv Drug Deliv Rev 2021; 177:113926. [PMID: 34403749 DOI: 10.1016/j.addr.2021.113926] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
The delivery of macromolecules via the gastrointestinal (GI) tract remains a significant challenge. A variety of technologies using physical modes of drug delivery have been developed and investigated to overcome the epithelial cell layer of the GI tract for local and systemic delivery. These technologies include direct injection, jetting, ultrasound, and iontophoresis, which have been largely adapted from transdermal drug delivery. Direct injection of agents using needles through endoscopy has been used clinically for over a century. Jetting, a needle-less method of drug delivery where a high-speed stream of fluid medication penetrates tissue, has been evaluated pre-clinically for delivery of agents into the buccal mucosa. Ultrasound has been shown to be beneficial in enhancing delivery of macromolecules, including nucleic acids, in pre-clinical animal models. The application of an electric field gradient to drive drugs into tissues through the technique of iontophoresis has been shown to deliver highly toxic chemotherapies into GI tissues. Here in, we provide an in-depth overview of these physical modes of drug delivery in the GI tract and their clinical and preclinical uses.
Collapse
Affiliation(s)
- James Byrne
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Harvard Radiation Oncology Program, Boston, MA 02114, USA; Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52240, USA
| | - Hen-Wei Huang
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - James C McRae
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sahab Babaee
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Amin Soltani
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sarah L Becker
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
68
|
Gümmer J, Schenke S, Denner F. Modelling Lipid-Coated Microbubbles in Focused Ultrasound Applications at Subresonance Frequencies. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2958-2979. [PMID: 34344560 DOI: 10.1016/j.ultrasmedbio.2021.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 06/15/2021] [Accepted: 06/20/2021] [Indexed: 06/13/2023]
Abstract
We present a computational study of the behaviour of a lipid-coated SonoVue microbubble with initial radius 1 µm ≤ R0 ≤ 2 µm, excited at frequencies (200-1500 kHz) significantly below the linear resonance frequency and pressure amplitudes of up to 1500 kPa-an excitation regime used in many applications of focused ultrasound. The bubble dynamics are simulated using the Rayleigh-Plesset equation and the Gilmore equation, in conjunction with the Marmottant model for the lipid monolayer coating. Also, a new continuously differentiable variant of the Marmottant model is introduced. Below the onset of inertial cavitation, a linear regime is identified in which the maximum pressure at the bubble wall is linearly proportional to the excitation pressure amplitude and the mechanical index. This linear regime is bounded by the Blake pressure, and, in line with recent in vitro experiments, the onset of inertial cavitation is found to occur at an excitation pressure amplitude of approximately 130-190 kPa, depending on the initial bubble size. In the nonlinear regime the maximum pressure at the bubble wall is found to be readily predicted by the maximum bubble radius, and both the Rayleigh-Plesset and Gilmore equations are shown to predict the onset of sub- and ultraharmonic frequencies of the acoustic emissions compared with in vitro experiments. Neither the surface dilational viscosity of the lipid monolayer nor the compressibility of the liquid has a discernible influence on the quantities studied, but accounting for the lipid coating is critical for accurate prediction of the bubble behaviour. The Gilmore equation is shown to be valid for the bubbles and excitation regime considered, and the Rayleigh-Plesset equation also provides accurate qualitative predictions, even though it is outside its range of validity for many of the cases considered.
Collapse
Affiliation(s)
- Jonas Gümmer
- Chair of Mechanical Process Engineering, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Sören Schenke
- Chair of Mechanical Process Engineering, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Fabian Denner
- Chair of Mechanical Process Engineering, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany.
| |
Collapse
|
69
|
Localized blood-brain barrier opening in infiltrating gliomas with MRI-guided acoustic emissions-controlled focused ultrasound. Proc Natl Acad Sci U S A 2021; 118:2103280118. [PMID: 34504017 DOI: 10.1073/pnas.2103280118] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological treatment of gliomas and other brain-infiltrating tumors remains challenging due to limited delivery of most therapeutics across the blood-brain barrier (BBB). Transcranial MRI-guided focused ultrasound (FUS), an emerging technology for noninvasive brain treatments, enables transient opening of the BBB through acoustic activation of circulating microbubbles. Here, we evaluate the safety and utility of transcranial microbubble-enhanced FUS (MB-FUS) for spatially targeted BBB opening in patients with infiltrating gliomas. In this Phase 0 clinical trial (NCT03322813), we conducted comparative and quantitative analyses of FUS exposures (sonications) and their effects on gliomas using MRI, histopathology, microbubble acoustic emissions (harmonic dose [HD]), and fluorescence-guided surgery metrics. Contrast-enhanced MRI and histopathology indicated safe and reproducible BBB opening in all patients. These observations occurred using a power cycling closed feedback loop controller, with the power varying by nearly an order of magnitude on average. This range underscores the need for monitoring and titrating the exposure on a patient-by-patient basis. We found a positive correlation between microbubble acoustic emissions (HD) and MR-evident BBB opening (P = 0.07) and associated interstitial changes (P < 0.01), demonstrating the unique capability to titrate the MB-FUS effects in gliomas. Importantly, we identified a 2.2-fold increase of fluorescein accumulation in MB-FUS-treated compared to untreated nonenhancing tumor tissues (P < 0.01) while accounting for vascular density. Collectively, this study demonstrates the capabilities of MB-FUS for safe, localized, controlled BBB opening and highlights the potential of this technology to improve the surgical and pharmacologic treatment of brain tumors.
Collapse
|
70
|
Ji R, Karakatsani ME, Burgess M, Smith M, Murillo MF, Konofagou EE. Cavitation-modulated inflammatory response following focused ultrasound blood-brain barrier opening. J Control Release 2021; 337:458-471. [PMID: 34324895 PMCID: PMC8440441 DOI: 10.1016/j.jconrel.2021.07.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 07/21/2021] [Accepted: 07/25/2021] [Indexed: 12/28/2022]
Abstract
Focused ultrasound (FUS) in combination with systemically injected microbubbles can be used to non-invasively open the blood-brain barrier (BBB) in targeted regions for a variety of therapeutic applications. Over the past two decades, preclinical research into the safety and efficacy of FUS-induced BBB opening has proven this technique to be transient and efficacious, propelling FUS-induced BBB opening into several clinical trials in recent years. However, as clinical trials further progress, the neuroinflammatory response to FUS-induced BBB opening needs to be better understood. In this study, we provide further insight into the relationship of microbubble cavitation and the resulting innate immune response to FUS-induced BBB opening. By keeping ultrasound parameters fixed (i.e. frequency, pressure, pulse length, etc.), three groups of mice were sonicated using a real-time cavitation controller until a target cavitation dose was reached (1 x 107 V2•s, 5 x 107 V2•s, 1 x 108 V2•s). The change in relative gene expression of the mouse inflammatory cytokines and receptors were evaluated at three different time-points (6 h, 24 h, and 72 h) after FUS. At both 6 and 24 h time-points, significant changes in relative gene expression of inflammatory cytokines and receptors were observed across all cavitation groups. However, the degree of changes in relative expression levels and the number of genes with significant changes in expression varied across the cavitation groups. Groups with a higher cavitation dose exhibited both greater changes in relative expression levels and greater number of significant changes. By 72 h post-opening, the gene expression levels returned to baseline in all cavitation dose groups, signifying a transient inflammatory response to FUS-induced BBB opening at the targeted cavitation dose levels. Furthermore, the real-time cavitation controller was able to produce consistent and significantly different BBB permeability enhancement volumes across the three different cavitation dose groups. These results indicate that cavitation monitoring and controlling during FUS-induced BBB opening can be used to potentially modulate or limit the degree of neuroinflammation, further emphasizing the importance of implementing cavitation controllers as FUS-induced BBB opening is translated into the clinic.
Collapse
Affiliation(s)
- Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Maria E Karakatsani
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mark Burgess
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Morgan Smith
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Maria F Murillo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA.
| |
Collapse
|
71
|
Ho YJ, Huang CC, Fan CH, Liu HL, Yeh CK. Ultrasonic technologies in imaging and drug delivery. Cell Mol Life Sci 2021; 78:6119-6141. [PMID: 34297166 PMCID: PMC11072106 DOI: 10.1007/s00018-021-03904-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Ultrasonic technologies show great promise for diagnostic imaging and drug delivery in theranostic applications. The development of functional and molecular ultrasound imaging is based on the technical breakthrough of high frame-rate ultrasound. The evolution of shear wave elastography, high-frequency ultrasound imaging, ultrasound contrast imaging, and super-resolution blood flow imaging are described in this review. Recently, the therapeutic potential of the interaction of ultrasound with microbubble cavitation or droplet vaporization has become recognized. Microbubbles and phase-change droplets not only provide effective contrast media, but also show great therapeutic potential. Interaction with ultrasound induces unique and distinguishable biophysical features in microbubbles and droplets that promote drug loading and delivery. In particular, this approach demonstrates potential for central nervous system applications. Here, we systemically review the technological developments of theranostic ultrasound including novel ultrasound imaging techniques, the synergetic use of ultrasound with microbubbles and droplets, and microbubble/droplet drug-loading strategies for anticancer applications and disease modulation. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theranostic tool.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
72
|
Jing B, Lindsey BD. Effect of Skull Porous Trabecular Structure on Transcranial Ultrasound Imaging in the Presence of Elastic Wave Mode Conversion at Varying Incidence Angle. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2734-2748. [PMID: 34140169 DOI: 10.1016/j.ultrasmedbio.2021.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 06/12/2023]
Abstract
With the advancement of aberration correction techniques, transcranial ultrasound imaging has exhibited great potential in applications such as imaging neurological function and guiding therapeutic ultrasound. However, the feasibility of transcranial imaging varies among individuals because of the differences in skull acoustic properties. To better understand the fundamental mechanisms underlying the variation in imaging performance, the effect of the structure of the porous trabecular bone on transcranial imaging performance (i.e., target localization errors and resolution) was investigated for the first time through the use of elastic wave simulations and experiments. Simulation studies using high-resolution computed tomography data from ex vivo skull samples revealed that imaging at large incidence angles reduced the target localization error for skulls having low porosity; however, as skull porosity increased, large angles of incidence resulted in degradation of resolution and increased target localization errors. Experimental results indicate that imaging at normal incidence introduced a localization error of 1.85 ± 0.10 mm, while imaging at a large incidence angle (40°) resulted in an increased localization error of 6.54 ± 1.33 mm and caused a single point target to no longer appear as a single, coherent target in the resulting image, which is consistent with simulation results. This first investigation of the effects of skull microstructure on transcranial ultrasound imaging indicates that imaging performance is highly dependent on the porosity of the skull, particularly at non-normal angles of incidence.
Collapse
Affiliation(s)
- Bowen Jing
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Brooks D Lindsey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA; School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
| |
Collapse
|
73
|
Emerging Therapeutic Strategies for Brain Tumors. Neuromolecular Med 2021; 24:23-34. [PMID: 34406634 DOI: 10.1007/s12017-021-08681-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022]
Abstract
Nearly thirty thousand incidences of primary and 300 thousand incidences of metastatic brain cancer are diagnosed in the USA each year. It has a high mortality rate and is often unresponsive to the standard of care, which includes surgical resection, radiation, and chemotherapy. These treatment strategies are also hindered by their invasiveness and toxic effects on healthy cells and tissues. Furthermore, the blood-brain/tumor barrier severely limits delivery of anti-cancer therapeutics administered intravenously to brain tumors, resulting in poor tumor response to the treatment. There is a critical need to develop new approaches to brain cancer therapy that can overcome these limitations. Focused ultrasound has emerged as a modality that addresses many of these limitations and has the potential to alter the treatment paradigm for brain cancer. Ultrasound transmitted through the skull can be focused on tumors and used for targeted ablation or opening the vascular barriers for drug delivery. This review provides insight on the current status of these unique ultrasound techniques, different strategies of using this technique for brain cancer, experience in preclinical models, and potential for clinical translation. We also debate the safety perspective of these techniques and discuss potential avenues for future work in noninvasive planning, monitoring, and evaluation of the ultrasonic neurointervention.
Collapse
|
74
|
Pouliopoulos AN, Kwon N, Jensen G, Meaney A, Niimi Y, Burgess MT, Ji R, McLuckie AJ, Munoz FA, Kamimura HAS, Teich AF, Ferrera VP, Konofagou EE. Safety evaluation of a clinical focused ultrasound system for neuronavigation guided blood-brain barrier opening in non-human primates. Sci Rep 2021; 11:15043. [PMID: 34294761 PMCID: PMC8298475 DOI: 10.1038/s41598-021-94188-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
An emerging approach with potential in improving the treatment of neurodegenerative diseases and brain tumors is the use of focused ultrasound (FUS) to bypass the blood-brain barrier (BBB) in a non-invasive and localized manner. A large body of pre-clinical work has paved the way for the gradual clinical implementation of FUS-induced BBB opening. Even though the safety profile of FUS treatments in rodents has been extensively studied, the histological and behavioral effects of clinically relevant BBB opening in large animals are relatively understudied. Here, we examine the histological and behavioral safety profile following localized BBB opening in non-human primates (NHPs), using a neuronavigation-guided clinical system prototype. We show that FUS treatment triggers a short-lived immune response within the targeted region without exacerbating the touch accuracy or reaction time in visual-motor cognitive tasks. Our experiments were designed using a multiple-case-study approach, in order to maximize the acquired data and support translation of the FUS system into human studies. Four NHPs underwent a single session of FUS-mediated BBB opening in the prefrontal cortex. Two NHPs were treated bilaterally at different pressures, sacrificed on day 2 and 18 post-FUS, respectively, and their brains were histologically processed. In separate experiments, two NHPs that were earlier trained in a behavioral task were exposed to FUS unilaterally, and their performance was tracked for at least 3 weeks after BBB opening. An increased microglia density around blood vessels was detected on day 2, but was resolved by day 18. We also detected signs of enhanced immature neuron presence within areas that underwent BBB opening, compared to regions with an intact BBB, confirming previous rodent studies. Logistic regression analysis showed that the NHP cognitive performance did not deteriorate following BBB opening. These preliminary results demonstrate that neuronavigation-guided FUS with a single-element transducer is a non-invasive method capable of reversibly opening the BBB, without substantial histological or behavioral impact in an animal model closely resembling humans. Future work should confirm the observations of this multiple-case-study work across animals, species and tasks.
Collapse
Affiliation(s)
- Antonios N. Pouliopoulos
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Nancy Kwon
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Greg Jensen
- grid.21729.3f0000000419368729Department of Neuroscience, Columbia University, New York City, NY 10032 USA
| | - Anna Meaney
- grid.21729.3f0000000419368729Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY 10027 USA
| | - Yusuke Niimi
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Mark T. Burgess
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Robin Ji
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Alicia J. McLuckie
- grid.21729.3f0000000419368729Institute of Comparative Medicine, Columbia University, New York City, NY 10032 USA
| | - Fabian A. Munoz
- grid.21729.3f0000000419368729Department of Neuroscience, Columbia University, New York City, NY 10032 USA ,grid.21729.3f0000000419368729Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY 10027 USA
| | - Hermes A. S. Kamimura
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA
| | - Andrew F. Teich
- grid.21729.3f0000000419368729Department of Pathology and Cell Biology, Columbia University, New York City, NY 10032 USA
| | - Vincent P. Ferrera
- grid.21729.3f0000000419368729Department of Neuroscience, Columbia University, New York City, NY 10032 USA ,grid.21729.3f0000000419368729Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY 10027 USA ,grid.21729.3f0000000419368729Department of Psychiatry, Columbia University, New York City, NY
10032
USA
| | - Elisa E. Konofagou
- grid.21729.3f0000000419368729Department of Biomedical Engineering, Columbia University, New York City, NY 10032 USA ,grid.21729.3f0000000419368729Department of Radiology, Columbia University, New York City, NY 10032 USA
| |
Collapse
|
75
|
Karakatsani ME, Pouliopoulos AN, Liu M, Jambawalikar SR, Konofagou EE. Contrast-Free Detection of Focused Ultrasound-Induced Blood-Brain Barrier Opening Using Diffusion Tensor Imaging. IEEE Trans Biomed Eng 2021; 68:2499-2508. [PMID: 33360980 DOI: 10.1109/tbme.2020.3047575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Focused ultrasound (FUS) has emerged as a non-invasive technique to locally and reversibly disrupt the blood-brain barrier (BBB). Here, we investigate the use of diffusion tensor imaging (DTI) as a means of detecting FUS-induced BBB opening at the absence of an MRI contrast agent. A non-human primate (NHP) was repeatedly treated with FUS and preformed circulating microbubbles to transiently disrupt the BBB (n = 4). T1- and diffusion-weighted MRI scans were acquired after the ultrasound treatment, with and without gadolinium-based contrast agent, respectively. Both scans were registered with a high-resolution T1-weighted scan of the NHP to investigate signal correlations. DTI detected an increase in fractional anisotropy from 0.21 ± 0.02 to 0.38 ± 0.03 (82.6 ± 5.2% change) within the targeted area one hour after BBB opening. Enhanced DTI contrast overlapped by 77.22 ± 9.2% with hyper-intense areas of gadolinium-enhanced T1-weighted scans, indicating diffusion anisotropy enhancement only within the BBB opening volume. Diffusion was highly anisotropic and unidirectional within the treated brain region, as indicated by the direction of the principal diffusion eigenvectors. Polar and azimuthal angle ranges decreased by 35.6% and 82.4%, respectively, following BBB opening. Evaluation of the detection methodology on a second NHP (n = 1) confirmed the across-animal feasibility of the technique. In conclusion, DTI may be used as a contrast-free MR imaging modality in lieu of contrast-enhanced T1 mapping for detecting BBB opening during focused-ultrasound treatment or evaluating BBB integrity in brain-related pathologies.
Collapse
|
76
|
Chen KT, Wei KC, Liu HL. Focused Ultrasound Combined with Microbubbles in Central Nervous System Applications. Pharmaceutics 2021; 13:pharmaceutics13071084. [PMID: 34371774 PMCID: PMC8308978 DOI: 10.3390/pharmaceutics13071084] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022] Open
Abstract
The blood–brain barrier (BBB) protects the central nervous system (CNS) from invasive pathogens and maintains the homeostasis of the brain. Penetrating the BBB has been a major challenge in the delivery of therapeutic agents for treating CNS diseases. Through a physical acoustic cavitation effect, focused ultrasound (FUS) combined with microbubbles achieves the local detachment of tight junctions of capillary endothelial cells without inducing neuronal damage. The bioavailability of therapeutic agents is increased only in the area targeted by FUS energy. FUS with circulating microbubbles is currently the only method for inducing precise, transient, reversible, and noninvasive BBB opening (BBBO). Over the past decade, FUS-induced BBBO (FUS-BBBO) has been preclinically confirmed to not only enhance the penetration of therapeutic agents in the CNS, but also modulate focal immunity and neuronal activity. Several recent clinical human trials have demonstrated both the feasibility and potential advantages of using FUS-BBBO in diseased patients. The promising results support adding FUS-BBBO as a multimodal therapeutic strategy in modern CNS disease management. This review article explores this technology by describing its physical mechanisms and the preclinical findings, including biological effects, therapeutic concepts, and translational design of human medical devices, and summarizes completed and ongoing clinical trials.
Collapse
Affiliation(s)
- Ko-Ting Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan;
- Ph.D. Program in Biomedical Engineering, Chang Gung University, Guishan, Taoyuan 333, Taiwan
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan;
- Neuroscience Research Center, Linkou Chang Gung Memorial Hospital, Guishan, Taoyuan 333, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, TuCheng, New Taipei 236, Taiwan
- School of Medicine, Chang Gung University, Guishan, Taoyuan 333, Taiwan
- Correspondence: (K.-C.W.); (H.-L.L.)
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Da’an, Taipei 106, Taiwan
- Department of Biomedical Engineering, National Taiwan University, Da’an, Taipei 106, Taiwan
- Correspondence: (K.-C.W.); (H.-L.L.)
| |
Collapse
|
77
|
Li M, Gu J, Vu T, Sankin G, Zhong P, Yao J, Jing Y. Time-Resolved Passive Cavitation Mapping Using the Transient Angular Spectrum Approach. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:2361-2369. [PMID: 33635787 PMCID: PMC8269954 DOI: 10.1109/tuffc.2021.3062357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Passive cavitation mapping (PCM), which generates images using bubble acoustic emission signals, has been increasingly used for monitoring and guiding focused ultrasound surgery (FUS). PCM can be used as an adjunct to magnetic resonance imaging to provide crucial information on the safety and efficacy of FUS. The most widely used algorithm for PCM is delay-and-sum (DAS). One of the major limitations of DAS is its suboptimal computational efficiency. Although frequency-domain DAS can partially resolve this issue, such an algorithm is not suitable for imaging the evolution of bubble activity in real time and for cases in which cavitation events occur asynchronously. This study investigates a transient angular spectrum (AS) approach for PCM. The working principle of this approach is to backpropagate the received signal to the domain of interest and reconstruct the spatial-temporal wavefield encoded with the bubble location and collapse time. The transient AS approach is validated using an in silico model and water bath experiments. It is found that the transient AS approach yields similar results to DAS, but it is one order of magnitude faster. The results obtained by this study suggest that the transient AS approach is promising for fast and accurate PCM.
Collapse
|
78
|
Fletcher SMP, Choi M, Ramesh R, O'Reilly MA. Focused Ultrasound-Induced Blood-Spinal Cord Barrier Opening Using Short-Burst Phase-Keying Exposures in Rats: A Parameter Study. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1747-1760. [PMID: 33879388 DOI: 10.1016/j.ultrasmedbio.2021.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 06/12/2023]
Abstract
Transient opening of the blood-spinal cord barrier has the potential to improve drug delivery options to the spinal cord. We previously developed short-burst phase-keying exposures to reduce focal depth of field and mitigate standing waves in the spinal canal. However, optimal short-burst phase-keying parameters for drug delivery have not been identified. Here, the effects of pressure, treatment duration, pulse length, burst repetition frequency and burst length on resulting tissue effects were investigated. Increased in situ pressures (0.23-0.33 MPa) led to increased post-treatment T1-weighted contrast enhancement in magnetic resonance imaging (p = 0.015). Increased treatment duration (120 vs. 300 s) led to increased enhancement, but without statistical significance (p = 0.056). Increased burst repetition frequency (20 vs. 40 kHz) yielded a non-significant increase in enhancement (p = 0.064) but corresponded with increased damage observed on histology. No difference was observed in enhancement between pulse lengths of 2 and 10 ms (p = 0.912), corresponding with a sharp drop in the recorded second harmonic signal during the first 2 ms of the pulse. Increasing the burst length from two to five cycles (514 kHz) led to increased enhancement (p = 0.014). Results indicate that increasing the burst length may be the most effective method to enhance drug delivery. Additionally, shorter pulse lengths may allow more interleaved targets, and therefore a larger treatment volume, within one sonication.
Collapse
Affiliation(s)
- Stecia-Marie P Fletcher
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Min Choi
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ranjith Ramesh
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
79
|
Sun T, Shi Q, Zhang Y, Power C, Hoesch C, Antonelli S, Schroeder MK, Caldarone BJ, Taudte N, Schenk M, Hettmann T, Schilling S, McDannold NJ, Lemere CA. Focused ultrasound with anti-pGlu3 Aβ enhances efficacy in Alzheimer's disease-like mice via recruitment of peripheral immune cells. J Control Release 2021; 336:443-456. [PMID: 34186148 DOI: 10.1016/j.jconrel.2021.06.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022]
Abstract
Pyroglutamate-3 amyloid-β (pGlu3 Aβ) is an N-terminally modified, pathogenic form of amyloid-β that is present in cerebral amyloid plaques and vascular deposits. Here, we used focused ultrasound (FUS) with microbubbles to enhance the intravenous delivery of an Fc-competent anti-pGlu3 Aβ monoclonal antibody, 07/2a mAb, across the blood brain barrier (BBB) in an attempt to improve Aβ removal and memory in aged APP/PS1dE9 mice, an Alzheimer's disease (AD)-like model of amyloidogenesis. First, we demonstrated that bilateral hippocampal FUS-BBB disruption (FUS-BBBD) led to a 5.5-fold increase of 07/2a mAb delivery to the brains compared to non-sonicated mice 72 h following a single treatment. Then, we determined that three weekly treatments with 07/2a mAb alone improved spatial learning and memory in aged, plaque-rich APP/PS1dE9 mice, and that this improvement occurred faster and in a higher percentage of animals when combined with FUS-BBBD. Mice given the combination treatment had reduced hippocampal plaque burden compared to PBS-treated controls. Furthermore, synaptic protein levels were higher in hippocampal synaptosomes from mice given the combination treatment compared to sham controls, and there were more CA3 synaptic puncta labeled in the APP/PS1dE9 mice given the combination treatment compared to those given mAb alone. Plaque-associated microglia were present in the hippocampi of APP/PS1dE9 mice treated with 07/2a mAb with and without FUS-BBBD. However, we discovered that plaque-associated Ly6G+ monocytes were only present in the hippocampi of APP/PS1dE9 mice that were given FUS-BBBD alone or even more so, the combination treatment. Lastly, FUS-BBBD did not increase the incidence of microhemorrhage in mice with or without 07/2a mAb treatment. Our findings suggest that FUS is a useful tool to enhance delivery and efficacy of an anti-pGlu3 Aβ mAb for immunotherapy either via an additive effect or an independent mechanism. We revealed a potential novel mechanism wherein the combination of 07/2a mAb with FUS-BBBD led to greater monocyte infiltration and recruitment to plaques in this AD-like model. Overall, these effects resulted in greater plaque removal, sparing of synapses and improved cognitive function without causing overt damage, suggesting the possibility of FUS-BBBD as a noninvasive method to increase the therapeutic efficacy of drugs or biologics in AD patients.
Collapse
Affiliation(s)
- Tao Sun
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Qiaoqiao Shi
- Ann Romney Center for Neurologic Diseases in the Department of Neurology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Yongzhi Zhang
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Chanikarn Power
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Camilla Hoesch
- Ann Romney Center for Neurologic Diseases in the Department of Neurology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America
| | - Shawna Antonelli
- Ann Romney Center for Neurologic Diseases in the Department of Neurology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America
| | - Maren K Schroeder
- Ann Romney Center for Neurologic Diseases in the Department of Neurology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America
| | - Barbara J Caldarone
- Harvard Medical School Mouse Behavior Core, Boston, MA, United States of America
| | - Nadine Taudte
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Halle (Saale), Germany
| | - Mathias Schenk
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Halle (Saale), Germany
| | | | - Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department Molecular Drug Biochemistry and Therapy, Halle (Saale), Germany; Vivoryon Therapeutics AG, Halle (Saale), Germany; Anhalt University of Applied Sciences, Köthen, Germany
| | - Nathan J McDannold
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America.
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases in the Department of Neurology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States of America; Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
80
|
Bunevicius A, McDannold NJ, Golby AJ. Focused Ultrasound Strategies for Brain Tumor Therapy. Oper Neurosurg (Hagerstown) 2021; 19:9-18. [PMID: 31853548 DOI: 10.1093/ons/opz374] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A key challenge in the medical treatment of brain tumors is the limited penetration of most chemotherapeutic agents across the blood-brain barrier (BBB) into the tumor and the infiltrative margin around the tumor. Magnetic resonance-guided focused ultrasound (MRgFUS) is a promising tool to enhance the delivery of chemotherapeutic agents into brain tumors. OBJECTIVE To review the mechanism of FUS, preclinical evidence, and clinical studies that used low-frequency FUS for a BBB opening in gliomas. METHODS Literature review. RESULTS The potential of externally delivered low-intensity ultrasound for a temporally and spatially precise and predictable disruption of the BBB has been investigated for over a decade, yielding extensive preclinical literature demonstrating that FUS can disrupt the BBB in a spatially targeted and temporally reversible manner. Studies in animal models documented that FUS enhanced the delivery of numerous chemotherapeutic and investigational agents across the BBB and into brain tumors, including temozolomide, bevacizumab, 1,3-bis (2-chloroethyl)-1-nitrosourea, doxorubicin, viral vectors, and cells. Chemotherapeutic interventions combined with FUS slowed tumor progression and improved animal survival. Recent advances of MRgFUS systems allow precise, temporally and spatially controllable, and safe transcranial delivery of ultrasound energy. Initial clinical evidence in glioma patients has shown the efficacy of MRgFUS in disrupting the BBB, as demonstrated by an enhanced gadolinium penetration. CONCLUSION Thus far, a temporary disruption of the BBB followed by the administration of chemotherapy has been both feasible and safe. Further studies are needed to determine the actual drug delivery, including the drug distribution at a tissue-level scale, as well as effects on tumor growth and patient prognosis.
Collapse
Affiliation(s)
- Adomas Bunevicius
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Nathan Judson McDannold
- Harvard Medical School, Harvard University, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alexandra J Golby
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Harvard University, Boston, Massachusetts.,Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
81
|
Xhima K, McMahon D, Ntiri E, Goubran M, Hynynen K, Aubert I. Intravenous and Non-invasive Drug Delivery to the Mouse Basal ForebrainUsing MRI-guided Focused Ultrasound. Bio Protoc 2021; 11:e4056. [PMID: 34262999 PMCID: PMC8260260 DOI: 10.21769/bioprotoc.4056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/26/2021] [Accepted: 03/14/2021] [Indexed: 11/02/2022] Open
Abstract
Basal forebrain cholinergic neurons (BFCNs) regulate circuit dynamics underlying cognitive processing, including attention, memory, and cognitive flexibility. In Alzheimer's disease and related neurodegenerative conditions, the degeneration of BFCNs has long been considered a key player in cognitive decline. The cholinergic system thus represents a key therapeutic target. A long-standing obstacle for the development of effective cholinergic-based therapies is not only the production of biologically active compounds but also a platform for safe and efficient drug delivery to the basal forebrain. The blood-brain barrier (BBB) presents a significant challenge for drug delivery to the brain, excluding approximately 98% of small-molecule biologics and nearly 100% of large-molecule therapeutic agents from entry into the brain parenchyma. Current modalities to achieve effective drug delivery to deep brain structures, such as the basal forebrain, are particularly limited. Direct intracranial injection via a needle or catheter carries risks associated with invasive neurosurgery. Intra-arterial injection of hyperosmotic solutions or therapeutics modified to penetrate the BBB using endogenous transport systems lack regional specificity, which may not always be desirable. Intranasal, intrathecal, and intraventricular administration have limited drug distribution beyond the brain surface. Here, we present a protocol for non-invasively, locally, and transiently increasing BBB permeability using MRI-guided focused ultrasound (MRIgFUS) in the murine basal forebrain for delivery of therapeutic agents targeting the cholinergic system. Ongoing work in preclinical models and clinical trials supports the safety and feasibility of MRIgFUS-mediated BBB modulation as a promising drug delivery modality for the treatment of debilitating neurological diseases.
Collapse
Affiliation(s)
- Kristiana Xhima
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Dallan McMahon
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Edward Ntiri
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Maged Goubran
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Kullervo Hynynen
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Isabelle Aubert
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
82
|
Yu K, Niu X, Krook-Magnuson E, He B. Intrinsic functional neuron-type selectivity of transcranial focused ultrasound neuromodulation. Nat Commun 2021; 12:2519. [PMID: 33947867 PMCID: PMC8097024 DOI: 10.1038/s41467-021-22743-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Transcranial focused ultrasound (tFUS) is a promising neuromodulation technique, but its mechanisms remain unclear. We hypothesize that if tFUS parameters exhibit distinct modulation effects in different neuron populations, then the mechanism can be understood through identifying unique features in these neuron populations. In this work, we investigate the effect of tFUS stimulation on different functional neuron types in in vivo anesthetized rodent brains. Single neuron recordings were separated into regular-spiking and fast-spiking units based on their extracellular spike shapes acquired through intracranial electrophysiological recordings, and further validated in transgenic optogenetic mice models of light-excitable excitatory and inhibitory neurons. We show that excitatory and inhibitory neurons are intrinsically different in response to ultrasound pulse repetition frequency (PRF). The results suggest that we can preferentially target specific neuron types noninvasively by tuning the tFUS PRF. Chemically deafened rats and genetically deafened mice were further tested for validating the directly local neural effects induced by tFUS without potential auditory confounds.
Collapse
Affiliation(s)
- Kai Yu
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Xiaodan Niu
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Bin He
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
83
|
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: 101] [Impact Index Per Article: 33.7] [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
|
84
|
D'Souza A, Dave KM, Stetler RA, S. Manickam D. Targeting the blood-brain barrier for the delivery of stroke therapies. Adv Drug Deliv Rev 2021; 171:332-351. [PMID: 33497734 DOI: 10.1016/j.addr.2021.01.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
A variety of neuroprotectants have shown promise in treating ischemic stroke, yet their delivery to the brain remains a challenge. The endothelial cells lining the blood-brain barrier (BBB) are emerging as a dynamic factor in the response to neurological injury and disease, and the endothelial-neuronal matrix coupling is fundamentally neuroprotective. In this review, we discuss approaches that target the endothelium for drug delivery both across the BBB and to the BBB as a viable strategy to facilitate neuroprotective effects, using the example of brain-derived neurotrophic factor (BDNF). We highlight the advances in cell-derived extracellular vesicles (EVs) used for CNS targeting and drug delivery. We also discuss the potential of engineered EVs as a potent strategy to deliver BDNF or other drug candidates to the ischemic brain, particularly when coupled with internal components like mitochondria that may increase cellular energetics in injured endothelial cells.
Collapse
|
85
|
Guo Y, Lee H, Fang Z, Velalopoulou A, Kim J, Thomas MB, Liu J, Abramowitz RG, Kim Y, Coskun AF, Krummel DP, Sengupta S, MacDonald TJ, Arvanitis C. Single-cell analysis reveals effective siRNA delivery in brain tumors with microbubble-enhanced ultrasound and cationic nanoparticles. SCIENCE ADVANCES 2021; 7:7/18/eabf7390. [PMID: 33931452 PMCID: PMC8087400 DOI: 10.1126/sciadv.abf7390] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/12/2021] [Indexed: 05/08/2023]
Abstract
RNA-based therapies offer unique advantages for treating brain tumors. However, tumor penetrance and uptake are hampered by RNA therapeutic size, charge, and need to be "packaged" in large carriers to improve bioavailability. Here, we have examined delivery of siRNA, packaged in 50-nm cationic lipid-polymer hybrid nanoparticles (LPHs:siRNA), combined with microbubble-enhanced focused ultrasound (MB-FUS) in pediatric and adult preclinical brain tumor models. Using single-cell image analysis, we show that MB-FUS in combination with LPHs:siRNA leads to more than 10-fold improvement in siRNA delivery into brain tumor microenvironments of the two models. MB-FUS delivery of Smoothened (SMO) targeting siRNAs reduces SMO protein production and markedly increases tumor cell death in the SMO-activated medulloblastoma model. Moreover, our analysis reveals that MB-FUS and nanoparticle properties can be optimized to maximize delivery in the brain tumor microenvironment, thereby serving as a platform for developing next-generation tunable delivery systems for RNA-based therapy in brain tumors.
Collapse
Affiliation(s)
- Yutong Guo
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hohyun Lee
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anastasia Velalopoulou
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jinhwan Kim
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Midhun Ben Thomas
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jingbo Liu
- Department of Pediatrics, Aflac Cancer and Blood, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan G Abramowitz
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - YongTae Kim
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Aflac Cancer and Blood, Emory University School of Medicine, Atlanta, GA, USA
| | - Costas Arvanitis
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
86
|
Fan CH, Kao WF, Kang ST, Ho YJ, Yeh CK. Exploring the Acoustic and Dynamic Characteristics of Phase-Change Droplets. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:1051-1061. [PMID: 33079650 DOI: 10.1109/tuffc.2020.3032441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Acoustic droplet vaporization (ADV) provides the on-demand production of bubbles for use in ultrasound (US)-based diagnostic and therapeutic applications. The droplet-to-bubble transition process has been shown to involve localized internal gas nucleation, followed by a volume expansion of threefold to fivefold and inertial bubble oscillation, all of which take place within a few microseconds. Monitoring these ADV processes is important in gauging the mechanical effects of phase-change droplets in a biological environment, but this is difficult to achieve using regular optical observations. In this study, we utilized acoustic characterization [i.e., simultaneous passive cavitation detection (PCD) and active cavitation detection (ACD)] to investigate the acoustic signatures emitted from phase-change droplets ADV and determined their correlations with the physical behaviors observed using high-speed optical imaging. The experimental results showed that activation with three-cycle 5-MHz US pulse resulted in the droplets (diameter: 3.0- [Formula: see text]) overexpanding and undergoing damped oscillation before settling to bubbles with a final diameter. Meanwhile, a broadband shock wave was observed at the beginning of the PCD signal. The intense fluctuations of the ACD signal revealed that the shock wave arose from the inertial cavitation of nucleated small gas pockets in the droplets. It was particularly interesting that another shock-wave signal with a much lower acoustic frequency (< 2 MHz) was observed at about [Formula: see text] after the first half signal. This signal coincided with the reduction of the ACD signal amplitude that indicated the rebound of the transforming bubble. Since internal gas nucleation is a crucial process of ADV, the first half signal may indicate the occurrence of an ADV event, and the second half signal may further reveal the degrees of expansion and oscillation of the bubble. These acoustic signatures provide opportunities for monitoring ADV dynamics based on the detection of acoustic signals.
Collapse
|
87
|
Wei P, Cornel EJ, Du J. Ultrasound-responsive polymer-based drug delivery systems. Drug Deliv Transl Res 2021; 11:1323-1339. [PMID: 33761101 PMCID: PMC7989687 DOI: 10.1007/s13346-021-00963-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
Ultrasound-responsive polymeric materials have received a tremendous amount of attention from scientists for several decades. Compared to other stimuli-responsive materials (such as UV-, thermal-, and pH-responsive materials), these smart materials are more applicable since they allow more efficient drug delivery and targeted treatment by fairly non-invasive means. This review describes the recent advances of such ultrasound-responsive polymer-based drug delivery systems and illustrates various applications. More specifically, the mechanism of ultrasound-induced drug delivery, typical formulations, and biomedical applications (tumor therapy, disruption of blood-brain barrier, fighting infectious diseases, transdermal drug delivery, and enhanced thrombolysis) are summarized. Finally, a perspective on the future research directions for the development of ultrasound-responsive polymeric materials to facilitate a clinical translation is given.
Collapse
Affiliation(s)
- Ping Wei
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Erik Jan Cornel
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai, 201804, China. .,Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
88
|
McMahon D, O'Reilly MA, Hynynen K. Therapeutic Agent Delivery Across the Blood-Brain Barrier Using Focused Ultrasound. Annu Rev Biomed Eng 2021; 23:89-113. [PMID: 33752471 DOI: 10.1146/annurev-bioeng-062117-121238] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Specialized features of vasculature in the central nervous system greatly limit therapeutic treatment options for many neuropathologies. Focused ultrasound, in combination with circulating microbubbles, can be used to transiently and noninvasively increase cerebrovascular permeability with a high level of spatial precision. For minutes to hours following sonication, drugs can be administered systemically to extravasate in the targeted brain regions and exert a therapeutic effect, after which permeability returns to baseline levels. With the wide range of therapeutic agents that can be delivered using this approach and the growing clinical need, focused ultrasound and microbubble (FUS+MB) exposure in the brain has entered human testing to assess safety. This review outlines the use of FUS+MB-mediated cerebrovascular permeability enhancement as a drug delivery technique, details several technical and biological considerations of this approach, summarizes results from the clinical trials conducted to date, and discusses the future direction of the field.
Collapse
Affiliation(s)
- Dallan McMahon
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N 3M5, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| |
Collapse
|
89
|
Characterization of ultrasound-mediated delivery of trastuzumab to normal and pathologic spinal cord tissue. Sci Rep 2021; 11:4412. [PMID: 33627726 PMCID: PMC7904756 DOI: 10.1038/s41598-021-83874-x] [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: 10/20/2020] [Accepted: 02/08/2021] [Indexed: 01/31/2023] Open
Abstract
Extensive studies on focused ultrasound (FUS)-mediated drug delivery through the blood-brain barrier have been published, yet little work has been published on FUS-mediated drug delivery through the blood-spinal cord barrier (BSCB). This work aims to quantify the delivery of the monoclonal antibody trastuzumab to rat spinal cord tissue and characterize its distribution within a model of leptomeningeal metastases. 10 healthy Sprague-Dawley rats were treated with FUS + trastuzumab and sacrificed at 2-h or 24-h post-FUS. A human IgG ELISA (Abcam) was used to measure trastuzumab concentration and a 12 ± fivefold increase was seen in treated tissue over control tissue at 2 h versus no increase at 24 h. Three athymic nude rats were inoculated with MDA-MB-231-H2N HER2 + breast cancer cells between the meninges in the thoracic region of the spinal cord and treated with FUS + trastuzumab. Immunohistochemistry was performed to visualize trastuzumab delivery, and semi-quantitative analysis revealed similar or more intense staining in tumor tissue compared to healthy tissue suggesting a comparable or greater concentration of trastuzumab was achieved. FUS can increase the permeability of the BSCB, improving drug delivery to specifically targeted regions of healthy and pathologic tissue in the spinal cord. The achieved concentrations within the healthy tissue are comparable to those reported in the brain.
Collapse
|
90
|
McMahon D, Deng L, Hynynen K. Comparing rapid short-pulse to tone burst sonication sequences for focused ultrasound and microbubble-mediated blood-brain barrier permeability enhancement. J Control Release 2021; 329:696-705. [PMID: 33022327 DOI: 10.1016/j.jconrel.2020.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Transcranial focused ultrasound and microbubble (FUS + MB) exposure enables targeted, noninvasive drug delivery to the brain. Given the protective nature of the blood-brain barrier (BBB), the development of sonication strategies that maximize therapeutic efficacy while minimizing the risk of tissue damage are essential. This work aimed to compare the safety of 10 ms tone bursts, widely used in the field, to a recently described rapid short-pulse (RaSP) sequence, while accounting for drug delivery potential. MATERIALS AND METHODS Forty-one male wild-type mice received FUS + MB exposure (1.78 MHz driving frequency; 0.5 Hz burst repetition frequency; 250 s duration; 40 μl/kg Definity) at a range of fixed pressure amplitudes. A RaSP sequence (13 five-cycle pulses/10 ms burst) was compared to 10 ms tone bursts (B10). For animals in cohort #1 (n = 26), T1 mapping was used to quantify gadobutrol extravasation. Three targets, temporarily separated by 10 min, were sonicated in each brain to compare the time dependence of BBB permeability enhancement between sequences. Red blood cell (RBC) extravasation was quantified to assess vascular damage. For animals in cohort #2 (n = 18), a single target was sonicated per brain. BBB permeability enhancement was compared between sequences by T1 mapping and the extravasation of a 3 kDa fluorescent dextran. RESULTS At a peak negative pressure of 400 kPa, the B10 sequence produced an order of magnitude greater gadobutrol and dextran extravasation compared to RaSP (p < 0.01). When accounting for BBB permeability enhancement magnitude, as measure by T1 mapping, no differences were observed between sequences in the pattern of dextran or albumin extravasation in tissue sections; however, the frequency of RBC extravasation was found to be 5 times greater with the RaSP sequence (p = 0.02). At pressure amplitudes resulting in similar levels of gadobutrol extravasation, no significant differences were observed in the time dependence of BBB permeability enhancement between sequences. CONCLUSION When accounting for the magnitude of BBB permeability enhancement, and thus the potential for drug delivery, the RaSP sequence tested here did not produce measurable improvements over the B10 sequence and may present an increased risk of vascular damage.
Collapse
Affiliation(s)
- Dallan McMahon
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| | - Lulu Deng
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
91
|
Kamimura HAS, Wu SY, Grondin J, Ji R, Aurup C, Zheng W, Heidmann M, Pouliopoulos AN, Konofagou EE. Real-Time Passive Acoustic Mapping Using Sparse Matrix Multiplication. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:164-177. [PMID: 32746182 PMCID: PMC7770101 DOI: 10.1109/tuffc.2020.3001848] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Passive acoustic mapping enables the spatiotemporal monitoring of cavitation with circulating microbubbles during focused ultrasound (FUS)-mediated blood-brain barrier opening. However, the computational load for processing large data sets of cavitation maps or more complex algorithms limit the visualization in real-time for treatment monitoring and adjustment. In this study, we implemented a graphical processing unit (GPU)-accelerated sparse matrix-based beamforming and time exposure acoustics in a neuronavigation-guided ultrasound system for real-time spatiotemporal monitoring of cavitation. The system performance was tested in silico through benchmarking, in vitro using nonhuman primate (NHP) and human skull specimens, and demonstrated in vivo in NHPs. We demonstrated the stability of the cavitation map for integration times longer than 62.5 [Formula: see text]. A compromise between real-time displaying and cavitation map quality obtained from beamformed RF data sets with a size of 2000 ×128 ×30 (axial [Formula: see text]) was achieved for an integration time of [Formula: see text], which required a computational time of 0.27 s (frame rate of 3.7 Hz) and could be displayed in real-time between pulses at PRF = 2 Hz. Our benchmarking tests show that the GPU sparse-matrix algorithm processed the RF data set at a computational rate of [Formula: see text]/pixel/sample, which enables adjusting the frame rate and the integration time as needed. The neuronavigation system with real-time implementation of cavitation mapping facilitated the localization of the cavitation activity and helped to identify distortions due to FUS phase aberration. The in vivo test of the method demonstrated the feasibility of GPU-accelerated sparse matrix computing in a close to a clinical condition, where focus distortions exemplify problems during treatment. These experimental conditions show the need for spatiotemporal monitoring of cavitation with real-time capability that enables the operator to correct or halt the sonication in case substantial aberrations are observed.
Collapse
|
92
|
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
|
93
|
Meng Y, Jones RM, Davidson B, Huang Y, Pople CB, Surendrakumar S, Hamani C, Hynynen K, Lipsman N. Technical Principles and Clinical Workflow of Transcranial MR-Guided Focused Ultrasound. Stereotact Funct Neurosurg 2020; 99:329-342. [PMID: 33302282 DOI: 10.1159/000512111] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/28/2020] [Indexed: 11/19/2022]
Abstract
Transcranial MR-guided focused ultrasound (MRgFUS) is a rapidly developing technology in neuroscience for manipulating brain structure and function without open surgery. The effectiveness of transcranial MRgFUS for thermoablation is well established, and the technique is actively employed worldwide for movement disorders including essential tremor. A growing number of centers are also investigating the potential of microbubble-mediated focused ultrasound-induced opening of the blood-brain barrier (BBB) for targeted drug delivery to the brain. Here, we provide a technical overview of the principles, clinical workflow, and operator considerations of transcranial MRgFUS procedures for both thermoablation and BBB opening.
Collapse
Affiliation(s)
- Ying Meng
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ryan M Jones
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Davidson
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Yuexi Huang
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Christopher B Pople
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | | | - Clement Hamani
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Nir Lipsman
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada, .,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada,
| |
Collapse
|
94
|
Pouliopoulos AN, Smith CAB, Bezer JH, El Ghamrawy A, Sujarittam K, Bouldin CJ, Morse SV, Tang MX, Choi JJ. Doppler Passive Acoustic Mapping. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:2692-2703. [PMID: 32746222 DOI: 10.1109/tuffc.2020.3011657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In therapeutic ultrasound using microbubbles, it is essential to drive the microbubbles into the correct type of activity and the correct location to produce the desired biological response. Although passive acoustic mapping (PAM) is capable of locating where microbubble activities are generated, it is well known that microbubbles rapidly move within the ultrasound beam. We propose a technique that can image microbubble movement by estimating their velocities within the focal volume. Microbubbles embedded within a wall-less channel of a tissue-mimicking material were sonicated using 1-MHz focused ultrasound. The acoustic emissions generated by the microbubbles were captured with a linear array (L7-4). PAM with robust Capon beamforming was used to localize the microbubble acoustic emissions. We spectrally analyzed the time trace of each position and isolated the higher harmonics. Microbubble velocity maps were constructed from the position-dependent Doppler shifts at different time points during sonication. Microbubbles moved primarily away from the transducer at velocities on the order of 1 m/s due to primary acoustic radiation forces, producing a time-dependent velocity distribution. We detected microbubble motion both away and toward the receiving array, revealing the influence of acoustic radiation forces and fluid motion due to the ultrasound exposure. High-speed optical images confirmed the acoustically measured microbubble velocities. Doppler PAM enables passive estimation of microbubble motion and may be useful in therapeutic applications, such as drug delivery across the blood-brain barrier, sonoporation, sonothrombolysis, and drug release.
Collapse
|
95
|
Shi L, Jiang Y, Zhang Y, Lan L, Huang Y, Cheng JX, Yang C. A fiber optoacoustic emitter with controlled ultrasound frequency for cell membrane sonoporation at submillimeter spatial resolution. PHOTOACOUSTICS 2020; 20:100208. [PMID: 33101926 PMCID: PMC7569214 DOI: 10.1016/j.pacs.2020.100208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 05/13/2023]
Abstract
Focused ultrasound has attracted great attention in minimally invasive therapeutic and mechanism studies. Frequency below 1 MHz is identified preferable for high-efficiency bio-modulation. However, the poor spatial confinement of several millimeters and large device diameter of ∼25 mm of typical sub-MHz ultrasound technology suffered from the diffraction limit, severely hindering its further applications. To address it, a fiber-based optoacoustic emitter (FOE) is developed, serving as a miniaturized ultrasound point source, with sub-millimeter confinement, composed of an optical diffusion layer and an expansion layer on an optical fiber. By modifying acoustic damping and light absorption performance, controllable frequencies in the range of 0.083 MHz-5.500 MHz are achieved and further induce cell membrane sonoporation with frequency dependent efficiency. By solving the problem of compromise between sub-MHz frequency and sub-millimeter precision via breaking the diffraction limit, the FOE shows a great potential in region-specific drug delivery, gene transfection and neurostimulation.
Collapse
Affiliation(s)
- Linli Shi
- Department of Chemistry, Boston University, 580 Commonwealth Avenue, Boston, MA 02215, USA
| | - Ying Jiang
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
| | - Yi Zhang
- Department of Physics, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA
| | - Lu Lan
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
| | - Yimin Huang
- Department of Chemistry, Boston University, 580 Commonwealth Avenue, Boston, MA 02215, USA
| | - Ji-Xin Cheng
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, 8 St. Mary’s Street, Boston, MA 02215, USA
- Corresponding authors at: Department of Electrical and Computer Engineering, 8 St. Mary’s Street, Boston, MA 02215, USA.
| | - Chen Yang
- Department of Chemistry, Boston University, 580 Commonwealth Avenue, Boston, MA 02215, USA
- Department of Electrical and Computer Engineering, 8 St. Mary’s Street, Boston, MA 02215, USA
- Corresponding authors at: Department of Electrical and Computer Engineering, 8 St. Mary’s Street, Boston, MA 02215, USA.
| |
Collapse
|
96
|
Zhang X, Ye D, Yang L, Yue Y, Sultan D, Pacia CP, Pang H, Detering L, Heo GS, Luehmann H, Choksi A, Sethi A, Limbrick DD, Becher OJ, Tai YC, Rubin JB, Chen H, Liu Y. Magnetic Resonance Imaging-Guided Focused Ultrasound-Based Delivery of Radiolabeled Copper Nanoclusters to Diffuse Intrinsic Pontine Glioma. ACS APPLIED NANO MATERIALS 2020; 3:11129-11134. [PMID: 34337344 PMCID: PMC8320805 DOI: 10.1021/acsanm.0c02297] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an invasive pediatric brainstem malignancy exclusively in children without effective treatment due to the often-intact blood-brain tumor barrier (BBTB), an impediment to the delivery of therapeutics. Herein, we used focused ultrasound (FUS) to transiently open BBTB and delivered radiolabeled nanoclusters (64Cu-CuNCs) to tumors for positron emission tomography (PET) imaging and quantification in a mouse DIPG model. First, we optimized FUS acoustic pressure to open the blood-brain barrier (BBB) for effective delivery of 64Cu-CuNCs to pons in wildtype mice. Then the optimized FUS pressure was used to deliver radiolabeled agents in DIPG mouse. Magnetic resonance imaging (MRI)-guided FUS-induced BBTB opening was demonstrated using a low molecular weight, short-lived 68Ga-DOTA-ECL1i radiotracer and PET/CT before and after treatment. We then compared the delivery efficiency of 64Cu-CuNCs to DIPG tumor with and without FUS treatment and demonstrated the FUS-enhanced delivery and time-dependent diffusion of 64Cu-CuNCs within the tumor.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neurosurgery, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Dezhuang Ye
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, Saint Louis, MO 63130, USA
- Department of Neurosurgery, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Lihua Yang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Pham Pacia
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hannah Pang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ankur Choksi
- School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Abhishek Sethi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Oren J Becher
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yuan-Chuan Tai
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO 63108, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
97
|
Recent Advances on Ultrasound Contrast Agents for Blood-Brain Barrier Opening with Focused Ultrasound. Pharmaceutics 2020; 12:pharmaceutics12111125. [PMID: 33233374 PMCID: PMC7700476 DOI: 10.3390/pharmaceutics12111125] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
The blood-brain barrier is the primary obstacle to efficient intracerebral drug delivery. Focused ultrasound, in conjunction with microbubbles, is a targeted and non-invasive way to disrupt the blood-brain barrier. Many commercially available ultrasound contrast agents and agents specifically designed for therapeutic purposes have been investigated in ultrasound-mediated blood-brain barrier opening studies. The new generation of sono-sensitive agents, such as liquid-core droplets, can also potentially disrupt the blood-brain barrier after their ultrasound-induced vaporization. In this review, we describe the different compositions of agents used for ultrasound-mediated blood-brain barrier opening in recent studies, and we discuss the challenges of the past five years related to the optimal formulation of agents.
Collapse
|
98
|
Huh H, Park TY, Seo H, Han M, Jung B, Choi HJ, Lee EH, Pahk KJ, Kim H, Park J. A local difference in blood-brain barrier permeability in the caudate putamen and thalamus of a rat brain induced by focused ultrasound. Sci Rep 2020; 10:19286. [PMID: 33159137 PMCID: PMC7648079 DOI: 10.1038/s41598-020-76259-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/08/2020] [Indexed: 01/17/2023] Open
Abstract
A blood–brain barrier (BBB) opening induced by focused ultrasound (FUS) has been widely studied as an effective way of treating brain diseases. We investigate the effect of ultrasound’s incidence angle at caudate putamen (Cp) and thalamus (Th) of the rat brain by inducing the same power of focused ultrasound that corresponds to the acoustic pressure of 0.65 MPa in free field. The BBB permeability (Ktrans) was quantitatively evaluated with dynamic contrast-enhanced magnetic resonance imaging. The group averaged (n = 11) maximum Ktrans at Cp (0.021 ± 0.012 min−1) was 1.39 times smaller than the Ktrans of Th (0.029 ± 0.01 min−1) with p = 0.00343. The group averaged (n = 6) ultrasound’s incidence angles measured using the computed tomography image of rat skulls were compared with the maximum Ktrans and showed a negatively linear relation R2 = 0.7972). The maximum acoustic pressure computed from the acoustic simulation showed higher average acoustic pressures at Th (0.37 ± 0.02 MPa) compared to pressures at Cp (0.32 ± 0.01 MPa) with p = 0.138 × 10−11. More red blood cell were observed at the Th region compared to the Cp region in the tissue staining. These results indicate that localized characteristics of the sonication target within the subject should be considered for safer and more efficient BBB disruption induced by FUS.
Collapse
Affiliation(s)
- Hyungkyu Huh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Tae Young Park
- Center for Bionics, Korea Institute of Science and Technology, Seoul, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Korea
| | - Hyeon Seo
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Mun Han
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Byeongjin Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Hyo Jin Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Eun-Hee Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Ki Joo Pahk
- Center for Bionics, Korea Institute of Science and Technology, Seoul, Korea
| | - Hyungmin Kim
- Center for Bionics, Korea Institute of Science and Technology, Seoul, Korea. .,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Korea.
| | - Juyoung Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea.
| |
Collapse
|
99
|
Lapin NA, Gill K, Shah BR, Chopra R. Consistent opening of the blood brain barrier using focused ultrasound with constant intravenous infusion of microbubble agent. Sci Rep 2020; 10:16546. [PMID: 33024157 PMCID: PMC7538995 DOI: 10.1038/s41598-020-73312-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 09/08/2020] [Indexed: 12/03/2022] Open
Abstract
The blood brain barrier (BBB) is a major obstacle to the delivery of therapeutics to the brain. Focused ultrasound (FUS) in combination with microbubbles can non-invasively open the BBB in a targeted manner. Bolus intravenous injections of microbubbles are standard practice, but dynamic influx and clearance mechanisms prevent delivery of a uniform dose with time. When multiple targets are selected for sonication in a single treatment, uniform serum concentrations of microbubbles are important for consistent BBB opening. Herein, we show that bubble infusions were able to achieve consistent BBB opening at multiple target sites. FUS exposures were conducted with different Definity microbubble concentrations at various acoustic pressures. To quantify the effects of infusion on BBB opening, we calculated the MRI contrast enhancement rate. When infusions were performed at rates of 7.2 µl microbubbles/kg/min or below, we were able to obtain consistent BBB opening without injury at all pressures. However, when infusion rates exceeded 20 µl/kg/min, signs of injury occurred at pressures from 0.39 to 0.56 MPa. When compared to bolus injections, a bubble infusion offers a more controlled and consistent approach to multi-target BBB disruption.
Collapse
Affiliation(s)
- Norman A Lapin
- Focused Ultrasound Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kirt Gill
- Focused Ultrasound Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Bhavya R Shah
- Focused Ultrasound Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Neurosurgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA.,Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rajiv Chopra
- Focused Ultrasound Laboratory, Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA. .,Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
100
|
Dauba A, Goulas J, Colin L, Jourdain L, Larrat B, Gennisson JL, Certon D, Novell A. Evaluation of capacitive micromachined ultrasonic transducers for passive monitoring of microbubble-assisted ultrasound therapies. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2020; 148:2248. [PMID: 33138521 DOI: 10.1121/10.0002096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Passive cavitation detection can be performed to monitor microbubble activity during brain therapy. Microbubbles under ultrasound exposure generate a response characterized by multiple nonlinear emissions. Here, the wide bandwidth of capacitive micromachined ultrasonic transducers (CMUTs) was exploited to monitor the microbubble signature through a rat skull and a macaque skull. The intrinsic nonlinearity of the CMUTs was characterized in receive mode. Indeed, undesirable nonlinear components generated by the CMUTs must be minimized as they can mask the microbubble harmonic response. The microbubble signature at harmonic and ultra-harmonic components (0.5-6 MHz) was successfully extracted through a rat skull using moderate bias voltage.
Collapse
Affiliation(s)
- Ambre Dauba
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, 91401, France
| | - Jordane Goulas
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, 91401, France
| | - Laurent Colin
- GREMAN CNRS UMR 7347, Université François Rabelais, INSA Centre Val de Loire, Tours, France
| | - Laurène Jourdain
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, 91401, France
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, Baobab, NeuroSpin, Gif-sur-Yvette, 91191, France
| | - Jean-Luc Gennisson
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, 91401, France
| | - Dominique Certon
- GREMAN CNRS UMR 7347, Université François Rabelais, INSA Centre Val de Loire, Tours, France
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, 91401, France
| |
Collapse
|