1
|
Pakdaman Zangabad R, Lee H, Zhang X, Sait Kilinc M, Arvanitis CD, Levent Degertekin F. A High Sensitivity CMUT-Based Passive Cavitation Detector for Monitoring Microbubble Dynamics During Focused Ultrasound Interventions. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1087-1096. [PMID: 39088497 DOI: 10.1109/tuffc.2024.3436918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Tracking and controlling microbubble (MB) dynamics in the human brain through acoustic emission (AE) monitoring during transcranial focused ultrasound (tFUS) therapy are critical for attaining safe and effective treatments. The low-amplitude MB emissions have harmonic and ultra-harmonic components, necessitating a broad bandwidth and low-noise system for monitoring transcranial MB activity. Capacitive micromachined ultrasonic transducers (CMUTs) offer high sensitivity and low noise over a broad bandwidth, especially when they are tightly integrated with electronics, making them a good candidate technology for monitoring the MB activity through human skull. In this study, we designed a 16-channel analog front-end (AFE) electronics with a low-noise transimpedance amplifier (TIA), a band-gap reference circuit, and an output buffer stage. To assess AFE performance and ability to detect MB AE, we combined it with a commercial CMUT array. The integrated system has 12.3 - [Formula: see text] receive sensitivity with 0.085 - [Formula: see text] minimum detectable pressure (MDP) up to 3 MHz for a single element CMUT with 3.78 [Formula: see text] area. Experiments with free MBs in a microfluidic channel demonstrate that our system is able to capture key spectral components of MBs' harmonics when sonicated at clinically relevant frequencies (0.5 MHz) and pressures (250 kPa). Together our results demonstrate that the proposed CMUT system can support the development of novel passive cavitation detectors (PCD) to track MB activity for attaining safe and effective focused ultrasound (FUS) treatments.
Collapse
|
2
|
Tang J, Feng M, Wang D, Zhang L, Yang K. Recent advancement of sonogenetics: A promising noninvasive cellular manipulation by ultrasound. Genes Dis 2024; 11:101112. [PMID: 38947740 PMCID: PMC11214298 DOI: 10.1016/j.gendis.2023.101112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 07/02/2024] Open
Abstract
Recent advancements in biomedical research have underscored the importance of noninvasive cellular manipulation techniques. Sonogenetics, a method that uses genetic engineering to produce ultrasound-sensitive proteins in target cells, is gaining prominence along with optogenetics, electrogenetics, and magnetogenetics. Upon stimulation with ultrasound, these proteins trigger a cascade of cellular activities and functions. Unlike traditional ultrasound modalities, sonogenetics offers enhanced spatial selectivity, improving precision and safety in disease treatment. This technology broadens the scope of non-surgical interventions across a wide range of clinical research and therapeutic applications, including neuromodulation, oncologic treatments, stem cell therapy, and beyond. Although current literature predominantly emphasizes ultrasonic neuromodulation, this review offers a comprehensive exploration of sonogenetics. We discuss ultrasound properties, the specific ultrasound-sensitive proteins employed in sonogenetics, and the technique's potential in managing conditions such as neurological disorders, cancer, and ophthalmic diseases, and in stem cell therapies. Our objective is to stimulate fresh perspectives for further research in this promising field.
Collapse
Affiliation(s)
- Jin Tang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingxuan Feng
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ke Yang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| |
Collapse
|
3
|
Seo JP, Trippett JS, Huang Z, Lee S, Nouraein S, Wang RZ, Szablowski JO. Acoustically targeted measurement of transgene expression in the brain. SCIENCE ADVANCES 2024; 10:eadj7686. [PMID: 39110811 PMCID: PMC11305388 DOI: 10.1126/sciadv.adj7686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Gene expression is a critical component of brain physiology, but monitoring this expression in the living brain represents a major challenge. Here, we introduce a new paradigm called recovery of markers through insonation (REMIS) for noninvasive measurement of gene expression in the brain with cell type, spatial, and temporal specificity. Our approach relies on engineered protein markers that are produced in neurons but exit into the brain's interstitium. When ultrasound is applied to targeted brain regions, it opens the blood-brain barrier and releases these markers into the bloodstream. Once in blood, the markers can be readily detected using biochemical techniques. REMIS can noninvasively confirm gene delivery and measure endogenous signaling in specific brain sites through a simple insonation and a subsequent blood test. REMIS is reliable and demonstrated consistent improvement in recovery of markers from the brain into the blood. Overall, this work establishes a noninvasive, spatially specific method of monitoring gene delivery and endogenous signaling in the brain.
Collapse
Affiliation(s)
- Joon Pyung Seo
- Applied Physics Program, Rice University, Houston, TX, USA
| | | | - Zhimin Huang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Sangsin Lee
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Shirin Nouraein
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, USA
| | - Ryan Z. Wang
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Jerzy O. Szablowski
- Applied Physics Program, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX, USA
- Rice Neuroengineering Initiative, Rice University, Houston, TX, USA
| |
Collapse
|
4
|
McDannold N, Wen PY, Reardon DA, Fletcher SM, Golby AJ. Cavitation monitoring, treatment strategy, and acoustic simulations of focused ultrasound blood-brain barrier disruption in patients with glioblastoma. J Control Release 2024; 372:194-208. [PMID: 38897294 PMCID: PMC11299340 DOI: 10.1016/j.jconrel.2024.06.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE We report our experience disrupting the blood-brain barrier (BBB) to improve drug delivery in glioblastoma patients receiving temozolomide chemotherapy. The goals of this retrospective analysis were to compare MRI-based measures of BBB disruption and vascular damage to the exposure levels, acoustic emissions data, and acoustic simulations. We also simulated the cavitation detectors. METHODS Monthly BBB disruption (BBBD) was performed using a 220 kHz hemispherical phased array focused ultrasound system (Exablate Neuro, InSightec) and Definity microbubbles (Lantheus) over 38 sessions in nine patients. Exposure levels were actively controlled via the cavitation dose obtained by monitoring subharmonic acoustic emissions. The acoustic field and sensitivity profile of the cavitation detection system were simulated. Exposure levels and cavitation metrics were compared to the level of BBBD evident in contrast-enhanced MRI and to hypointense regions in T2*-weighted MRI. RESULTS Our treatment strategy evolved from using a relatively high cavitation dose goal to a lower goal and longer sonication duration and ultimately resulted in BBBD across the treatment volume with minimal petechiae. Subsonication-level feedback control of the exposure using acoustic emissions also improved consistency. Simulations of the acoustic field suggest that reflections and standing waves appear when the focus is placed near the skull, but their effects can be mitigated with aberration correction. Simulating the cavitation detectors suggest variations in the sensitivity profile across the treatment volume and between patients. A correlation was observed with the cavitation dose, BBBD and petechial hemorrhage in 8/9 patients, but substantial variability was evident. Analysis of the cavitation spectra found that most bursts did not contain wideband emissions, a signature of inertial cavitation, but biggest contribution to the cavitation dose - the metric used to control the procedure - came from bursts with wideband emissions. CONCLUSION Using a low subharmonic cavitation dose with a longer duration resulted in BBBD with minimal petechiae. The correlation between cavitation dose and outcomes demonstrates the benefits of feedback control based on acoustic emissions, although more work is needed to reduce variability. Acoustic simulations could improve focusing near the skull and inform our analysis of acoustic emissions. Monitoring additional frequency bands and improving the sensitivity of the cavitation detection could provide signatures of microbubble activity associated with BBB disruption that were undetected here and could improve our ability to achieve BBB disruption without vascular damage.
Collapse
Affiliation(s)
- Nathan McDannold
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America.
| | - Patrick Y Wen
- Department of Neuro-oncology, Dana Farber Cancer Institute, Boston, MA, United States of America
| | - David A Reardon
- Department of Neuro-oncology, Dana Farber Cancer Institute, Boston, MA, United States of America
| | - Stecia-Marie Fletcher
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Alexandra J Golby
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, United States of America
| |
Collapse
|
5
|
Martinez PJ, Song JJ, Garay FG, Song KH, Mufford T, Steiner J, DeSisto J, Ellens N, Serkova NJ, Green AL, Borden M. Comprehensive assessment of blood-brain barrier opening and sterile inflammatory response: unraveling the therapeutic window. Sci Rep 2024; 14:17036. [PMID: 39043894 PMCID: PMC11266505 DOI: 10.1038/s41598-024-67916-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
Microbubbles (MBs) combined with focused ultrasound (FUS) has emerged as a promising noninvasive technique to permeabilize the blood-brain barrier (BBB) for drug delivery into the brain. However, the safety and biological consequences of BBB opening (BBBO) remain incompletely understood. This study aims to investigate the effects of two parameters mediating BBBO: microbubble volume dose (MVD) and mechanical index (MI). High-resolution MRI-guided FUS was employed in mouse brains to assess BBBO by manipulating these two parameters. Afterward, the sterile inflammatory response (SIR) was studied 6 h post-FUS treatment. Results demonstrated that both MVD and MI significantly influenced the extent of BBBO, with higher MVD and MI leading to increased permeability. Moreover, RNA sequencing revealed upregulation of major inflammatory pathways and immune cell infiltration after BBBO, indicating the presence and extent of SIR. Gene set enrichment analysis identified 12 gene sets associated with inflammatory responses that were significantly upregulated at higher MVD or MI. A therapeutic window was established between therapeutically relevant BBBO and the onset of SIR, providing operating regimes to avoid damage from stimulation of the NFκB pathway via TNFɑ signaling to apoptosis. These results contribute to the optimization and standardization of BBB opening parameters for safe and effective drug delivery to the brain and further elucidate the underlying molecular mechanisms driving sterile inflammation.
Collapse
Affiliation(s)
- Payton J Martinez
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Jane J Song
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Francis G Garay
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kang-Ho Song
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Toni Mufford
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jenna Steiner
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - John DeSisto
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Nicholas Ellens
- Alpheus Medical, Inc., 1266 Park Rd., Chanhassen, MN, 55317, USA
| | - Natalie J Serkova
- Department of Radiology, Cancer Center Animal Imaging Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Adam L Green
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Mark Borden
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO, 80303, USA.
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA.
| |
Collapse
|
6
|
Bae S, Liu K, Pouliopoulos AN, Ji R, Jiménez-Gambín S, Yousefian O, Kline-Schoder AR, Batts AJ, Tsitsos FN, Kokossis D, Mintz A, Honig LS, Konofagou EE. Transcranial blood-brain barrier opening in Alzheimer's disease patients using a portable focused ultrasound system with real-time 2-D cavitation mapping. Theranostics 2024; 14:4519-4535. [PMID: 39113808 PMCID: PMC11303073 DOI: 10.7150/thno.94206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/14/2024] [Indexed: 08/10/2024] Open
Abstract
Background : Focused ultrasound (FUS) in combination with microbubbles has recently shown great promise in facilitating blood-brain barrier (BBB) opening for drug delivery and immunotherapy in Alzheimer's disease (AD). However, it is currently limited to systems integrated within the MRI suites or requiring post-surgical implants, thus restricting its widespread clinical adoption. In this pilot study, we investigate the clinical safety and feasibility of a portable, non-invasive neuronavigation-guided FUS (NgFUS) system with integrated real-time 2-D microbubble cavitation mapping. Methods : A phase 1 clinical study with mild to moderate AD patients (N = 6) underwent a single session of microbubble-mediated NgFUS to induce transient BBB opening (BBBO). Microbubble activity under FUS was monitored with real-time 2-D cavitation maps and dosing to ensure the efficacy and safety of the NgFUS treatment. Post-operative MRI was used for BBB opening and closure confirmation as well as safety assessment. Changes in AD biomarker levels in both blood serum and extracellular vesicles (EVs) were evaluated, while changes in amyloid-beta (Aβ) load in the brain were assessed through 18F-florbetapir PET. Results : BBBO was achieved in 5 out of 6 subjects with an average volume of 983 ± 626 mm3 following FUS at the right frontal lobe both in white and gray matter regions. The outpatient treatment was completed within 34.8 ± 10.7 min. Cavitation dose significantly correlated with the BBBO volume (R 2 > 0.9, N = 4), demonstrating the portable NgFUS system's capability of predicting opening volumes. The cavitation maps co-localized closely with the BBBO location, representing the first report of real-time transcranial 2-D cavitation mapping in the human brain. Larger opening volumes correlated with increased levels of AD biomarkers, including Aβ42 (R 2 = 0.74), Tau (R 2 = 0.95), and P-Tau181 (R 2 = 0.86), assayed in serum-derived EVs sampled 3 days after FUS (N = 5). From PET scans, subjects showed a lower Aβ load increase in the treated frontal lobe region compared to the contralateral region. Reduction in asymmetry standardized uptake value ratios (SUVR) correlated with the cavitation dose (R 2 > 0.9, N = 3). Clinical changes in the mini-mental state examination over 6 months were within the expected range of cognitive decline with no additional changes observed as a result of FUS. Conclusion : We showed the safety and feasibility of this cost-effective and time-efficient portable NgFUS treatment for BBBO in AD patients with the first demonstration of real-time 2-D cavitation mapping. The cavitation dose correlated with BBBO volume, a slowed increase in pathology, and serum detection of AD proteins. Our study highlights the potential for accessible FUS treatment in AD, with or without drug delivery.
Collapse
Affiliation(s)
- Sua Bae
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Keyu Liu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | | | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | | | - Omid Yousefian
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | | | - Alec J. Batts
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Fotios N. Tsitsos
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Danae Kokossis
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lawrence S. Honig
- Department of Neurology and Taub Institute, Columbia University Irving Medical Center 10032, New York, NY, USA
| | - Elisa E. Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
7
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
8
|
Zhao P, Wu T, Tian Y, You J, Cui X. Recent advances of focused ultrasound induced blood-brain barrier opening for clinical applications of neurodegenerative diseases. Adv Drug Deliv Rev 2024; 209:115323. [PMID: 38653402 DOI: 10.1016/j.addr.2024.115323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/21/2023] [Accepted: 04/20/2024] [Indexed: 04/25/2024]
Abstract
With the aging population on the rise, neurodegenerative disorders have taken center stage as a significant health concern. The blood-brain barrier (BBB) plays an important role to maintain the stability of central nervous system, yet it poses a formidable obstacle to delivering drugs for neurodegenerative disease therapy. Various methods have been devised to confront this challenge, each carrying its own set of limitations. One particularly promising noninvasive approach involves the utilization of focused ultrasound (FUS) combined with contrast agents-microbubbles (MBs) to achieve transient and reversible BBB opening. This review provides a comprehensive exploration of the fundamental mechanisms behind FUS/MBs-mediated BBB opening and spotlights recent breakthroughs in its application for neurodegenerative diseases. Furthermore, it addresses the current challenges and presents future perspectives in this field.
Collapse
Affiliation(s)
- Pengxuan Zhao
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Pharmacy, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Tiantian Wu
- School of Pharmacy, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Yu Tian
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai 200000, China
| | - Jia You
- School of Pharmacy, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Xinwu Cui
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
9
|
Chen L, Song J, Richter-Stretton G, Lee W, Padmanabhan P, Götz J. Multimodal evaluation of blood-brain barrier opening in mice in response to low-intensity ultrasound and a claudin-5 binder. Nanotheranostics 2024; 8:427-441. [PMID: 38961889 PMCID: PMC11217787 DOI: 10.7150/ntno.95146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/05/2024] [Indexed: 07/05/2024] Open
Abstract
Background: The blood-brain barrier (BBB) is a major bottleneck in delivering therapeutics to the brain. Treatment strategies to transiently open this barrier include focused ultrasound combined with intravenously injected microbubbles (FUS+MB) and targeting of molecules that regulate BBB permeability. Methods: Here, we investigated BBB opening mediated by the claudin-5 binder cCPEm (a microorganismal toxin in a truncated form) and FUS+MB at a centre frequency of 1 MHz, assessing dextran uptake, broadband emission, and endogenous immunoglobulin G (IgG) extravasation. Results: FUS+MB-induced BBB opening was detectable at a pressure ≥0.35 MPa when assessed for leakage of 10 and 70 kDa dextran, and at ≥0.2 MPa for uptake of endogenous IgG. Treating mice with 20 mg/kg cCPEm failed to open the BBB, and pre-treatment with cCPEm followed by FUS+MB at 0.2 and 0.3 MPa did not overtly increase BBB opening compared to FUS+MB alone. Using passive cavitation detection (PCD), we found that broadband emission correlated with the peak negative pressure (PNP) and dextran leakage, indicating the possibility of using broadband emission for developing a feedback controller to monitor BBB opening. Conclusions: Together, our study highlights the challenges in developing combinatorial approaches to open the BBB and presents an additional IgG-based histological detection method for BBB opening.
Collapse
Affiliation(s)
| | | | | | | | | | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
10
|
Memari E, Khan D, Alkins R, Helfield B. Focused ultrasound-assisted delivery of immunomodulating agents in brain cancer. J Control Release 2024; 367:283-299. [PMID: 38266715 DOI: 10.1016/j.jconrel.2024.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Focused ultrasound (FUS) combined with intravascularly circulating microbubbles can transiently increase the permeability of the blood-brain barrier (BBB) to enable targeted therapeutic delivery to the brain, the clinical testing of which is currently underway in both adult and pediatric patients. Aside from traditional cancer drugs, this technique is being extended to promote the delivery of immunomodulating therapeutics to the brain, including antibodies, immune cells, and cytokines. In this manner, FUS approaches are being explored as a tool to improve and amplify the effectiveness of immunotherapy for both primary and metastatic brain cancer, a particularly challenging solid tumor to treat. Here, we present an overview of the latest groundbreaking research in FUS-assisted delivery of immunomodulating agents to the brain in pre-clinical models of brain cancer, and place it within the context of the current immunotherapy approaches. We follow this up with a discussion on new developments and emerging strategies for this rapidly evolving approach.
Collapse
Affiliation(s)
- Elahe Memari
- Department of Physics, Concordia University, Montreal H4B 1R6, Canada
| | - Dure Khan
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Ryan Alkins
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Division of Neurosurgery, Department of Surgery, Kingston Health Sciences Centre, Queen's University, Kingston, ON, Canada
| | - Brandon Helfield
- Department of Physics, Concordia University, Montreal H4B 1R6, Canada; Department of Biology, Concordia University, Montreal H4B 1R6, Canada.
| |
Collapse
|
11
|
Manuel TJ, Sigona MK, Phipps MA, Kusunose J, Luo H, Yang PF, Newton AT, Gore JC, Grissom W, Chen LM, Caskey CF. Small volume blood-brain barrier opening in macaques with a 1 MHz ultrasound phased array. J Control Release 2023; 363:707-720. [PMID: 37827222 DOI: 10.1016/j.jconrel.2023.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/14/2023]
Abstract
The use of focused ultrasound to open the blood-brain barrier (BBB) has the potential to deliver drugs to specific regions of the brain. The size of the BBB opening and ability to localize the opening determines the spatial extent and is a limiting factor in many applications of BBB opening where targeting a small brain region is desired. Here we evaluate the performance of a system designed for small opening volumes and highlight the unique challenges associated with pushing the spatial precision of this technique. To achieve small volume openings in cortical regions of the macaque brain, we tested a custom 1 MHz array transducer integrated into a magnetic resonance image-guided focused ultrasound system. Using real-time cavitation monitoring, we demonstrated twelve instances of single sonication, small volume BBB opening with average volumes of 59 ± 37 mm3 and 184 ± 2 mm3 in cortical and subcortical targets, respectively. We found high correlation between subject-specific acoustic simulations and observed openings when incorporating grey matter segmentation (R2 = 0.8577), and the threshold for BBB opening based on simulations was 0.53 MPa. Analysis of MRI-based safety assessment and cavitation signals indicate a safe pressure range for 1 MHz BBB opening and suggest that our system can be used to deliver drugs and gene therapy to small brain regions.
Collapse
Affiliation(s)
- Thomas J Manuel
- Vanderbilt University, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Michelle K Sigona
- Vanderbilt University, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - M Anthony Phipps
- Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Jiro Kusunose
- Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Huiwen Luo
- Vanderbilt University, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Pai-Feng Yang
- Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Allen T Newton
- Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - John C Gore
- Vanderbilt University, Nashville, TN, USA; Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - William Grissom
- Vanderbilt University, Nashville, TN, USA; Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Li Min Chen
- Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Charles F Caskey
- Vanderbilt University, Nashville, TN, USA; Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Institute of Imaging Science, Nashville, TN, USA.
| |
Collapse
|
12
|
Mondou P, Mériaux S, Nageotte F, Vappou J, Novell A, Larrat B. State of the art on microbubble cavitation monitoring and feedback control for blood-brain-barrier opening using focused ultrasound. Phys Med Biol 2023; 68:18TR03. [PMID: 37369229 DOI: 10.1088/1361-6560/ace23e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/27/2023] [Indexed: 06/29/2023]
Abstract
Focused ultrasound (FUS) is a non-invasive and highly promising method for targeted and reversible blood-brain barrier permeabilization. Numerous preclinical studies aim to optimize the localized delivery of drugs using this method in rodents and non-human primates. Several clinical trials have been initiated to treat various brain diseases in humans using simultaneous BBB permeabilization and drug injection. This review presents the state of the art ofin vitroandin vivocavitation control algorithms for BBB permeabilization using microbubbles (MB) and FUS. Firstly, we describe the different cavitation states, their physical significance in terms of MB behavior and their translation into the spectral composition of the backscattered signal. Next, we report the different indexes calculated and used during the ultrasonic monitoring of cavitation. Finally, the differentin vitroandin vivocavitation control strategies described in the literature are presented and compared.
Collapse
Affiliation(s)
- Paul Mondou
- Université de Strasbourg, CNRS, ICube, UMR7357, Strasbourg, France
- Université Paris-Saclay, CEA, CNRS, BAOBAB, NeuroSpin, 91191, Gif-sur-Yvette, France
| | - Sébastien Mériaux
- Université Paris-Saclay, CEA, CNRS, BAOBAB, NeuroSpin, 91191, Gif-sur-Yvette, France
| | - Florent Nageotte
- Université de Strasbourg, CNRS, ICube, UMR7357, Strasbourg, France
| | - Jonathan Vappou
- Université de Strasbourg, CNRS, ICube, UMR7357, Strasbourg, France
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, BAOBAB, NeuroSpin, 91191, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, 91401 , Orsay, France
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, BAOBAB, NeuroSpin, 91191, Gif-sur-Yvette, France
| |
Collapse
|
13
|
Navarro-Becerra JA, Borden MA. Targeted Microbubbles for Drug, Gene, and Cell Delivery in Therapy and Immunotherapy. Pharmaceutics 2023; 15:1625. [PMID: 37376072 DOI: 10.3390/pharmaceutics15061625] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Microbubbles are 1-10 μm diameter gas-filled acoustically-active particles, typically stabilized by a phospholipid monolayer shell. Microbubbles can be engineered through bioconjugation of a ligand, drug and/or cell. Since their inception a few decades ago, several targeted microbubble (tMB) formulations have been developed as ultrasound imaging probes and ultrasound-responsive carriers to promote the local delivery and uptake of a wide variety of drugs, genes, and cells in different therapeutic applications. The aim of this review is to summarize the state-of-the-art of current tMB formulations and their ultrasound-targeted delivery applications. We provide an overview of different carriers used to increase drug loading capacity and different targeting strategies that can be used to enhance local delivery, potentiate therapeutic efficacy, and minimize side effects. Additionally, future directions are proposed to improve the tMB performance in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, CO 80309, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
14
|
Seo JP, Trippett JS, Huang Z, Wang RZ, Lee S, Szablowski JO. Acoustically-Targeted Measurement of Transgene Expression in the Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541868. [PMID: 37292585 PMCID: PMC10245922 DOI: 10.1101/2023.05.23.541868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gene expression is a critical component of brain physiology and activity, but monitoring this expression in the living brain represents a significant challenge. Here, we introduce a new paradigm called Recovery of Markers through InSonation (REMIS) for noninvasive measurement of gene expression in the brain with cell-type, spatial, and temporal specificity. Our approach relies on engineered protein markers that are designed to be expressed in neurons and exit into the interstitium. By applying ultrasound to targeted brain regions, these markers are released into the bloodstream, where they can be readily detected using biochemical techniques. REMIS can noninvasively confirm gene delivery and measure endogenous signaling in specific brain sites through a simple insonation and a subsequent blood test. Using REMIS, we successfully measured chemogenetic induction of neuronal activity in ultrasound-tar-geted brain regions. REMIS recovery of markers is reliable and demonstrated improved recovery of markers from the brain into the blood in every tested animal. Overall, our work establishes a noninvasive, spatially-specific means of monitoring gene delivery outcomes and endogenous signaling in mammalian brains, opening up possibilities for brain research and noninvasive monitoring of gene therapies in the brain.
Collapse
|
15
|
Qin D, Zou Q, Zhong X, Zhang B, Li Z. Effects of medium viscoelasticity on bubble collapse strength of interacting polydisperse bubbles. ULTRASONICS SONOCHEMISTRY 2023; 95:106375. [PMID: 36965309 PMCID: PMC10060372 DOI: 10.1016/j.ultsonch.2023.106375] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/27/2023] [Accepted: 03/14/2023] [Indexed: 06/06/2023]
Abstract
Due to its physical and/or chemical effects, acoustic cavitation plays a crucial role in various emerging applications ranging from advanced materials to biomedicine. The cavitation bubbles usually undergo oscillatory dynamics and violent collapse within a viscoelastic medium, which are closely related to the cavitation-associated effects. However, the role of medium viscoelasticity on the cavitation dynamics has received little attention, especially for the bubble collapse strength during multi-bubble cavitation with the complex interactions between size polydisperse bubbles. In this study, modified Gilmore equations accounting for inter-bubble interactions were coupled with the Zener viscoelastic model to simulate the dynamics of multi-bubble cavitation in viscoelastic media. Results showed that the cavitation dynamics (e.g., acoustic resonant response, nonlinear oscillation behavior and bubble collapse strength) of differently-sized bubbles depend differently on the medium viscoelasticity and each bubble is affected by its neighboring bubbles to a different degree. More specifically, increasing medium viscosity drastically dampens the bubble dynamics and weakens the bubble collapse strength, while medium elasticity mainly affects the bubble resonance at which the bubble collapse strength is maximum. Differently-sized bubbles can achieve resonances and even subharmonic resonances at high driving acoustic pressures as the elasticity changes to certain values, and the resonance frequency of each bubble increases with the elasticity increasing. For the interactions between the size polydisperse bubbles, it indicated that the largest bubble generally has a dominant effect on the dynamics of smaller ones while in turn it is almost unaffected, exhibiting a pattern of destructive and constructive interactions. This study provides a valuable insight into the acoustic cavitation dynamics of multiple interacting polydisperse bubbles in viscoelastic media, which may offer a potential of controlling the medium viscoelasticity to appropriately manipulate the dynamics of multi-bubble cavitation for achieving proper cavitation effects according to the desired application.
Collapse
Affiliation(s)
- Dui Qin
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, People's Republic of China; Postdoctoral Workstation of Chongqing People's Hospital, Chongqing, People's Republic of China.
| | - Qingqin Zou
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, People's Republic of China
| | - Xianhua Zhong
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, People's Republic of China
| | - Bingyu Zhang
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, People's Republic of China
| | - Zhangyong Li
- Department of Biomedical Engineering, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, People's Republic of China.
| |
Collapse
|
16
|
Manuel TJ, Sigona MK, Phipps MA, Kusunose J, Luo H, Yang PF, Newton AT, Gore JC, Grissom W, Chen LM, Caskey CF. Small volume blood-brain barrier opening in macaques with a 1 MHz ultrasound phased array. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.02.530815. [PMID: 36909495 PMCID: PMC10002751 DOI: 10.1101/2023.03.02.530815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Focused ultrasound blood-brain barrier (BBB) opening is a promising tool for targeted delivery of therapeutic agents into the brain. The volume of opening determines the extent of therapeutic administration and sets a lower bound on the size of targets which can be selectively treated. We tested a custom 1 MHz array transducer optimized for cortical regions in the macaque brain with the goal of achieving small volume openings. We integrated this device into a magnetic resonance image guided focused ultrasound system and demonstrated twelve instances of small volume BBB opening with average opening volumes of 59 ± 37 mm 3 and 184 ± 2 mm 3 in cortical and subcortical targets, respectively. We developed real-time cavitation monitoring using a passive cavitation detector embedded in the array and characterized its performance on a bench-top flow phantom mimicking transcranial BBB opening procedures. We monitored cavitation during in-vivo procedures and compared cavitation metrics against opening volumes and safety outcomes measured with FLAIR and susceptibility weighted MR imaging. Our findings show small BBB opening at cortical targets in macaques and characterize the safe pressure range for 1 MHz BBB opening. Additionally, we used subject-specific simulations to investigate variance in measured opening volumes and found high correlation (R 2 = 0.8577) between simulation predictions and observed measurements. Simulations suggest the threshold for 1 MHz BBB opening was 0.53 MPa. This system enables BBB opening for drug delivery and gene therapy to be targeted to more specific brain regions.
Collapse
|
17
|
Lin Y, O’Reilly MA, Hynynen K. A PVDF Receiver for Acoustic Monitoring of Microbubble-Mediated Ultrasound Brain Therapy. SENSORS (BASEL, SWITZERLAND) 2023; 23:1369. [PMID: 36772406 PMCID: PMC9921684 DOI: 10.3390/s23031369] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
The real-time monitoring of spectral characteristics of microbubble (MB) acoustic emissions permits the prediction of increases in blood-brain barrier (BBB) permeability and of tissue damage in MB-mediated focused ultrasound (FUS) brain therapy. Single-element passive cavitation detectors provide limited spatial information regarding MB activity, greatly affecting the performance of acoustic control. However, an array of receivers can be used to spatially map cavitation events and thus improve treatment control. The spectral content of the acoustic emissions provides additional information that can be correlated with the bio-effects, and wideband receivers can thus provide the most complete spectral information. Here, we develop a miniature polyvinylidene fluoride (PVDF thickness = 110 μm, active area = 1.2 mm2) broadband receiver for the acoustic monitoring of MBs. The receiver has superior sensitivity (2.36-3.87 V/MPa) to those of a commercial fibre-optic hydrophone in the low megahertz frequency range (0.51-5.4 MHz). The receiver also has a wide -6 dB acceptance angle (54 degrees at 1.1 MHz and 13 degrees at 5.4 MHz) and the ability to detect subharmonic and higher harmonic MB emissions in phantoms. The overall acoustic performance of this low-cost receiver indicates its suitability for the eventual use within an array for MB monitoring and mapping in preclinical studies.
Collapse
Affiliation(s)
- Yi Lin
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Meaghan A. O’Reilly
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Kullervo Hynynen
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
18
|
Batts AJ, Ji R, Noel RL, Kline-Schoder AR, Bae S, Kwon N, Konofagou EE. Using a novel rapid alternating steering angles pulse sequence to evaluate the impact of theranostic ultrasound-mediated ultra-short pulse length on blood-brain barrier opening volume and closure, cavitation mapping, drug delivery feasibility, and safety. Theranostics 2023; 13:1180-1197. [PMID: 36793858 PMCID: PMC9925313 DOI: 10.7150/thno.76199] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/08/2022] [Indexed: 02/16/2023] Open
Abstract
Background: Focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening is a noninvasive, safe and reversible technique for targeted drug delivery to the brain. Most preclinical systems developed to perform and monitor BBB opening are comprised of a separate geometrically focused transducer and passive cavitation detector (PCD) or imaging array. This study builds upon previous work from our group developing a single imaging phased array configuration for simultaneous BBB opening and monitoring called theranostic ultrasound (ThUS), leveraging ultra-short pulse lengths (USPLs) and a novel rapid alternating steering angles (RASTA) pulse sequence design for simultaneous bilateral sonications with target-specific USPL. The RASTA sequence was further employed to evaluate the impact of USPL on BBB opening volume, power cavitation imaging (PCI) pixel intensity, BBB closing timeline, drug delivery efficiency, and safety. Methods: A P4-1 phased array transducer driven by a Verasonics Vantage ultrasound system was operated using a custom script to run the RASTA sequence which consisted of interleaved steered, focused transmits and passive imaging. Contrast-enhanced magnetic resonance imaging (MRI) confirmed initial opening volume and closure of the BBB by longitudinal imaging through 72 hours post-BBB opening. For drug delivery experiments, mice were systemically administered a 70 kDa fluorescent dextran or adeno-associated virus serotype 9 (AAV9) for fluorescence microscopy or enzyme-linked immunosorbent assay (ELISA) to evaluate ThUS-mediated molecular therapeutic delivery. Additional brain sections were also H&E-stained to evaluate histological damage, and IBA1- and GFAP-stained to elucidate the effects of ThUS-mediated BBB opening on stimulation of key cell types involved in the neuro-immune response, microglia and astrocytes. Results: The ThUS RASTA sequence induced distinct BBB openings simultaneously in the same mouse where volume, PCI pixel intensity, level of dextran delivery, and AAV reporter transgene expression were correlated with brain hemisphere-specific USPL, consistent with statistically significant differences between 1.5, 5, and 10-cycle USPL groups. BBB closure after ThUS required 2-48 hours depending on USPL. The potential for acute damage and neuro-immune activation increased with USPL, but such observable damage was nearly reversed 96 hours post-ThUS. Conclusion: ThUS is a versatile single-array technique which exhibits the potential for investigating a variety of non-invasive therapeutic delivery applications in the brain.
Collapse
Affiliation(s)
- Alec J Batts
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Rebecca L Noel
- Department of Biomedical Engineering, Columbia University, New York, USA
| | | | - Sua Bae
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, USA.,Department of Radiology, Columbia University, New York, USA
| |
Collapse
|
19
|
Choi HJ, Han M, Seo H, Park CY, Lee EH, Park J. The new insight into the inflammatory response following focused ultrasound-mediated blood-brain barrier disruption. Fluids Barriers CNS 2022; 19:103. [PMID: 36564820 PMCID: PMC9783406 DOI: 10.1186/s12987-022-00402-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Despite the great potential of FUS-BBB disruption (FUS-BBBD), it is still controversial whether FUS-BBBD acts as an inducing factor of neuro-inflammation or not, and the biological responses after FUS-BBBD triggers the inflammatory process are poorly understood. The aim of this study is to investigate the safety window for FUS levels based on a comprehensive safety assessment. METHODS The mice were treated with two different ultrasound parameters (0.25 MPa and 0.42 MPa) in the thalamus region of brain. The efficacy of BBB opening was verified by dynamic contrast-enhanced MRI (DCE-MRI) and the cavitation monitoring. The transcriptome analysis was performed to investigate the molecular response for the two BBBD conditions after FUS-mediated BBB opening in time-dependent manners. Histological analysis was used for evaluation of the tissue damage, neuronal degeneration, and activation of glial cells induced by FUS-BBBD. RESULTS The BBBD, as quantified by the Ktrans, was approximately threefold higher in 0.42 MPa-treated group than 0.25 MPa-treated group. While the minimal tissue/cellular damage was found in 0.25 MPa-treated group, visible damages containing microhemorrhages and degenerating neurons were detected in 0.42 MPa-treated group in accordance with the extent of BBBD. In transcriptome analysis, 0.42 MPa-treated group exhibited highly dynamic changes in the expression levels of an inflammatory response or NF-κB pathway-relative genes in a time-dependent manner whereas, 0.25 MPa was not altered. Interestingly, although it is clear that 0.42 MPa induces neuroinflammation through glial activation, neuroprotective properties were evident by the expression of A2-type astrocytes. CONCLUSIONS Our findings propose that a well-defined BBBD parameter of 0.25 MPa could ensure the safety without cellular/tissue damage or sterile inflammatory response in the brain. Furthermore, the fact that the excessive sonication parameters at 0.42 MPa could induce a sterile inflammation response via glial activation suggested the possibility that could lead to tissue repair toward the homeostasis of the brain microenvironment through A2-type reactive astrocytes.
Collapse
Affiliation(s)
- Hyo Jin Choi
- grid.496160.c0000 0004 6401 4233Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hubub), 80, Cheombok-Ro, Dong-Gu, Daegu, 41061 Republic of Korea
| | - Mun Han
- grid.496160.c0000 0004 6401 4233Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hubub), 80, Cheombok-Ro, Dong-Gu, Daegu, 41061 Republic of Korea
| | - Hyeon Seo
- grid.256681.e0000 0001 0661 1492Department of Computer Science, Gyeongsang National University, 501, Jinju-Daero, Jinju, Gyeongsangnam-Do 52828 Republic of Korea
| | - Chan Yuk Park
- grid.496160.c0000 0004 6401 4233Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hubub), 80, Cheombok-Ro, Dong-Gu, Daegu, 41061 Republic of Korea
| | - Eun-Hee Lee
- grid.496160.c0000 0004 6401 4233Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hubub), 80, Cheombok-Ro, Dong-Gu, Daegu, 41061 Republic of Korea
| | - Juyoung Park
- grid.256155.00000 0004 0647 2973College of Future Industry, Department of High-Tech Medical Device, Gachon University, 1342, Seongnam-Daero, Sujeong-Gu, Seongnam, Gyeonggi 13120 Republic of Korea
| |
Collapse
|
20
|
Regulation of P-glycoprotein and Breast Cancer Resistance Protein Expression Induced by Focused Ultrasound-Mediated Blood-Brain Barrier Disruption: A Pilot Study. Int J Mol Sci 2022; 23:ijms232415488. [PMID: 36555129 PMCID: PMC9779754 DOI: 10.3390/ijms232415488] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) controls brain homeostasis; it is formed by vascular endothelial cells that are physically connected by tight junctions (TJs). The BBB expresses efflux transporters such as P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), which limit the passage of substrate molecules from blood circulation to the brain. Focused ultrasound (FUS) with microbubbles can create a local and reversible detachment of the TJs. However, very little is known about the effect of FUS on the expression of efflux transporters. We investigated the in vivo effects of moderate acoustic pressures on both P-gp and BCRP expression for up to two weeks after sonication. Magnetic resonance-guided FUS was applied in the striatum of 12 rats. P-gp and BCRP expression were determined by immunohistochemistry at 1, 3, 7, and 14 days postFUS. Our results indicate that FUS-induced BBB opening is capable of (i) decreasing P-gp expression up to 3 days after sonication in both the treated and in the contralateral brain regions and is capable of (ii) overexpressing BCRP up to 7 days after FUS in the sonicated regions only. Our findings may help improve FUS-aided drug delivery strategies by considering both the mechanical effect on the TJs and the regulation of P-gp and BCRP.
Collapse
|
21
|
Wang J, Li Z, Pan M, Fiaz M, Hao Y, Yan Y, Sun L, Yan F. Ultrasound-mediated blood-brain barrier opening: An effective drug delivery system for theranostics of brain diseases. Adv Drug Deliv Rev 2022; 190:114539. [PMID: 36116720 DOI: 10.1016/j.addr.2022.114539] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Blood-brain barrier (BBB) remains a significant obstacle to drug therapy for brain diseases. Focused ultrasound (FUS) combined with microbubbles (MBs) can locally and transiently open the BBB, providing a potential strategy for drug delivery across the BBB into the brain. Nowadays, taking advantage of this technology, many therapeutic agents, such as antibodies, growth factors, and nanomedicine formulations, are intensively investigated across the BBB into specific brain regions for the treatment of various brain diseases. Several preliminary clinical trials also have demonstrated its safety and good tolerance in patients. This review gives an overview of the basic mechanisms, ultrasound contrast agents, evaluation or monitoring methods, and medical applications of FUS-mediated BBB opening in glioblastoma, Alzheimer's disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Jieqiong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 201206, China
| | - Zhenzhou Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, China
| | - Min Pan
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Muhammad Fiaz
- Department of Radiology, Azra Naheed Medical College, Lahore, Pakistan
| | - Yongsheng Hao
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yiran Yan
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
22
|
Singh A, Kusunose J, Phipps MA, Wang F, Chen LM, Caskey CF. Guiding and monitoring focused ultrasound mediated blood-brain barrier opening in rats using power Doppler imaging and passive acoustic mapping. Sci Rep 2022; 12:14758. [PMID: 36042266 PMCID: PMC9427847 DOI: 10.1038/s41598-022-18328-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
The blood-brain barrier (BBB) prevents harmful toxins from entering brain but can also inhibit therapeutic molecules designed to treat neurodegenerative diseases. Focused ultrasound (FUS) combined with microbubbles can enhance permeability of BBB and is often performed under MRI guidance. We present an all-ultrasound system capable of targeting desired regions to open BBB with millimeter-scale accuracy in two dimensions based on Doppler images. We registered imaging coordinates to FUS coordinates with target registration error of 0.6 ± 0.3 mm and used the system to target microbubbles flowing in cellulose tube in two in vitro scenarios (agarose-embedded and through a rat skull), while receiving echoes on imaging transducer. We created passive acoustic maps from received echoes and found error between intended location in imaging plane and location of pixel with maximum intensity after passive acoustic maps reconstruction to be within 2 mm in 5/6 cases. We validated ultrasound-guided procedure in three in vivo rat brains by delivering MRI contrast agent to cortical regions of rat brains after BBB opening. Landmark-based registration of vascular maps created with MRI and Doppler ultrasound revealed BBB opening inside the intended focus with targeting accuracy within 1.5 mm. Combined use of power Doppler imaging with passive acoustic mapping demonstrates an ultrasound-based solution to guide focused ultrasound with high precision in rodents.
Collapse
Affiliation(s)
- Aparna Singh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Jiro Kusunose
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - M Anthony Phipps
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Feng Wang
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Li Min Chen
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Charles F Caskey
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA.
| |
Collapse
|
23
|
Shen Y, Hu M, Li W, Chen Y, Xu Y, Sun L, Liu D, Chen S, Gu Y, Ma Y, Chen X. Delivery of DNA octahedra enhanced by focused ultrasound with microbubbles for glioma therapy. J Control Release 2022; 350:158-174. [PMID: 35981634 DOI: 10.1016/j.jconrel.2022.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/18/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
DNA nanostructures, with good biosafety, highly programmable assembly, flexible modification, and precise control, are tailored as drug carriers to deliver therapeutic agents for cancer therapy. However, they face considerable challenges regarding their delivery into the brain, mainly due to the blood-brain barrier (BBB). By controlling the acoustic parameters, focused ultrasound combined with microbubbles (FUS/MB) can temporarily, noninvasively, and reproducibly open the BBB in a localized region. We investigated the delivery outcome of pH-responsive DNA octahedra loading Epirubicin (Epr@DNA-Octa) via FUS/MB and its therapeutic efficiency in a mouse model bearing intracranial glioma xenograft. Using FUS/MB to locally disrupt the BBB or the blood-tumor barrier (BTB) and systemic administration of Epr@DNA-Octa (Epr@DNA-Octa + FUS/MB) (2 mg/kg of loaded Epr), we achieved an Epr concentration of 292.3 ± 10.1 ng/g tissue in glioma, a 4.4-fold increase compared to unsonicated animals (p < 0.001). The in vitro findings indicated that Epr released from DNA strands accumulated in lysosomes and induced enhanced cytotoxicity compared to free Epr. Further two-photon intravital imaging of spatiotemporal patterns of the DNA-Octa leakage revealed that the FUS/MB treatment enhanced DNA-Octa delivery across several physiological barriers at microscopic level, including the first extravasation across the BBB/BTB and then deep penetration into the glioma center and engulfment of DNA-Octa into the tumor cell body. Longitudinal in vivo bioluminescence imaging and histological analysis indicated that the intracranial glioma progression in nude mice treated with Epr@DNA-Octa + FUS/MB was effectively retarded compared to other groups. The beneficial effect on survival was most significant in the Epr@DNA-Octa + FUS/MB group, with a 50% increase in median survival and a 73% increase in the maximum survival compared to control animals. Our work demonstrates the potential viability of FUS/MB as an alternative strategy for glioma delivery of anticancer drugs using DNA nanostructures as the drug delivery platform for brain cancer therapy.
Collapse
Affiliation(s)
- Yuanyuan Shen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518071, China
| | - Mengni Hu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518071, China
| | - Wen Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yiling Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518071, China
| | - Yiluo Xu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518071, China
| | - Litao Sun
- Department of Ultrasound, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China
| | - Dongzhe Liu
- Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen 518071, China
| | - Siping Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518071, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Ma
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Xin Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518071, China.
| |
Collapse
|
24
|
Thies M, Oelze ML. Combined Therapy Planning, Real-Time Monitoring, and Low Intensity Focused Ultrasound Treatment Using a Diagnostic Imaging Array. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:1410-1419. [PMID: 34986094 PMCID: PMC9199060 DOI: 10.1109/tmi.2021.3140176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Low intensity focused ultrasound (FUS) therapies use low intensity focused ultrasound waves, typically in combination with microbubbles, to non-invasively induce a variety of therapeutic effects. FUS therapies require pre-therapy planning and real-time monitoring during treatment to ensure the FUS beam is correctly targeted to the desired tissue region. To facilitate more streamlined FUS treatments, we present a system for pre-therapy planning, real-time FUS beam visualization, and low intensity FUS treatment using a single diagnostic imaging array. Therapy planning was accomplished by manually segmenting a B-mode image captured by the imaging array and calculating a sonication pattern for the treatment based on the user-input region of interest. For real-time monitoring, the imaging array transmitted a visualization pulse which was focused to the same location as the FUS therapy beam and ultrasonic backscatter from this pulse was used to reconstruct the intensity field of the FUS beam. The therapy planning and beam monitoring techniques were demonstrated in a tissue-mimicking phantom and in a rat tumor in vivo while a mock FUS treatment was carried out. The FUS pulse from the imaging array was excited with an MI of 0.78, which suggests that the array could be used to administer select low intensity FUS treatments involving microbubble activation.
Collapse
|
25
|
Fan X, Gu L, Lv S, Zhang M, Zhuang L, Zhang Y, Chen P. Suppression of the transforming growth factor-β signaling pathway produces a synergistic effect of combination therapy with programmed death receptor 1 blockade and radiofrequency ablation against hepatic carcinoma in mice. Bioengineered 2022; 13:9046-9058. [PMID: 35354382 PMCID: PMC9162004 DOI: 10.1080/21655979.2022.2051688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/19/2022] Open
Abstract
Primary liver cancer (PLC) significantly affects the health of patients globally owing to its high morbidity and low survival rate. Radiofrequency ablation (RFA) has recently been introduced for the clinical treatment of PLC. However, significant immunosuppressive effects are induced by RFA, which limits its application. This study aimed to explore the potential of combination therapy with RFA by investigating the effects of siRNAs against programmed death receptor 1 (PD-1) and transforming growth factor-β (TGF-β) on the antitumor effect induced by RFA. We observed that compared with si-NC, cell viability was reduced, apoptosis rate was elevated, release of inflammatory factors and percentage of CD3+CD8+ cells were increased, and the PI3K/AKT/mTOR pathway was repressed in the co-culture of RFA-treated H22 cells and CD8+ T cells by transfection with si-PD-1 and si-TGF-β; these effects were further enhanced by co-transfection with si-PD-1 and si-TGF-β. Additionally, in H22 cell xenograft-bearing mice treated with RFA, compared with the si-NC group, repressed tumor growth, prolonged survival, increased production of inflammatory factors and expression of CD3 and CD8 in tumor tissues, and downregulation of the PI3K/AKT/mTOR pathway were observed in the si-PD-1 and si-TGF-β groups; these effects were further enhanced in the si-PD-1 + si-TGF-β group. Taken together, our data revealed that suppression of the TGF-β signaling pathway produced a synergistic antitumor effect of combination therapy with PD-1 blockade and RFA against PLC. [Figure: see text].
Collapse
Affiliation(s)
- Xiaoxiang Fan
- Department of Interventional Therapy, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Lihu Gu
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
- Department of General Surgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Shuyi Lv
- Department of Interventional Therapy, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Meiwu Zhang
- Department of Interventional Therapy, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Luhui Zhuang
- Department of Interventional Therapy, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Yan Zhang
- Department of Interventional Therapy, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
| | - Ping Chen
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, Zhejiang, China
- Department of General Surgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Schoen S, Kilinc MS, Lee H, Guo Y, Degertekin FL, Woodworth GF, Arvanitis C. Towards controlled drug delivery in brain tumors with microbubble-enhanced focused ultrasound. Adv Drug Deliv Rev 2022; 180:114043. [PMID: 34801617 PMCID: PMC8724442 DOI: 10.1016/j.addr.2021.114043] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/27/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
Brain tumors are particularly challenging malignancies, due to their location in a structurally and functionally distinct part of the human body - the central nervous system (CNS). The CNS is separated and protected by a unique system of brain and blood vessel cells which together prevent most bloodborne therapeutics from entering the brain tumor microenvironment (TME). Recently, great strides have been made through microbubble (MB) ultrasound contrast agents in conjunction with ultrasound energy to locally increase the permeability of brain vessels and modulate the brain TME. As we elaborate in this review, this physical method can effectively deliver a wide range of anticancer agents, including chemotherapeutics, antibodies, and nanoparticle drug conjugates across a range of preclinical brain tumors, including high grade glioma (glioblastoma), diffuse intrinsic pontine gliomas, and brain metastasis. Moreover, recent evidence suggests that this technology can promote the effective delivery of novel immunotherapeutic agents, including immune check-point inhibitors and chimeric antigen receptor T cells, among others. With early clinical studies demonstrating safety, and several Phase I/II trials testing the preclinical findings underway, this technology is making firm steps towards shaping the future treatments of primary and metastatic brain cancer. By elaborating on its key components, including ultrasound systems and MB technology, along with methods for closed-loop spatial and temporal control of MB activity, we highlight how this technology can be tuned to enable new, personalized treatment strategies for primary brain malignancies and brain metastases.
Collapse
Affiliation(s)
- Scott Schoen
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - M. Sait Kilinc
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Hohyun Lee
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yutong Guo
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - F. Levent Degertekin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Graeme F. Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA,Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, College Park, MD 20742, USA,Fischell Department of Bioengineering A. James Clarke School of Engineering, University of Maryland
| | - Costas Arvanitis
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA,Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| |
Collapse
|
30
|
Song R, Zhang C, Teng F, Tu J, Guo X, Fan Z, Zheng Y, Zhang D. Cavitation-facilitated transmembrane permeability enhancement induced by acoustically vaporized nanodroplets. ULTRASONICS SONOCHEMISTRY 2021; 79:105790. [PMID: 34662804 PMCID: PMC8526759 DOI: 10.1016/j.ultsonch.2021.105790] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 05/05/2023]
Abstract
Ultrasound-facilitated transmembrane permeability enhancement has attracted broad attention in the treatment of central nervous system (CNS) diseases, by delivering gene/drugs into the deep site of brain tissues with a safer and more effective way. Although the feasibility of using acoustically vaporized nanodroplets to open the blood-brain-barrier (BBB) has previously been reported, the relevant physical mechanisms and impact factors are not well known. In the current study, a nitrocellulose (NC) membrane was used to mimic the multi-layered pore structure of BBB. The cavitation activity and the penetration ability of phase-changed nanodroplets were systemically evaluated at different concentration levels, and compared with the results obtained for SonoVue microbubbles. Passive cavitation detection showed that less intensified but more sustained inertial cavitation (IC) activity would be generated by vaporized nanodroplets than microbubbles. As the results, with a sufficiently high concentration (∼5 × 108/mL), phase-changed nanodroplets were more effective than microbubbles in enabling a fluorescent tracer agent (FITC, 150 kDa) to penetrate deeper and more homogeneously through the NC membrane, and a positive correlation was observed between accumulated IC dose and the amount of penetrated FITC. In vivo studies further confirmed acoustically vaporized nanodroplets performed better than microbubbles by opening the BBB in rats' brains. These results indicated that phase-changed nanodroplets can be used as a safe, efficient and durable agent to achieve satisfactory cavitation-mediated permeability enhancement effect in biomedical applications.
Collapse
Affiliation(s)
- Renjie Song
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Chunbing Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fengmeng Teng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China.
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Zheng Fan
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Yinfei Zheng
- Research Center for Intelligent Sensing, Zhejiang Lab, Hangzhou 311100, China.
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China.
| |
Collapse
|
31
|
Wang F, Dong L, Wei X, Wang Y, Chang L, Wu H, Liu S, Chang Y, Yin Y, Luo X, Jia X, Yan F, Li N. Effect of Gambogic Acid-Loaded Porous-Lipid/PLGA Microbubbles in Combination With Ultrasound-Triggered Microbubble Destruction on Human Glioma. Front Bioeng Biotechnol 2021; 9:711787. [PMID: 34604184 PMCID: PMC8479098 DOI: 10.3389/fbioe.2021.711787] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
Gambogic acid (GA) is a highly effective antitumor agent, and it is used for the treatment of a wide range of cancers. It is challenging to deliver drugs to the central nervous system due to the inability of GA to cross the blood-brain barrier (BBB). Studies have shown that ultrasound-targeted microbubble destruction can be used for transient and reversible BBB disruption, significantly facilitating intracerebral drug delivery. We first prepared GA-loaded porous-lipid microbubbles (GA porous-lipid/PLGA MBs), and an in vitro BBB model was established. The cell viability was detected by CCK-8 assay and flow cytometry. The results indicate that U251 human glioma cells were killed by focused ultrasound (FUS) combined with GA/PLGA microbubbles. FUS combined with GA/PLGA microbubbles was capable of locally and transiently enhancing the permeability of BBB under certain conditions. This conformational change allows the release of GA to extracellular space. This study provides novel targets for the treatment of glioma.
Collapse
Affiliation(s)
- Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lei Dong
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xixi Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yongling Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Liansheng Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hongwei Wu
- Department of Chemistry, Xinxiang Medical University, Xinxiang, China
| | - Shuyuan Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China
| | - Yuqiao Chang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaoqiu Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaojian Jia
- Shenzhen Kangning Hospital and Shenzhen Mental Health Center, Shenzhen, China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
32
|
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
|
33
|
Grygorczyk R, Boudreault F, Ponomarchuk O, Tan JJ, Furuya K, Goldgewicht J, Kenfack FD, Yu F. Lytic Release of Cellular ATP: Physiological Relevance and Therapeutic Applications. Life (Basel) 2021; 11:life11070700. [PMID: 34357072 PMCID: PMC8307140 DOI: 10.3390/life11070700] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 07/13/2021] [Indexed: 01/01/2023] Open
Abstract
The lytic release of ATP due to cell and tissue injury constitutes an important source of extracellular nucleotides and may have physiological and pathophysiological roles by triggering purinergic signalling pathways. In the lungs, extracellular ATP can have protective effects by stimulating surfactant and mucus secretion. However, excessive extracellular ATP levels, such as observed in ventilator-induced lung injury, act as a danger-associated signal that activates NLRP3 inflammasome contributing to lung damage. Here, we discuss examples of lytic release that we have identified in our studies using real-time luciferin-luciferase luminescence imaging of extracellular ATP. In alveolar A549 cells, hypotonic shock-induced ATP release shows rapid lytic and slow-rising non-lytic components. Lytic release originates from the lysis of single fragile cells that could be seen as distinct spikes of ATP-dependent luminescence, but under physiological conditions, its contribution is minimal <1% of total release. By contrast, ATP release from red blood cells results primarily from hemolysis, a physiological mechanism contributing to the regulation of local blood flow in response to tissue hypoxia, mechanical stimulation and temperature changes. Lytic release of cellular ATP may have therapeutic applications, as exemplified by the use of ultrasound and microbubble-stimulated release for enhancing cancer immunotherapy in vivo.
Collapse
Affiliation(s)
- Ryszard Grygorczyk
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
- Département de Médecine, Université de Montréal, Montréal, QC H2X 0A9, Canada
- Correspondence: (R.G.); (F.Y.)
| | - Francis Boudreault
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
| | - Olga Ponomarchuk
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
| | - Ju Jing Tan
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
| | - Kishio Furuya
- Graduate School of Medicine, Nagoya University, Nagoya 464-8601, Japan;
| | - Joseph Goldgewicht
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
| | - Falonne Démèze Kenfack
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
| | - François Yu
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada; (F.B.); (O.P.); (J.J.T.); (J.G.); (F.D.K.)
- Département de Radiologie, Radio-Oncologie et Médecine Nucléaire, Université de Montréal, Montréal, QC H2X 0A9, Canada
- Institut de Génie Biomédical, Université de Montréal, Montréal, QC H2X 0A9, Canada
- Correspondence: (R.G.); (F.Y.)
| |
Collapse
|
34
|
Quantitative analysis of in-vivo microbubble distribution in the human brain. Sci Rep 2021; 11:11797. [PMID: 34083642 PMCID: PMC8175375 DOI: 10.1038/s41598-021-91252-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 05/21/2021] [Indexed: 02/04/2023] Open
Abstract
Microbubbles (MB) are widely used as contrast agents to perform contrast-enhanced ultrasound (CEUS) imaging and as acoustic amplifiers of mechanical bioeffects incited by therapeutic-level ultrasound. The distribution of MBs in the brain is not yet fully understood, thereby limiting intra-operative CEUS guidance or MB-based FUS treatments. In this paper we describe a robust platform for quantification of MB distribution in the human brain, allowing to quantitatively discriminate between tumoral and normal brain tissues and we provide new information regarding real-time cerebral MBs distribution. Intraoperative CEUS imaging was performed during surgical tumor resection using an ultrasound machine (MyLab Twice, Esaote, Italy) equipped with a multifrequency (3-11 MHz) linear array probe (LA332) and a specific low mechanical index (MI < 0.4) CEUS algorithm (CnTi, Esaote, Italy; section thickness, 0.245 cm) for non-destructive continuous MBs imaging. CEUS acquisition is started by enabling the CnTI PEN-M algorithm automatically setting the MI at 0.4 with a center frequency of 2.94 MHz-10 Hz frame rate at 80 mm-allowing for continuous non-destructive MBs imaging. 19 ultrasound image sets of adequate length were selected and retrospectively analyzed using a custom image processing software for quantitative analysis of echo power. Regions of interest (ROIs) were drawn on key structures (artery-tumor-white matter) by a blinded neurosurgeon, following which peak enhancement and time intensity curves (TICs) were quantified. CEUS images revealed clear qualitative differences in MB distribution: arteries showed the earliest and highest enhancement among all structures, followed by tumor and white matter regions, respectively. The custom software built for quantitative analysis effectively captured these differences. Quantified peak intensities showed regions containing artery, tumor or white matter structures having an average MB intensity of 0.584, 0.436 and 0.175 units, respectively. Moreover, the normalized area under TICs revealed the time of flight for MB to be significantly lower in brain tissue as compared with tumor tissue. Significant heterogeneities in TICs were also observed within different regions of the same brain lesion. In this study, we provide the most comprehensive strategy for accurate quantitative analysis of MBs distribution in the human brain by means of CEUS intraoperative imaging. Furthermore our results demonstrate that CEUS imaging quantitative analysis enables discernment between different types of brain tumors as well as regions and structures within the brain. Similar considerations will be important for the planning and implementation of MB-based imaging or treatments in the future.
Collapse
|
35
|
Zhou H, Liu Y, Long X, Qiao Y, Lee J, Liu X, Zheng H, Zou C. MR-guided blood-brain barrier opening induced by rapid short-pulse ultrasound in non-human primates. Quant Imaging Med Surg 2021; 11:2415-2427. [PMID: 34079712 DOI: 10.21037/qims-20-1047] [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] [Indexed: 12/24/2022]
Abstract
Background Opening the blood-brain barrier (BBB) with focused ultrasound and microbubbles (MBs) has potential use in non-invasive targeted therapy for central nervous system (CNS) diseases. Rapid short-pulse (RaSP) ultrasound with a microsecond sequence has been proposed as a minimally disruptive and efficient method for opening the BBB. This work aimed to test the feasibility and safety of BBB opening in a non-human primate model using combined RaSP ultrasound sequence and MBs. Methods The BBB of 2 rhesus macaques were opened with RaSP and the commonly used 10 millisecond long pulse (LP), combined with microbubble (SonoVueTM, 0.2 µL/g) injection in a bolus. The transducer's central frequency was 300 kHz, and the acoustic pressure was set to 0.56 MPa calibrated in water. The BBB opening procedure was guided and evaluated with contrast-enhanced magnetic resonance imaging. The relative signal enhancement was compared between RaSP and LP sonication. T2-weighted fast-spin echo (FSE) and T2*-weighted gradient echo (GRE) sequences were scanned to evaluate edema and micro-bleeding at the end of the procedure. Results The relative signal enhancement was significantly higher (P<0.01) in the focal area compared to a similar area of the opposite hemisphere at all time points after sonication in each monkey, indicating the successful opening of the BBB. The relative signal enhancement in RaSP reached more than 60% of that with LP in our experiment, while the energy deposition was only 6% of LP. No edema or hemorrhage was found on magnetic resonance images after RaSP. Conclusions Combined RaSP ultrasound and MBs for the BBB opening is a practical method in large animal models.
Collapse
Affiliation(s)
- Hui Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,The Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, China
| | - Yang Liu
- Research Center for Medical AI, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaojing Long
- Research Center for Medical AI, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yangzi Qiao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jo Lee
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chao Zou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen, China
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
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
|
39
|
Sujarittam K, Choi JJ. Angular dependence of the acoustic signal of a microbubble cloud. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2020; 148:2958. [PMID: 33261381 DOI: 10.1121/10.0002490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023]
Abstract
Microbubble-mediated ultrasound therapies have a common need for methods that can noninvasively monitor the treatment. One approach is to use the bubbles' acoustic emissions as feedback to the operator or a control unit. Current methods interpret the emissions' frequency content to infer the microbubble activities and predict therapeutic outcomes. However, different studies placed their sensors at different angles relative to the emitter and bubble cloud. Here, it is evaluated whether such angles influence the captured emissions such as the frequency content. In computer simulations, 128 coupled bubbles were sonicated with a 0.5-MHz, 0.35-MPa pulse, and the acoustic emissions generated by the bubbles were captured with two sensors placed at different angles. The simulation was replicated in experiments using a microbubble-filled gel channel (0.5-MHz, 0.19-0.75-MPa pulses). A hydrophone captured the emissions at two different angles. In both the simulation and the experiments, one angle captured periodic time-domain signals, which had high contributions from the first three harmonics. In contrast, the other angle captured visually aperiodic time-domain features, which had much higher harmonic and broadband content. Thus, by placing acoustic sensors at different positions, substantially different acoustic emissions were captured, potentially leading to very different conclusions about the treatment outcome.
Collapse
Affiliation(s)
- Krit Sujarittam
- Department of Bioengineering, Imperial College London, 2 Imperial College Road, South Kensington, London, SW7 2AZ, United Kingdom
| | - James J Choi
- Department of Bioengineering, Imperial College London, 2 Imperial College Road, South Kensington, London, SW7 2AZ, United Kingdom
| |
Collapse
|
40
|
Dong W, Huang A, Huang J, Wu P, Guo S, Liu H, Qin M, Yang X, Zhang B, Wan M, Zong Y. Plasmid-loadable magnetic/ultrasound-responsive nanodroplets with a SPIO-NP dispersed perfluoropentane core and lipid shell for tumor-targeted intracellular plasmid delivery. Biomater Sci 2020; 8:5329-5345. [PMID: 32793943 DOI: 10.1039/d0bm00699h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Using ultrasound activating contrast agents to induce sonoporation is a potential strategy for effective lesion-targeted gene delivery. Previous reports have proven that submicron nanodroplets have a better advantage than microbubbles in that they can pass through tumor vasculature endothelial gaps by passive targeting; however, they cannot achieve an adequate dose in tumors to facilitate ultrasound-enhanced gene delivery. Additionally, a few studies focused on delivering macromolecular genetic materials (i.e. overexpression plasmid and CRISPR plasmid) have presented more unique advantages than small-molecular genetic materials (i.e. miRNA mimics, siRNA and shRNA etc.), such as enhancing the expression of target genes with long-term effectiveness. Thereby, we constructed novel plasmid-loadable magnetic/ultrasound-responsive nanodroplets, where superparamagnetic iron oxide nanoparticle dispersed perfluoropentane was encapsulated with lipids to which plasmids could be adhered, and branched polyethylenimine was used to protect the plasmids from enzymolysis. Furthermore, in vitro and in vivo studies were performed to verify the magnetic tumor-targeting ability of the plasmid-loadable magnetic/ultrasound-responsive nanodroplets and focused ultrasound enhanced intracellular plasmid delivery. The plasmid-loadable magnetic/ultrasound-responsive nanodroplets, carrying 16-19 plasmids per droplet, had desirable diameters less than 300 nm, and integrated the merits of excellent magnetic targeting capabilities and phase transition sensitivity to focused ultrasound. Under programmable focused ultrasound exposure, the plasmid-loadable magnetic/ultrasound-responsive nanodroplets underwent a phase-transition into echogenic microbubbles and the subsequent inertial cavitation of the microbubbles achieved an ∼40% in vitro plasmid delivery efficiency. Following intravenous administration, T2-weighted magnet resonance imaging, scanning electron microscopy and inductively coupled plasma optical emission spectrometry of the tumors showed significantly enhanced intratumoral accumulation of the plasmid-loadable magnetic/ultrasound-responsive nanodroplets under an external magnetic field. And a GFP ELISA assay and immunofluorescence staining indicated that focused ultrasound-induced inertial cavitation of the plasmid-loadable magnetic/ultrasound-responsive nanodroplets significantly enhanced the intracellular delivery of plasmids within the tumor after magnet-assisted accumulation, while only lower GFP levels were observed in the tumors on applying focused ultrasound or an external magnet alone. Taken together, utilizing the excellent plasmid-loadable magnetic/ultrasound-responsive nanodroplets combined with magnetism and ultrasound could efficiently deliver plasmids to cancer cells, which could be a potential strategy for macromolecular genetic material delivery in the clinic to treat cancer.
Collapse
Affiliation(s)
- Wei Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Shen Y, Hua L, Yeh CK, Shen L, Ying M, Zhang Z, Liu G, Li S, Chen S, Chen X, Yang X. Ultrasound with microbubbles improves memory, ameliorates pathology and modulates hippocampal proteomic changes in a triple transgenic mouse model of Alzheimer's disease. Am J Cancer Res 2020; 10:11794-11819. [PMID: 33052247 PMCID: PMC7546002 DOI: 10.7150/thno.44152] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease manifested by cognitive impairment. As a unique approach to open the blood-brain barrier (BBB) noninvasively and temporarily, a growing number of studies showed that low-intensity focused ultrasound in combination with microbubbles (FUS/MB), in the absence of therapeutic agents, is capable of ameliorating amyloid or tau pathology, concurrent with improving memory deficits of AD animal models. However, the effects of FUS/MB on both the two pathologies simultaneously, as well as the memory behaviors, have not been reported so far. Methods: In this study, female triple transgenic AD (3×Tg-AD) mice at eight months of age with both amyloid-β (Aβ) deposits and tau phosphorylation were treated by repeated FUS/MB in the unilateral hippocampus twice per week for six weeks. The memory behaviors were investigated by the Y maze, the Morris water maze and the step-down passive avoidance test following repeated FUS/MB treatments. Afterwards, the involvement of Aβ and tau pathology were assessed by immunohistochemical analysis. Neuronal health and phagocytosis of Aβ deposits by microglia in the hippocampus were examined by confocal microscopy. Further, hippocampal proteomic alterations were analyzed by employing two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) combined with mass spectrometry. Results: The three independent memory tasks were indicative of evident learning and memory impairments in eight-month-old 3×Tg-AD mice, which developed intraneuronal Aβ, extracellular diffuse Aβ deposits and phosphorylated tau in the hippocampus and amygdala. Following repeated FUS/MB treatments, significant improvement in learning and memory ability of the 3×Tg-AD mice was achieved. Amelioration in both Aβ deposits and phosphorylated tau in the sonicated hemisphere was induced in FUS/MB-treated 3×Tg-AD mice. Albeit without increase in neuron density, enhancement in axonal neurofilaments emerged from the FUS/MB treatment. Confocal microscopy revealed activated microglia engulfing Aβ deposits in the FUS/MB-treated hippocampus. Further, proteomic analysis revealed 20 differentially expressed proteins, associated with glycolysis, neuron projection, mitochondrial pathways, metabolic process and ubiquitin binding etc., in the hippocampus between FUS/MB-treated and sham-treated 3×Tg-AD mice. Conclusions: Our findings reinforce the positive therapeutic effects on AD models with both Aβ and tau pathology induced by FUS/MB-mediated BBB opening, further supporting the potential of this treatment regime for clinical applications.
Collapse
|
42
|
Todd N, McDannold N, Borsook D. Targeted manipulation of pain neural networks: The potential of focused ultrasound for treatment of chronic pain. Neurosci Biobehav Rev 2020; 115:238-250. [PMID: 32534900 PMCID: PMC7483565 DOI: 10.1016/j.neubiorev.2020.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/24/2020] [Accepted: 06/04/2020] [Indexed: 12/29/2022]
Abstract
Focused ultrasound (FUS) is a promising technology for facilitating treatment of brain diseases including chronic pain. Focused ultrasound is a unique modality for delivering therapeutic levels of energy into the body, including the central nervous system (CNS). It is non-invasive and can target spatially localized effects through the intact skull to cortical or subcortical regions of the brain. FUS can achieve three different mechanisms of action in the brain that are relevant for chronic pain treatment: (1) localized thermal ablation of neural tissue; (2) localized and transient disruption of the blood-brain barrier for targeted drug delivery to CNS structures; and (3) inhibition or stimulation of neuronal activity in targeted regions. This review provides an in-depth look at the technology of FUS with emphasis placed on applications to CNS-based treatments of chronic pain. While still in the early stages of clinical translation and with some technical challenges remaining, we suggest that FUS has great potential as a novel approach for manipulating CNS networks involved in pain treatment.
Collapse
Affiliation(s)
- Nick Todd
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Center for Pain and the Brain, 1 Autumn Street, Boston Children's Hospital, Boston, MA, 02115, United States.
| | - Nathan McDannold
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - David Borsook
- Center for Pain and the Brain, 1 Autumn Street, Boston Children's Hospital, Boston, MA, 02115, United States; Department of Anesthesia, Perioperative, and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, United States
| |
Collapse
|
43
|
Wang JB, Di Ianni T, Vyas DB, Huang Z, Park S, Hosseini-Nassab N, Aryal M, Airan RD. Focused Ultrasound for Noninvasive, Focal Pharmacologic Neurointervention. Front Neurosci 2020; 14:675. [PMID: 32760238 PMCID: PMC7372945 DOI: 10.3389/fnins.2020.00675] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
A long-standing goal of translational neuroscience is the ability to noninvasively deliver therapeutic agents to specific brain regions with high spatiotemporal resolution. Focused ultrasound (FUS) is an emerging technology that can noninvasively deliver energy up the order of 1 kW/cm2 with millimeter and millisecond resolution to any point in the human brain with Food and Drug Administration-approved hardware. Although FUS is clinically utilized primarily for focal ablation in conditions such as essential tremor, recent breakthroughs have enabled the use of FUS for drug delivery at lower intensities (i.e., tens of watts per square centimeter) without ablation of the tissue. In this review, we present strategies for image-guided FUS-mediated pharmacologic neurointerventions. First, we discuss blood–brain barrier opening to deliver therapeutic agents of a variety of sizes to the central nervous system. We then describe the use of ultrasound-sensitive nanoparticles to noninvasively deliver small molecules to millimeter-sized structures including superficial cortical regions and deep gray matter regions within the brain without the need for blood–brain barrier opening. We also consider the safety and potential complications of these techniques, with attention to temporal acuity. Finally, we close with a discussion of different methods for mapping the ultrasound field within the brain and describe future avenues of research in ultrasound-targeted drug therapies.
Collapse
Affiliation(s)
- Jeffrey B Wang
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Tommaso Di Ianni
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Daivik B Vyas
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Zhenbo Huang
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Sunmee Park
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Niloufar Hosseini-Nassab
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Muna Aryal
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| | - Raag D Airan
- Neuroradiology Division, Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
44
|
Sun T, Dasgupta A, Zhao Z, Nurunnabi M, Mitragotri S. Physical triggering strategies for drug delivery. Adv Drug Deliv Rev 2020; 158:36-62. [PMID: 32589905 DOI: 10.1016/j.addr.2020.06.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Physically triggered systems hold promise for improving drug delivery by enhancing the controllability of drug accumulation and release, lowering non-specific toxicity, and facilitating clinical translation. Several external physical stimuli including ultrasound, light, electric fields and magnetic fields have been used to control drug delivery and they share some common features such as spatial targeting, spatiotemporal control, and minimal invasiveness. At the same time, they possess several distinctive features in terms of interactions with biological entities and/or the extent of stimulus response. Here, we review the key advances of such systems with a focus on discussing their physical mechanisms, the design rationales, and translational challenges.
Collapse
Affiliation(s)
- Tao Sun
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anshuman Dasgupta
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, TX 79902, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
45
|
A new safety index based on intrapulse monitoring of ultra-harmonic cavitation during ultrasound-induced blood-brain barrier opening procedures. Sci Rep 2020; 10:10088. [PMID: 32572103 PMCID: PMC7308405 DOI: 10.1038/s41598-020-66994-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/29/2020] [Indexed: 11/25/2022] Open
Abstract
Ultrasound-induced blood-brain barrier (BBB) opening using microbubbles is a promising technique for local delivery of therapeutic molecules into the brain. The real-time control of the ultrasound dose delivered through the skull is necessary as the range of pressure for efficient and safe BBB opening is very narrow. Passive cavitation detection (PCD) is a method proposed to monitor the microbubble activity during ultrasound exposure. However, there is still no consensus on a reliable safety indicator able to predict potential damage in the brain. Current approaches for the control of the beam intensity based on PCD employ a full-pulse analysis and may suffer from a lack of sensitivity and poor reaction time. To overcome these limitations, we propose an intra-pulse analysis to monitor the evolution of the frequency content during ultrasound bursts. We hypothesized that the destabilization of microbubbles exposed to a critical level of ultrasound would result in the instantaneous generation of subharmonic and ultra-harmonic components. This specific signature was exploited to define a new sensitive indicator of the safety of the ultrasound protocol. The approach was validated in vivo in rats and non-human primates using a retrospective analysis. Our results demonstrate that intra-pulse monitoring was able to exhibit a sudden appearance of ultra-harmonics during the ultrasound excitation pulse. The repeated detection of such a signature within the excitation pulse was highly correlated with the occurrence of side effects such as hemorrhage and edema. Keeping the acoustic pressure at levels where no such sign of microbubble destabilization occurred resulted in safe BBB openings, as shown by MR images and gross pathology. This new indicator should be more sensitive than conventional full-pulse analysis and can be used to distinguish between potentially harmful and safe ultrasound conditions in the brain with very short reaction time.
Collapse
|
46
|
Fletcher SMP, Choi M, Ogrodnik N, O'Reilly MA. A Porcine Model of Transvertebral Ultrasound and Microbubble-Mediated Blood-Spinal Cord Barrier Opening. Am J Cancer Res 2020; 10:7758-7774. [PMID: 32685018 PMCID: PMC7359082 DOI: 10.7150/thno.46821] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
Blood-spinal cord barrier opening, using focused ultrasound and microbubbles, has the potential to improve drug delivery for the treatment of spinal cord pathologies. Delivering and detecting ultrasound through the spine is a challenge for clinical translation. We have previously developed short burst, phase keying exposures, which can be used in a dual-aperture configuration to address clinical scale targeting challenges. Here we demonstrate the use of these pulses for blood-spinal cord barrier opening, in vivo in pigs. Methods: The spinal cords of Yorkshire pigs (n=8) were targeted through the vertebral laminae, in the lower thoracic to upper lumbar region using focused ultrasound (486 kHz) and microbubbles. Four animals were treated with a combination of pulsed sinusoidal exposures (1.0-4.0 MPa, non-derated) and pulsed short burst, phase keying exposures (1.0-2.0 MPa, non-derated). Four animals were treated using ramped short burst, phase keying exposures (1.8-2.1 MPa, non-derated). A 250 kHz narrowband receiver was used to detect acoustic emissions from microbubbles. Blood-spinal cord barrier opening was assessed by the extravasation of Evans blue dye. Histological analysis of the spinal cords was used to assess tissue damage and excised vertebral samples were used in benchtop experiments. Results: Ramped short burst, phase keying exposures successfully modified the blood-spinal cord barrier at 16/24 targeted locations, as assessed by the extravasation of Evans blue dye. At 4 of these locations, opening was confirmed with minimal adverse effects observed through histology. Transmission measurements through excised vertebrae indicated a mean transmission of (47.0 ± 7.0 %) to the target. Conclusions: This study presents the first evidence of focused ultrasound-induced blood-spinal cord barrier opening in a large animal model, through the intact spine. This represents an important step towards clinical translation.
Collapse
|
47
|
Lin HC, Fan CH, Ho YJ, Yeh CK. Dual-Frequency Chirp Excitation for Passive Cavitation Imaging in the Brain. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2020; 67:1127-1140. [PMID: 31940528 DOI: 10.1109/tuffc.2020.2964786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the main challenges that impede cavitation-mediated imaging in the brain is restricted opening of the blood-brain barrier (BBB) making it difficult to locate cavitating microbubbles (MBs). Passive cavitation imaging (PCI) has received attention due to the possibility of performing real-time monitoring by listening to acoustic cavitation. However, the long excitation pulses associated with PCI degrade its axial resolution. The present study combined a coded excitation technique with a dual-frequency chirp (DFC) excitation method to prevent interference from the nonlinear components of MBs' cavitation. The use of DFC excitation generates a low-frequency (0.4, 0.5, or 0.6 MHz) chirp component as the envelope of the signal-driving MBs' cavitation with a dual-frequency pulse ( ω1 = 1.35 MHz and ω2 = 1.65 MHz, ω1 = 1.3 MHz and ω2 = 1.7 MHz, and ω1 = 1.25 MHz and ω2 = 1.75 MHz). The cavitation of MBs was passively imaged utilizing a chirp component with pulse compression to maintain abundant insonation energy without any reduction in the axial imaging resolution. In vitro experiments showed that the DFC method improved the signal-to-noise ratio by 42.2% and the axial resolution by 4.1-fold compared with using a conventional long-pulse waveform. Furthermore, the cavitating MBs driven by different ultrasound (US) energy (0, 0.3, 0.6, and 0.9 MPa, N = 3 for each group) in the rat brain with an intact skull still could be mapped by DFC. Our successful demonstration of using the DFC method to image cavitation-induced BBB opening affords an alternative tool for assessing cavitation-dependent drug delivery to the brain, with the benefit of real-time and high convenient integration with current US imaging devices.
Collapse
|
48
|
Blood-brain barrier disruption and delivery of irinotecan in a rat model using a clinical transcranial MRI-guided focused ultrasound system. Sci Rep 2020; 10:8766. [PMID: 32472017 PMCID: PMC7260193 DOI: 10.1038/s41598-020-65617-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/17/2020] [Indexed: 11/09/2022] Open
Abstract
We investigated controlled blood-brain barrier (BBB) disruption using a low-frequency clinical transcranial MRI-guided focused ultrasound (TcMRgFUS) device and evaluated enhanced delivery of irinotecan chemotherapy to the brain and a rat glioma model. Animals received three weekly sessions of FUS, FUS and 10 mg/kg irinotecan, or irinotecan alone. In each session, four volumetric sonications targeted 36 locations in one hemisphere. With feedback control based on recordings of acoustic emissions, 98% of the sonication targets (1045/1071) reached a pre-defined level of acoustic emission, while the probability of wideband emission (a signature for inertial cavitation) was than 1%. BBB disruption, evaluated by mapping the R1 relaxation rate after administration of an MRI contrast agent, was significantly higher in the sonicated hemisphere (P < 0.01). Histological evaluation found minimal tissue effects. Irinotecan concentrations in the brain were significantly higher (P < 0.001) with BBB disruption, but SN-38 was only detected in <50% of the samples and only with an excessive irinotecan dose. Irinotecan with BBB disruption did not impede tumor growth or increase survival. Overall these results demonstrate safe and controlled BBB disruption with a low-frequency clinical TcMRgFUS device. While irinotecan delivery to the brain was not neurotoxic, it did not improve outcomes in the F98 glioma model.
Collapse
|
49
|
Pouliopoulos AN, Jimenez DA, Frank A, Robertson A, Zhang L, Kline-Schoder AR, Bhaskar V, Harpale M, Caso E, Papapanou N, Anderson R, Li R, Konofagou EE. Temporal stability of lipid-shelled microbubbles during acoustically-mediated blood-brain barrier opening. FRONTIERS IN PHYSICS 2020; 8:137. [PMID: 32457896 PMCID: PMC7250395 DOI: 10.3389/fphy.2020.00137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Non-invasive blood-brain barrier (BBB) opening using focused ultrasound (FUS) is being tested as a means to locally deliver drugs into the brain. Such FUS therapies require injection of preformed microbubbles, currently used as contrast agents in ultrasound imaging. Although their behavior during exposure to imaging sequences has been well described, our understanding of microbubble stability within a therapeutic field is still not complete. Here, we study the temporal stability of lipid-shelled microbubbles during therapeutic FUS exposure in two timescales: the short time scale (i.e., μs of low-frequency ultrasound exposure) and the long time scale (i.e., days post-activation). We first simulated the microbubble response to low-frequency sonication, and found a strong correlation between viscosity and fragmentation pressure. Activated microbubbles had a concentration decay constant of 0.02 d-1 but maintained a quasi-stable size distribution for up to 3 weeks (< 10% variation). Microbubbles flowing through a 4-mm vessel within a tissue-mimicking phantom (5% gelatin) were exposed to therapeutic pulses (fc: 0.5 MHz, peak-negative pressure: 300 kPa, pulse length: 1 ms, pulse repetition frequency: 1 Hz, n=10). We recorded and analyzed their acoustic emissions, focusing on emitted energy and its temporal evolution, alongside the frequency content. Measurements were repeated with concentration-matched samples (107 microbubbles/ml) on day 0, 7, 14, and 21 after activation. Temporal stability decreased while inertial cavitation response increased with storage time both in vitro and in vivo, possibly due to changes in the shell lipid content. Using the same parameters and timepoints, we performed BBB opening in a mouse model (n=3). BBB opening volume measured through T1-weighted contrast-enhanced MRI was equal to 19.1 ± 7.1 mm3, 21.8 ± 14 mm3, 29.3 ± 2.5 mm3, and 38 ± 20.1 mm3 on day 0, 7, 14, and 21, respectively, showing no significant difference over time (p-value: 0.49). Contrast enhancement was 24.9 ± 1.7 %, 23.7 ± 11.7 %, 28.9 ± 5.3 %, and 35 ± 13.4 %, respectively (p-value: 0.63). In conclusion, the in-house made microbubbles studied here maintain their capacity to produce similar therapeutic effects over a period of 3 weeks after activation, as long as the natural concentration decay is accounted for. Future work should focus on stability of commercially available microbubbles and tailoring microbubble shell properties towards therapeutic applications.
Collapse
Affiliation(s)
| | - Daniella A. Jimenez
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Alexander Frank
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Alexander Robertson
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Lin Zhang
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Alina R. Kline-Schoder
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Vividha Bhaskar
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Mitra Harpale
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Elizabeth Caso
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Nicholas Papapanou
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Rachel Anderson
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Rachel Li
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
| | - Elisa E. Konofagou
- Department of Biomedical Engineering, Columbia University, New York City, New York 10032, USA
- Department of Radiology, Columbia University, New York City, New York 10032, USA
- Correspondence: Elisa E. Konofagou 351 Engineering Terrace, 1210 Amsterdam Avenue Mail Code: 8904, New York, NY, USA 10027 Phone: 212-342-0863, 212-854-9661
| |
Collapse
|
50
|
Valdez MA, Fernandez E, Matsunaga T, Erickson RP, Trouard TP. Distribution and Diffusion of Macromolecule Delivery to the Brain via Focused Ultrasound using Magnetic Resonance and Multispectral Fluorescence Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:122-136. [PMID: 31585767 PMCID: PMC6937597 DOI: 10.1016/j.ultrasmedbio.2019.08.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/22/2019] [Accepted: 08/30/2019] [Indexed: 05/15/2023]
Abstract
Focused ultrasound (FUS), in combination with microbubble contrast agents, can be used to transiently open the blood-brain barrier (BBB) to allow intravascular agents to cross into the brain. Often, FUS is carried out in conjunction with magnetic resonance imaging (MRI) to evaluate BBB opening to gadolinium-based MRI contrast agents. Although MRI allows direct visualization of the distribution of gadolinium-based contrast agents in the brain parenchyma, it does not allow measurements of the distribution of other molecules crossing the BBB. Therapeutic molecules (e.g., monoclonal antibodies) are much different in size than MRI contrast agents and have been found to have different distributions in the brain after FUS-mediated BBB opening. In the work described here, we combined in vivo MRI and ex vivo multispectral fluorescence imaging to compare the distributions of MRI contrast and dextran molecules of different molecular weights (3, 70 and 500 kDa) after FUS-mediated BBB opening through a range of ultrasound pressures (0.18-0.46 MPa) in laboratory mice. The volume of brain exposed was calculated from the MRI and fluorescence images and was significantly dependent on both molecular weight and ultrasound pressure. Diffusion coefficients of the different-molecular-weight dextran molecules in the brain parenchyma were also calculated from the fluorescence images and were negatively correlated with the molecular weight of the dextran molecules. The results of this work build on a body of knowledge that is critically important for the FUS technique to be used in clinical delivery of therapeutics to the brain.
Collapse
Affiliation(s)
- Michael A Valdez
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Elizabeth Fernandez
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Terry Matsunaga
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA; Department of Medical Imaging, University of Arizona, Tucson, Arizona, USA
| | - Robert P Erickson
- Department of Pediatrics, University of Arizona, Tucson, Arizona, USA; BIO5 Research Institute, University of Arizona, Tucson, Arizona, USA
| | - Theodore P Trouard
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA; Department of Medical Imaging, University of Arizona, Tucson, Arizona, USA; BIO5 Research Institute, University of Arizona, Tucson, Arizona, USA; Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, Arizona, USA.
| |
Collapse
|