1
|
Seo H, Han M, Choi JR, Kim S, Park J, Lee EH. Numerical Investigation of Layered Homogeneous Skull Model for Simulations of Transcranial Focused Ultrasound. Neuromodulation 2025; 28:103-114. [PMID: 38691075 DOI: 10.1016/j.neurom.2024.04.001] [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: 02/07/2024] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND AND OBJECTIVES The influence of the intracranial pressure field must be discussed with the development of a single-element transducer for low-intensity transcranial focused ultrasound because the skull plays a significant role in blocking and dispersing ultrasound wave propagation. Ultrasound propagation is mainly affected by the structure and acoustic properties of the skull; thus, we aimed to investigate the impact of simplifying the acoustic properties of the skull on the simulation of the transcranial pressure field to present guidance for efficient skull modeling in full-wave simulations. MATERIALS AND METHODS We constructed a three-dimensional computational model for ultrasound transmission with the same structure but varying acoustic properties of the skull. The structural information and heterogeneous acoustic properties of the skull were acquired from computed tomography images, and we segmented the skull into three layers (3 L), including spongy and compact bones. We then assigned homogeneous acoustic properties to a single layer (1 L) or 3 L of the skull. In addition, we investigated the influence of different types of transducers and different ultrasound frequencies (1.1 MHz, 0.5 MHz, and 0.25 MHz) on the intracranial pressure field to provide a comparison of the heterogenous and homogeneous models. RESULTS We indicated the importance of numerical simulations in estimating the intracranial pressure field of the skull owing to beam distortions. When we simplified the skull model, both the 1 L and 3 L models showed contours of the acoustic focus comparable to those of the heterogeneous model. When we evaluated the peak pressure and volume of the acoustic focus, the 1 L model produced a better estimation of peak pressure with a difference <10%, and the 3 L model is suitable to obtain smaller errors in the volume of the acoustic focus. CONCLUSIONS In conclusion, we examined the possibility of simplification of skull models using 1 L and 3 L homogeneous properties in the numerical simulation for focused ultrasound. The results show that the layered homogeneous model can provide characteristics comparable to those of the acoustic focus in heterogeneous models.
Collapse
Affiliation(s)
- Hyeon Seo
- Department of AI Convergence Engineering, Gyeongsang National University, Jinju, Republic of Korea; Department of Computer Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Mun Han
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Korea
| | - Jong-Ryul Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Korea
| | - Seungmin Kim
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Korea
| | - Juyoung Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Korea; Department of High-Tech Medical Device, College of Future Industry, Gachon University, Seongnam, Korea
| | - Eun-Hee Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu, Korea.
| |
Collapse
|
2
|
Seo M, Shin M, Noh G, Yoo SS, Yoon K. Multi-modal networks for real-time monitoring of intracranial acoustic field during transcranial focused ultrasound therapy. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108458. [PMID: 39437458 DOI: 10.1016/j.cmpb.2024.108458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/22/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Transcranial focused ultrasound (tFUS) is an emerging non-invasive therapeutic technology that offers new brain stimulation modality. Precise localization of the acoustic focus to the desired brain target throughout the procedure is needed to ensure the safety and effectiveness of the treatment, but acoustic distortion caused by the skull poses a challenge. Although computational methods can provide the estimated location and shape of the focus, the computation has not reached sufficient speed for real-time inference, which is demanded in real-world clinical situations. Leveraging the advantages of deep learning, we propose multi-modal networks capable of generating intracranial pressure map in real-time. METHODS The dataset consisted of free-field pressure maps, intracranial pressure maps, medical images, and transducer placements was obtained from 11 human subjects. The free-field and intracranial pressure maps were computed using the k-space method. We developed network models based on convolutional neural networks and the Swin Transformer, featuring a multi-modal encoder and a decoder. RESULTS Evaluations on foreseen data achieved high focal volume conformity of approximately 93% for both computed tomography (CT) and magnetic resonance (MR) data. For unforeseen data, the networks achieved the focal volume conformity of 88% for CT and 82% for MR. The inference time of the proposed networks was under 0.02 s, indicating the feasibility for real-time simulation. CONCLUSIONS The results indicate that our networks can effectively and precisely perform real-time simulation of the intracranial pressure map during tFUS applications. Our work will enhance the safety and accuracy of treatments, representing significant progress for low-intensity focused ultrasound (LIFU) therapies.
Collapse
Affiliation(s)
- Minjee Seo
- Yonsei University, School of Mathematics and Computing (Computational Science and Engineering), Seoul, 03722, Republic of Korea
| | - Minwoo Shin
- Yonsei University, School of Mathematics and Computing (Computational Science and Engineering), Seoul, 03722, Republic of Korea
| | - Gunwoo Noh
- Korea University, School of Mechanical Engineering, Seoul, 02841, Republic of Korea
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Kyungho Yoon
- Yonsei University, School of Mathematics and Computing (Computational Science and Engineering), Seoul, 03722, Republic of Korea.
| |
Collapse
|
3
|
Jiang Z, Hua M, Li J, Mau HL, Choi J, Gormley WB, Amich JM, Sha RM. SCOUT: Skull-Corrected Optimization for Ultrasound Transducers. Bioengineering (Basel) 2024; 11:1144. [PMID: 39593804 PMCID: PMC11592240 DOI: 10.3390/bioengineering11111144] [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: 10/22/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Transcranial focused ultrasound has been studied for non-invasive and localized treatment of many brain diseases. The biggest challenge for focusing ultrasound onto the brain is the skull, which attenuates ultrasound and changes its propagation direction, leading to pressure drop, focus shift, and defocusing. We presented an optimization algorithm which automatically found the optimal location for placing a single-element focused transducer. At this optimal location, the focus shift was in an acceptable range and the ultrasound was tightly focused. The algorithm simulated the beam profiles of placing the transducer at different locations and compared the results. Locations with a normalized peak-negative pressure (PNP) above threshold were first found. Then, the optimal location was identified as the location with the smallest focal volume. The optimal location found in this study had a normalized PNP of 0.966 and a focal volume of 6.8% smaller than without the skull. A Zeta navigation system was used to automatically place the transducer and track the error caused by movement. These results demonstrated that the algorithm could find the optimal transducer location to avoid large focus shift and defocusing. With the Zeta navigation system, our algorithm can help to make transcranial focused ultrasound treatment safer and more successful.
Collapse
Affiliation(s)
- Zheng Jiang
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
| | - Michelle Hua
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
| | - Jacqueline Li
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
| | - Hieu Le Mau
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
| | - James Choi
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - William B. Gormley
- Harvard Medical School, Boston, MA 02115, USA;
- Computational Neurosurgical Outcomes Center, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Neurological Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jose M. Amich
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
| | - Raahil M. Sha
- Zeta Surgical Inc., Boston, MA 02111, USA; (Z.J.); (M.H.); (J.L.); (H.L.M.); (J.C.); (J.M.A.)
| |
Collapse
|
4
|
Jimenez-Gambin S, Bae S, Ji R, Tsitsos F, Konofagou EE. Feasibility of Hologram-Assisted Bilateral Blood-Brain Barrier Opening in Non-Human Primates. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1172-1185. [PMID: 39196737 PMCID: PMC11656487 DOI: 10.1109/tuffc.2024.3451289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Focused ultrasound (FUS) and microbubbles facilitate blood-brain barrier opening (BBBO) noninvasively, transiently, and safely for targeted drug delivery. Unlike state-of-the-art approaches, in this study, we demonstrate for the first time the simultaneous, bilateral BBBO in non-human primates (NHPs) using acoustic holograms at caudate and putamen structures. The simple and low-cost system with a single-element FUS transducer and 3-D printed acoustic hologram was guided by neuronavigation and a robotic arm. The advantages of holograms are transcranial aberration correction, simultaneous multifocus and high localization, and target-independent transducer positioning, defining a promising alternative for time- and cost-efficient FUS procedures. Holograms were designed with the k-space method by time-reversal techniques. T1-weighted MRI was used for treatment planning, while the computed tomography (CT) scan provided the head tissues acoustic properties. For the BBBO procedure, a robotic arm allowed transducer positioning errors below 0.1 mm and 0.1°. Following positioning, 0.5-0.6-MPa, 513-kHz microbubble-enhanced FUS was applied for 4 min. For BBBO assessment, Post-FUS T1-weighted MRI was acquired, and contrast enhancement indicated bilateral gadolinium extravasation at both caudate or putamen structures. The two BBBO locations were separated by 13.13 mm with a volume of 91.81 mm3 in the caudate, compared with 9.40 mm with a volume of 124.52 mm3 in simulation, while they were separated by 21.74 mm with a volume of 145.38 mm3 in the putamen and compared with 22.32 mm with a volume of 156.42 mm3 in simulation. No neurological damage was observed through T2-weighted and susceptibility-weighted imaging. This study demonstrates the feasibility and safety of hologram-assisted neuronavigation-guided-FUS for BBBO in NHP, providing thus an avenue for clinical translation.
Collapse
|
5
|
Kusunose J, Rodriguez WJ, Luo H, Manuel TJ, Phipps MA, Yang PF, Grissom WA, Konrad PE, Chen LM, Dawant BM, Caskey CF. Design and Validation of a Patient-Specific Stereotactic Frame for Transcranial Ultrasound Therapy. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1030-1041. [PMID: 39024077 PMCID: PMC11465451 DOI: 10.1109/tuffc.2024.3420242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Transcranial-focused ultrasound (tFUS) procedures such as neuromodulation and blood-brain barrier (BBB) opening require precise focus placement within the brain. MRI is currently the most reliable tool for focus localization but can be prohibitive for procedures requiring recurrent therapies. We designed, fabricated, and characterized a patient-specific, 3-D-printed, stereotactic frame for repeated tFUS therapy. The frame is compact, with minimal footprint, can be removed and re-secured between treatments while maintaining sub-mm accuracy, and will allow for precise and repeatable transcranial FUS treatment without the need for MR-guidance following the initial calibration scan. Focus localization and repeatability were assessed via MR-thermometry and MR-acoustic radiation force imaging (ARFI) on an ex vivo skull phantom and in vivo nonhuman primates (NHPs), respectively. Focal localization, registration, steering, and re-steering were accomplished during the initial MRI calibration scan session. Keeping steering coordinates fixed in subsequent therapy and imaging sessions, we found good agreement between steered foci and the intended target, with target registration error (TRE) of 1.2 ± 0.3 ( n = 4 , ex vivo) and 1.0 ± 0.5 ( n = 3 , in vivo) mm. Focus position (steered and non-steered) was consistent, with sub-mm variation in each dimension between studies. Our 3-D-printed, patient-specific stereotactic frame can reliably position and orient the ultrasound transducer for repeated targeting of brain regions using a single MR-based calibration. The compact frame allows for high-precision tFUS to be carried out outside the magnet and could help reduce the cost of tFUS treatments where repeated application of an ultrasound focus is required with high precision.
Collapse
|
6
|
Choi SW, Komaiha M, Choi D, Lu N, Gerhardson TI, Fox A, Chaudhary N, Camelo-Piragua S, Hall TL, Pandey AS, Xu Z, Sukovich JR. Neuronavigation-Guided Transcranial Histotripsy (NaviTH) System. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1155-1166. [PMID: 38789304 DOI: 10.1016/j.ultrasmedbio.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 05/26/2024]
Abstract
OBJECTIVE The goal of the work described here was to develop the first neuronavigation-guided transcranial histotripsy (NaviTH) system and associated workflow for transcranial ablation. METHODS The NaviTH system consists of a 360-element, 700 kHz transmitter-receiver-capable transcranial histotripsy array, a clinical neuronavigation system and associated equipment for patient-to-array co-registration and therapy planning and targeting software systems. A workflow for NaviTH treatments, including pre-treatment aberration correction, was developed. Targeting errors stemming from target registration errors (TREs) during the patient-to-array co-registration process, as well as focal shifts caused by skull-induced aberrations, were investigated and characterized. The NaviTH system was used in treatments of two <96 h post-mortem human cadavers and in experiments in two excised human skullcaps. RESULTS The NaviTH was successfully used to create ablations in the cadaver brains as confirmed in post-treatment magnetic resonance imaging A total of three ablations were created in the cadaver brains, and targeting errors of 9, 3.4 and 4.4 mm were observed in corpus callosum, septum and thalamus targets, respectively. Errors were found to be caused primarily by TREs resulting from transducer tracking instrument design flaws and imperfections in the treatment workflow. Transducer tracking instrument design and workflow improvements reduced TREs to <2 mm, and skull-induced focal shifts, following pre-treatment aberration correction, were 0.3 mm. Total targeting errors of the NaviTH system following the noted improvements were 2.5 mm. CONCLUSIONS The feasibility of using the first NaviTH system in a human cadaver model has been determined. Although accuracy still needs to be improved, the proposed system has the potential to allow for transcranial histotripsy therapies without requiring active magnetic resonance treatment guidance.
Collapse
Affiliation(s)
- Sang Won Choi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mahmoud Komaiha
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Dave Choi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Ning Lu
- Department of Biomedical Engineering, Stanford University, Stanford, CA, USA
| | - Tyler I Gerhardson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Adam Fox
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Neeraj Chaudhary
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Timothy L Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aditya S Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan R Sukovich
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
López-Aguirre M, Castillo-Ortiz M, Viña-González A, Blesa J, Pineda-Pardo JA. The road ahead to successful BBB opening and drug-delivery with focused ultrasound. J Control Release 2024; 372:901-913. [PMID: 38971426 DOI: 10.1016/j.jconrel.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
This review delves into the innovative technology of Blood-Brain Barrier (BBB) opening with low-intensity focused ultrasound in combination with microbubbles (LIFU-MB), a promising therapeutic modality aimed at enhancing drug delivery to the central nervous system (CNS). The BBB's selective permeability, while crucial for neuroprotection, significantly hampers the efficacy of pharmacological treatments for CNS disorders. LIFU-MB emerges as a non-invasive and localized method to transiently increase BBB permeability, facilitating the delivery of therapeutic molecules. Here, we review the procedural stages of LIFU-MB interventions, including planning and preparation, sonication, evaluation, and delivery, highlighting the technological diversity and methodological challenges encountered in current clinical applications. With an emphasis on safety and efficacy, we discuss the crucial aspects of ultrasound delivery, microbubble administration, acoustic feedback monitoring and assessment of BBB permeability. Finally, we explore the critical choices for effective BBB opening with LIFU-MB, focusing on selecting therapeutic agents, optimizing delivery methods, and timing for delivery. Overcoming existing barriers to integrate this technology into clinical practice could potentially revolutionize CNS drug delivery and treatment paradigms in the near future.
Collapse
Affiliation(s)
- Miguel López-Aguirre
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; PhD Program in Physics, Complutense University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Castillo-Ortiz
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; PhD Program in Technologies for Health and Well-being, Polytechnic University of Valencia, Valencia, Spain; Molecular Imaging Technologies Research Institute (I3M), Polytechnic University of Valencia, Valencia, Spain
| | - Ariel Viña-González
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; PhD Program in Biomedical Engineering, Polytechnic University of Madrid, Madrid, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain; Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain
| | - José A Pineda-Pardo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Spain.
| |
Collapse
|
8
|
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
|
9
|
Antoniou A, Evripidou N, Damianou C. Focused ultrasound heating in brain tissue/skull phantoms with 1 MHz single-element transducer. J Ultrasound 2024; 27:263-274. [PMID: 37517052 PMCID: PMC11178743 DOI: 10.1007/s40477-023-00810-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 07/09/2023] [Indexed: 08/01/2023] Open
Abstract
PURPOSE The study aims to provide insights on the practicality of using single-element transducers for transcranial Focused Ultrasound (tFUS) thermal applications. METHODS FUS sonications were performed through skull phantoms embedding agar-based tissue mimicking gels using a 1 MHz single-element spherically focused transducer. The skull phantoms were 3D printed with Acrylonitrile Butadiene Styrene (ABS) and Resin thermoplastics having the exact skull bone geometry of a healthy volunteer. The temperature field distribution during and after heating was monitored in a 3 T Magnetic Resonance Imaging (MRI) scanner using MR thermometry. The effect of the skull's thickness on intracranial heating was investigated. RESULTS A single FUS sonication at focal acoustic intensities close to 1580 W/cm2 for 60 s in free field heated up the agar phantom to ablative temperatures reaching about 90 °C (baseline of 37 °C). The ABS skull strongly blocked the ultrasonic waves, resulting in zero temperature increase within the phantom. Considerable heating was achieved through the Resin skull, but it remained at hyperthermia levels. Conversely, tFUS through a 1 mm Resin skull showed enhanced ultrasonic penetration and heating, with the focal temperature reaching 70 °C. CONCLUSIONS The ABS skull demonstrated poorer performance in terms of tFUS compared to the Resin skull owing to its higher ultrasonic attenuation and porosity. The thin Resin phantom of 1 mm thickness provided an efficient acoustic window for delivering tFUS and heating up deep phantom areas. The results of such studies could be particularly useful for accelerating the establishment of a wider range of tFUS applications.
Collapse
Affiliation(s)
- Anastasia Antoniou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, 30 Archbishop Kyprianou Street, 3036, Limassol, Cyprus
| | - Nikolas Evripidou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, 30 Archbishop Kyprianou Street, 3036, Limassol, Cyprus
| | - Christakis Damianou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, 30 Archbishop Kyprianou Street, 3036, Limassol, Cyprus.
| |
Collapse
|
10
|
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
|
11
|
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. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.12.21.23300222. [PMID: 38196636 PMCID: PMC10775403 DOI: 10.1101/2023.12.21.23300222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
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
|
12
|
Natera-Villalba E, Ruiz-Yanzi MA, Gasca-Salas C, Matarazzo M, Martínez-Fernández R. MR-guided focused ultrasound in movement disorders and beyond: Lessons learned and new frontiers. Parkinsonism Relat Disord 2024; 122:106040. [PMID: 38378311 DOI: 10.1016/j.parkreldis.2024.106040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
The development of MR-guided focused ultrasound (MRgFUS) has provided a new therapeutic tool for neuropsychiatric disorders. In contrast to previously available neurosurgical techniques, MRgFUS allows precise impact on deep brain structures without the need for incision and yields an immediate effect. In its high-intensity modality (MRgHIFU), it produces accurate therapeutic thermoablation in previously selected brain targets. Importantly, the production of the lesion is progressive and highly controlled in real-time by both neuroimaging and clinical means. MRgHIFU ablation is already an accepted and widely used treatment for medically-refractory Parkinson's disease and essential tremor. Notably, other neurological disorders and diverse brain targets, including bilateral treatments, are currently under examination. Conversely, the low-intensity modality (MRgLIFU) shows promising prospects in neuromodulation and transient blood-brain barrier opening (BBBO). In the former circumstance, MRgLIFU could serve as a powerful clinical and research tool for non-invasively modulating brain activity and function. BBBO, on the other hand, emerges as a potentially impactful method to influence disease pathogenesis and progression by increasing brain target engagement of putative therapeutic agents. While promising, these applications remain experimental. As a recently developed technology, MRgFUS is not without challenges and questions to be addressed. Further developments and broader experience are necessary to enhance MRgFUS capabilities in both research and clinical practice, as well as to define device constraints. This clinical mini-review aims to provide an overview of the main evidence of MRgFUS application and to highlight unmet needs and future potentialities of the technique.
Collapse
Affiliation(s)
- Elena Natera-Villalba
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; PhD Medicine Program, Universidad Autónoma de Madrid, Madrid, Spain
| | - María-Agustina Ruiz-Yanzi
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain
| | - Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain; University CEU-San Pablo, Madrid, Spain
| | - Michele Matarazzo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain
| | - Raúl Martínez-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta Del Sur, HM Hospitales, Madrid, Spain; CIBERNED, Instituto Carlos III, Madrid, Spain; University CEU-San Pablo, Madrid, Spain.
| |
Collapse
|
13
|
Tazhibi M, McQuillan N, Wei HJ, Gallitto M, Bendau E, Webster Carrion A, Berg X, Kokossis D, Zhang X, Zhang Z, Jan CI, Mintz A, Gartrell RD, Syed HR, Fonseca A, Pavisic J, Szalontay L, Konofagou EE, Zacharoulis S, Wu CC. Focused ultrasound-mediated blood-brain barrier opening is safe and feasible with moderately hypofractionated radiotherapy for brainstem diffuse midline glioma. J Transl Med 2024; 22:320. [PMID: 38555449 PMCID: PMC10981822 DOI: 10.1186/s12967-024-05096-9] [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: 03/22/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Diffuse midline glioma (DMG) is a pediatric tumor with dismal prognosis. Systemic strategies have been unsuccessful and radiotherapy (RT) remains the standard-of-care. A central impediment to treatment is the blood-brain barrier (BBB), which precludes drug delivery to the central nervous system (CNS). Focused ultrasound (FUS) with microbubbles can transiently and non-invasively disrupt the BBB to enhance drug delivery. This study aimed to determine the feasibility of brainstem FUS in combination with clinical doses of RT. We hypothesized that FUS-mediated BBB-opening (BBBO) is safe and feasible with 39 Gy RT. METHODS To establish a safety timeline, we administered FUS to the brainstem of non-tumor bearing mice concurrent with or adjuvant to RT; our findings were validated in a syngeneic brainstem murine model of DMG receiving repeated sonication concurrent with RT. The brainstems of male B6 (Cg)-Tyrc-2J/J albino mice were intracranially injected with mouse DMG cells (PDGFB+, H3.3K27M, p53-/-). A clinical RT dose of 39 Gy in 13 fractions (39 Gy/13fx) was delivered using the Small Animal Radiation Research Platform (SARRP) or XRAD-320 irradiator. FUS was administered via a 0.5 MHz transducer, with BBBO and tumor volume monitored by magnetic resonance imaging (MRI). RESULTS FUS-mediated BBBO did not affect cardiorespiratory rate, motor function, or tissue integrity in non-tumor bearing mice receiving RT. Tumor-bearing mice tolerated repeated brainstem BBBO concurrent with RT. 39 Gy/13fx offered local control, though disease progression occurred 3-4 weeks post-RT. CONCLUSION Repeated FUS-mediated BBBO is safe and feasible concurrent with RT. In our syngeneic DMG murine model, progression occurs, serving as an ideal model for future combination testing with RT and FUS-mediated drug delivery.
Collapse
Affiliation(s)
- Masih Tazhibi
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Nicholas McQuillan
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Hong-Jian Wei
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Matthew Gallitto
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Ethan Bendau
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Andrea Webster Carrion
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Xander Berg
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Danae Kokossis
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Xu Zhang
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhiguo Zhang
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Chia-Ing Jan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Akiva Mintz
- Department of Radiology, Columbia University, New York, NY, 10027, USA
| | - Robyn D Gartrell
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Hasan R Syed
- Department of Neurosurgery, Children's National Hospital, Washington, DC, USA
- George Washington University, Washington, DC, USA
| | - Adriana Fonseca
- George Washington University, Washington, DC, USA
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
- The Brain Tumor Institute, Children's National Hospital, Washington, DC, USA
| | - Jovana Pavisic
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Luca Szalontay
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Stergios Zacharoulis
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA.
- Bristol Myers Squibb, Princeton, NJ, 08901, USA.
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA.
| |
Collapse
|
14
|
Aurup C, Bendig J, Blackman SG, McCune EP, Bae S, Jimenez-Gambin S, Ji R, Konofagou EE. Transcranial Functional Ultrasound Imaging Detects Focused Ultrasound Neuromodulation Induced Hemodynamic Changes in Mouse and Nonhuman Primate Brains In Vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.583971. [PMID: 38559149 PMCID: PMC10979885 DOI: 10.1101/2024.03.08.583971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Focused ultrasound (FUS) is an emerging noinvasive technique for neuromodulation in the central nervous system (CNS). To evaluate the effects of FUS-induced neuromodulation, many studies used behavioral changes, functional magnetic resonance imaging (fMRI) or electroencephalography (EEG). However, behavioral readouts are often not easily mapped to specific brain activity, EEG has low spatial resolution limited to the surface of the brain and fMRI requires a large importable scanner that limits additional readouts and manipulations. In this context, functional ultrasound imaging (fUSI) holds promise to directly monitor the effects of FUS neuromodulation with high spatiotemporal resolution in a large field of view, with a comparatively simple and flexible setup. fUSI uses ultrafast Power Doppler Imaging (PDI) to measure changes in cerebral blood volume, which correlates well with neuronal activity and local field potentials. We designed a setup that aligns a FUS transducer with a linear array to allow immediate subsequent monitoring of the hemodynamic response with fUSI during and after FUS neuromodulation. We established a positive correlation between FUS pressure and the size of the activated area, as well as changes in cerebral blood volume (CBV) and found that unilateral sonications produce bilateral hemodynamic changes with ipsilateral accentuation in mice. We further demonstrated the ability to perform fully noninvasive, transcranial FUS-fUSI in nonhuman primates for the first time by using a lower-frequency transducer configuration.
Collapse
Affiliation(s)
- Christian Aurup
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jonas Bendig
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Samuel G. Blackman
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Erica P. McCune
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Sua Bae
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Robin Ji
- 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
|
15
|
Newman M, Rasiah PK, Kusunose J, Rex TS, Mahadevan-Jansen A, Hardenburger J, Jansen ED, Millis B, Caskey CF. Ultrasound Modulates Calcium Activity in Cultured Neurons, Glial Cells, Endothelial Cells and Pericytes. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:341-351. [PMID: 38087717 DOI: 10.1016/j.ultrasmedbio.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 01/23/2024]
Abstract
OBJECTIVE Ultrasound is being researched as a method to modulate the brain. Studies of the interaction of sound with neurons support the hypothesis that mechanosensitive ion channels play an important role in ultrasound neuromodulation. The response of cells other than neurons (e.g., astrocytes, pericytes and endothelial cells) have not been fully characterized, despite playing an important role in brain function. METHODS To address this gap in knowledge, we examined cultured murine primary cortical neurons, astrocytes, endothelial cells and pericytes in an in vitro widefield microscopy setup during application of a 500 ms burst of 250 kHz focused ultrasound over a pressure range known to elicit neuromodulation. We examined cell membrane health in response to a range of pulses and used optical calcium indicators in conjunction with pharmacological antagonists to selectively block different groups of thermo- and mechanosensitive ion channels known to be responsive to ultrasound. RESULTS All cell types experienced an increase in calcium fluorescence in response to ultrasound. Gadolinium (Gad), 2-aminoethoxydiphenyl borate (2-APB) and ruthenium red (RR) reduced the percentage of responding neurons and magnitude of response. The percentage of astrocytes responding was significantly lowered only by Gad, whereas both 2-APB and Gad decreased the amplitude of the fluorescence response. 2-APB decreased the percentage of responding endothelial cells, whereas only Gad reduced the magnitude of responses. Pericytes exposed to RR or Gad were less likely to respond to stimulation. RR had no detectable effect on the magnitude of the pericyte responses while 2-APB and Gad significantly decreased the fluorescence intensity, despite not affecting the percentage responding. CONCLUSION Our study highlights the role of non-neuronal cells during FUS neuromodulation. All of the investigated cell types are sensitive to mechanical ultrasound stimulation and rely on mechanosensitive ion channels to undergo ultrasound neuromodulation.
Collapse
Affiliation(s)
- Malachy Newman
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Pratheepa Kumari Rasiah
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Biophotonics Center, Nashville, TN, USA
| | - Jiro Kusunose
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA; Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tonia S Rex
- Department of Opthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anita Mahadevan-Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Biophotonics Center, Nashville, TN, USA
| | - Jacob Hardenburger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Biophotonics Center, Nashville, TN, USA
| | - E Duco Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Biophotonics Center, Nashville, TN, USA
| | - Bryan Millis
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Biophotonics Center, Nashville, TN, USA
| | - Charles F Caskey
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
16
|
Jiang Z, Cudeiro-Blanco J, Ilbilgi Yildiz B, Sujarittam K, Dickinson RJ, Guasch L, Tang M, Hall TL, Choi JJ. An Ultrasound Array of Emitter-Receiver Stacks for Microbubble-Based Therapy. IEEE Trans Biomed Eng 2024; 71:467-476. [PMID: 37607156 DOI: 10.1109/tbme.2023.3307462] [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: 08/24/2023]
Abstract
Most therapeutic ultrasound devices place emitters and receivers in separate locations, so that the long therapeutic pulses (>1 ms) can be emitted while receivers monitor the procedure. However, with such placement, emitters and receivers are competing for the same space, producing a trade-off between emission efficiency and reception sensitivity. Taking advantage of recent studies demonstrating that short-pulse ultrasound can be used therapeutically, we aimed to develop a device that overcomes such trade-offs. The array was composed of emitter-receiver stacks, which enabled both emission and reception from the same location. Each element was made of a lead zirconate titanate (PZT)-polyvinylidene fluoride (PVDF) stack. The PZT (frequency: 500 kHz, diameter: 16 mm) was used for emission and the PVDF (thickness: 28 μm, diameter: 16 mm) for broadband reception. 32 elements were assembled in a 3D-printed dome-shaped frame (focal length: 150 mm; [Formula: see text]-number: 1) and was tested in free-field and through an ex-vivo human skull. In free-field, the array had a 4.5 × 4.5 × 32 mm focus and produced a peak-negative pressure (PNP) of 2.12 MPa at its geometric center. The electronic steering range was ±15 mm laterally and larger than ±15 mm axially. Through the skull, the array produced a PNP of 0.63 MPa. The PVDF elements were able to localize broadband microbubble emissions across the skull. We built the first multi-element array for short-pulse and microbubble-based therapeutic applications. Stacked arrays overcome traditional trade-offs between the transmission and reception quality and have the potential to create a step change in treatment safety and efficacy.
Collapse
|
17
|
Singh A, Jiménez-Gambín S, Konofagou EE. An all-ultrasound cranial imaging method to establish the relationship between cranial FUS incidence angle and transcranial attenuation in non-human primates in 3D. Sci Rep 2024; 14:1488. [PMID: 38233480 PMCID: PMC10794232 DOI: 10.1038/s41598-024-51623-5] [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: 06/03/2023] [Accepted: 01/08/2024] [Indexed: 01/19/2024] Open
Abstract
Focused ultrasound (FUS) is a non-invasive and non-ionizing technique which deploys ultrasound waves to induce bio-effects. When paired with acoustically active particles such as microbubbles (MBs), it can open the blood brain barrier (BBB) to facilitate drug delivery otherwise inhibited due to the presence of BBB. One of the parameters that affects the FUS beam propagation is the beam incidence angle on the skull. Prior work by our group has shown that, as incidence angles deviate from 90°, FUS focal pressures attenuate and result in a smaller BBB opening volume. The incidence angles calculated in our prior studies were in 2D and used skull information from CT. The study presented herein develops methods to calculate incidence angle in 3D in non-human primate (NHP) skull fragments using harmonic ultrasound imaging without using ionizing radiation. Our results show that ultrasound harmonic imaging is capable of accurately depicting features such as sutures and eye-sockets of the skull. Furthermore, we were able to reproduce previously reported relationships between the incidence angle and FUS beam attenuation. We also show feasibility of performing ultrasound harmonic imaging in in-vivo non-human primates. The all-ultrasound method presented herein combined with our neuronavigation system stands to increase more widespread adoption of FUS and render it accessible by eliminating the need for CT cranial mapping.
Collapse
Affiliation(s)
- Aparna Singh
- 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
|
18
|
Antoniou A, Damianou C. Feasibility of Ultrasonic Heating through Skull Phantom Using Single-element Transducer. J Med Ultrasound 2024; 32:32-40. [PMID: 38665339 PMCID: PMC11040484 DOI: 10.4103/jmu.jmu_3_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/14/2023] [Accepted: 03/31/2023] [Indexed: 04/28/2024] Open
Abstract
Background Noninvasive neurosurgery has become possible through the use of transcranial focused ultrasound (FUS). This study assessed the heating ability of single element spherically focused transducers operating at 0.4 and 1.1 MHz through three-dimensional (3D) printed thermoplastic skull phantoms. Methods Phantoms with precise skull bone geometry of a male patient were 3D printed using common thermoplastic materials following segmentation on a computed tomography head scan image. The brain tissue was mimicked by an agar-based gel phantom developed in-house. The selection of phantom materials was mainly based on transmission-through attenuation measurements. Phantom sonications were performed through water, and then, with the skull phantoms intervening the beam path. In each case, thermometry was performed at the focal spot using thermocouples. Results The focal temperature change in the presence of the skull phantoms was reduced to less than 20 % of that recorded in free field when using the 0.4 MHz transducer, whereas the 1.1 MHz trans-skull sonication produced minimal or no change in focal temperature. The 0.4 MHz transducer showed better performance in trans-skull transmission but still not efficient. Conclusion The inability of both tested single element transducers to steer the beam through the high attenuating skull phantoms and raise the temperature at the focus was confirmed, underlying the necessity to use a correction technique to compensate for energy losses, such those provided by phased arrays. The proposed phantom could be used as a cost-effective and ergonomic tool for trans-skull FUS preclinical studies.
Collapse
Affiliation(s)
- Anastasia Antoniou
- Department of Electrical Engineering, Computer Engineering and Informatics, Cyprus University of Technology, Limassol, Cyprus
| | - Christakis Damianou
- Department of Electrical Engineering, Computer Engineering and Informatics, Cyprus University of Technology, Limassol, Cyprus
| |
Collapse
|
19
|
Datta MS, Chen Y, Chauhan S, Zhang J, De La Cruz ED, Gong C, Tomer R. Whole-brain mapping reveals the divergent impact of ketamine on the dopamine system. Cell Rep 2023; 42:113491. [PMID: 38052211 PMCID: PMC10843582 DOI: 10.1016/j.celrep.2023.113491] [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: 11/07/2022] [Revised: 10/22/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
Ketamine is a multifunctional drug with clinical applications as an anesthetic, pain management medication, and a fast-acting antidepressant. However, it is also recreationally abused for its dissociative effects. Recent studies in rodents are revealing the neuronal mechanisms mediating its actions, but the impact of prolonged exposure to ketamine on brain-wide networks remains less understood. Here, we develop a sub-cellular resolution whole-brain phenotyping approach and utilize it in male mice to show that repeated ketamine administration leads to a dose-dependent decrease in dopamine neurons in midbrain regions linked to behavioral states, alongside an increase in the hypothalamus. Additionally, diverse changes are observed in long-range innervations of the prefrontal cortex, striatum, and sensory areas. Furthermore, the data support a role for post-transcriptional regulation in enabling ketamine-induced neural plasticity. Through an unbiased, high-resolution whole-brain analysis, this study provides important insights into how chronic ketamine exposure reshapes brain-wide networks.
Collapse
Affiliation(s)
- Malika S Datta
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Yannan Chen
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Shradha Chauhan
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Jing Zhang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | - Cheng Gong
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Raju Tomer
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
20
|
Cornelssen C, Finlinson E, Rolston JD, Wilcox KS. Ultrasonic therapies for seizures and drug-resistant epilepsy. Front Neurol 2023; 14:1301956. [PMID: 38162441 PMCID: PMC10756913 DOI: 10.3389/fneur.2023.1301956] [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: 09/25/2023] [Accepted: 11/09/2023] [Indexed: 01/03/2024] Open
Abstract
Ultrasonic therapy is an increasingly promising approach for the treatment of seizures and drug-resistant epilepsy (DRE). Therapeutic focused ultrasound (FUS) uses thermal or nonthermal energy to either ablate neural tissue or modulate neural activity through high- or low-intensity FUS (HIFU, LIFU), respectively. Both HIFU and LIFU approaches have been investigated for reducing seizure activity in DRE, and additional FUS applications include disrupting the blood-brain barrier in the presence of microbubbles for targeted-drug delivery to the seizure foci. Here, we review the preclinical and clinical studies that have used FUS to treat seizures. Additionally, we review effective FUS parameters and consider limitations and future directions of FUS with respect to the treatment of DRE. While detailed studies to optimize FUS applications are ongoing, FUS has established itself as a potential noninvasive alternative for the treatment of DRE and other neurological disorders.
Collapse
Affiliation(s)
- Carena Cornelssen
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - Eli Finlinson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
| | - John D. Rolston
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Karen S. Wilcox
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, United States
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
21
|
Bismuth M, Eck M, Ilovitsh T. Nanobubble-mediated cancer cell sonoporation using low-frequency ultrasound. NANOSCALE 2023; 15:17899-17909. [PMID: 37899700 DOI: 10.1039/d3nr03226d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Ultrasound insonation of microbubbles can form transient pores in cell membranes that enable the delivery of non-permeable extracellular molecules to the cells. Reducing the size of microbubble contrast agents to the nanometer range could facilitate cancer sonoporation. This size reduction can enhance the extravasation of nanobubbles into tumors after an intravenous injection, thus providing a noninvasive sonoporation platform. However, drug delivery efficacy depends on the oscillations of the bubbles, the ultrasound parameters and the size of the target compared to the membrane pores. The formation of large pores is advantageous for the delivery of large molecules, however the small size of the nanobubbles limit the bioeffects when operating near the nanobubble resonance frequency at the MHz range. Here, we show that by coupling nanobubbles with 250 kHz low frequency ultrasound, high amplitude oscillations can be achieved, which facilitate low energy sonoporation of cancer cells. This is beneficial both for increasing the uptake of a specific molecule and to improve large molecule delivery. The method was optimized for the delivery of four fluorescent molecules ranging in size from 1.2 to 70 kDa to breast cancer cells, while comparing the results to targeted microbubbles. Depending on the fluorescent molecule size, the optimal ultrasound peak negative pressure was found to range between 300 and 500 kPa. Increasing the pressure to 800 kPa reduced the fraction of fluorescent cells for all molecules sizes. The optimal uptake for the smaller molecule size of 4 kDa resulted in a fraction of 19.9 ± 1.8% of fluorescent cells, whereas delivery of 20 kDa and 70 kDa molecules yielded 14 ± 0.8% and 4.1 ± 1.1%, respectively. These values were similar to targeted microbubble-mediated sonoporation, suggesting that nanobubbles can serve as noninvasive sonoporation agents with a similar potency, and at a reduced bubble size. The nanobubbles effectively reduced cell viability and may thus potentially reduce the tumor burden, which is crucial for the success of cancer treatment. This method provides a non-invasive and low-energy tumor sonoporation theranostic platform, which can be combined with other therapies to maximize the therapeutic benefits of cancer treatment or be harnessed in gene therapy applications.
Collapse
Affiliation(s)
- Mike Bismuth
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Michal Eck
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
22
|
Song G, Plumlee P, Ahn JY, Wong ST, Zhao H. Translational strategies and systems biology insights for blood-brain barrier opening and delivery in brain tumors and Alzheimer's disease. Biomed Pharmacother 2023; 167:115450. [PMID: 37703663 PMCID: PMC10591819 DOI: 10.1016/j.biopha.2023.115450] [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: 07/23/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023] Open
Abstract
The blood-brain barrier (BBB) plays a critical role in determining the effectiveness of systemic treatments for brain diseases. Over the years, several innovative approaches in BBB opening and drug delivery have been developed and progressed into clinical testing phases, including focused ultrasound (FUS) with circulating microbubbles, mannitol-facilitated delivery of anti-neoplastic drugs, receptor-mediated transcytosis (RMT) by antibody-drug conjugates (ADCs), and viral vectors for gene therapy. We provided a comprehensive review of the most recent clinical applications of these approaches in managing brain tumors and Alzheimer's disease (AD), two major devastating brain diseases. Moreover, the spatial-temporal molecular heterogeneity of the BBB under disease states emphasized the importance of utilizing emerging spatial systems biology approaches to unravel novel targets for intervention within BBB and tailor strategies for enhancing drug delivery to the brain. SEARCH STRATEGY AND SELECTION CRITERIA: Data for this Review were identified by searches of clinicaltrials.gov, MEDLINE, Current Contents, PubMed, and references from relevant articles using the search terms "blood-brain barrier", "CNS drug delivery", "BBB modulation", "clinical trials", "systems biology", "primary or metastatic brain tumors", "Alzheimer's disease". Abstracts and reports from meetings were included only when they related directly to previously published work. Only articles published in English between 1980 and 2023 were included.
Collapse
Affiliation(s)
- Gefei Song
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA
| | - Pierce Plumlee
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA
| | - Ju Young Ahn
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Tc Wong
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA.
| | - Hong Zhao
- T. T. and W. F. Chao Center for BRAIN and Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston TX 77030, USA.
| |
Collapse
|
23
|
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
|
24
|
Bae S, Liu K, Pouliopoulos AN, Ji R, Konofagou EE. Real-Time Passive Acoustic Mapping With Enhanced Spatial Resolution in Neuronavigation-Guided Focused Ultrasound for Blood-Brain Barrier Opening. IEEE Trans Biomed Eng 2023; 70:2874-2885. [PMID: 37159313 PMCID: PMC10538424 DOI: 10.1109/tbme.2023.3266952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
OBJECTIVE Passive acoustic mapping (PAM) provides the spatial information of acoustic energy emitted from microbubbles during focused ultrasound (FUS), which can be used for safety and efficacy monitoring of blood-brain barrier (BBB) opening. In our previous work with a neuronavigation-guided FUS system, only part of the cavitation signal could be monitored in real time due to the computational burden although full-burst analysis is required to detect transient and stochastic cavitation activity. In addition, the spatial resolution of PAM can be limited for a small-aperture receiving array transducer. For full-burst real-time PAM with enhanced resolution, we developed a parallel processing scheme for coherence-factor-based PAM (CF-PAM) and implemented it onto the neuronavigation-guided FUS system using a co-axial phased-array imaging transducer. METHODS Simulation and in-vitro human skull studies were conducted for the performance evaluation of the proposed method in terms of spatial resolution and processing speed. We also carried out real-time cavitation mapping during BBB opening in non-human primates (NHPs). RESULTS CF-PAM with the proposed processing scheme provided better resolution than that of traditional time-exposure-acoustics PAM with a higher processing speed than that of eigenspace-based robust Capon beamformer, which facilitated the full-burst PAM with the integration time of 10 ms at a rate of 2 Hz. In vivo feasibility of PAM with the co-axial imaging transducer was also demonstrated in two NHPs, showing the advantages of using real-time B-mode and full-burst PAM for accurate targeting and safe treatment monitoring. SIGNIFICANCE This full-burst PAM with enhanced resolution will facilitate the clinical translation of online cavitation monitoring for safe and efficient BBB opening.
Collapse
|
25
|
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: 1.5] [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
|
26
|
Lao YH, Ji R, Zhou JK, Snow KJ, Kwon N, Saville E, He S, Chauhan S, Chi CW, Datta MS, Zhang H, Quek CH, Cai SS, Li M, Gaitan Y, Bechtel L, Wu SY, Lutz CM, Tomer R, Murray SA, Chavez A, Konofagou EE, Leong KW. Focused ultrasound-mediated brain genome editing. Proc Natl Acad Sci U S A 2023; 120:e2302910120. [PMID: 37579143 PMCID: PMC10450663 DOI: 10.1073/pnas.2302910120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/28/2023] [Indexed: 08/16/2023] Open
Abstract
Gene editing in the brain has been challenging because of the restricted transport imposed by the blood-brain barrier (BBB). Current approaches mainly rely on local injection to bypass the BBB. However, such administration is highly invasive and not amenable to treating certain delicate regions of the brain. We demonstrate a safe and effective gene editing technique by using focused ultrasound (FUS) to transiently open the BBB for the transport of intravenously delivered CRISPR/Cas9 machinery to the brain.
Collapse
Affiliation(s)
- Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY10027
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY14214
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | - Joyce K. Zhou
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | | | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | | | - Siyu He
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | - Shradha Chauhan
- Department of Biological Sciences, Columbia University, New York, NY10027
| | - Chun-Wei Chi
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY14214
| | - Malika S. Datta
- Department of Biological Sciences, Columbia University, New York, NY10027
| | - Hairong Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | - Chai Hoon Quek
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | - S. Sarah Cai
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | - Mingqiang Li
- Department of Biomedical Engineering, Columbia University, New York, NY10027
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou510630, China
| | | | | | - Shih-Ying Wu
- Department of Biomedical Engineering, Columbia University, New York, NY10027
| | | | - Raju Tomer
- Department of Biological Sciences, Columbia University, New York, NY10027
| | | | - Alejandro Chavez
- Department of Pediatrics, University of California San Diego, La Jolla, CA92037
| | - Elisa E. Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY10027
- Department of Radiology, Columbia University Medical Center, New York, NY10032
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY10027
- Department of Systems Biology, Columbia University Medical Center, New York, NY10032
| |
Collapse
|
27
|
Rivandi H, Costa TL. A 2D Ultrasound Phased-Array Transmitter ASIC for High-Frequency US Stimulation and Powering. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2023; 17:701-712. [PMID: 37352088 DOI: 10.1109/tbcas.2023.3288891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2023]
Abstract
Ultrasound (US) neuromodulation and ultrasonic power transfer to implanted devices demand novel ultrasound transmitters capable of steering focused ultrasound waves in 3D with high spatial resolution and US pressure, while having a miniaturized form factor. Meeting these requirements needs a 2D array of ultrasound transducers directly integrated with a high-frequency 2D phased-array ASIC. However, this imposes severe challenges on the design of the ASIC. In order to avoid the generation of grating lobes, the elements in the 2D phased-array should have a pitch of half of the ultrasound wavelength, which, as frequency increases, highly reduces the area available for the design of high-voltage beamforming channels. This article addresses these challenges by presenting the system-level optimization and implementation of a high-frequency 2D phased-array ASIC. The system-level study focuses on the optimization of the US transmitter toward high-frequency operation while minimizing power consumption. This study resulted in the implementation of two ASICs in TSMC 180 nm BCD technology: firstly, an individual beamforming channel was designed to demonstrate the tradeoffs between frequency, driving voltage, and beamforming capabilities. Finally, a 12-MHz pitch matched 12 × 12 phased-array ASIC working at 20-V amplitude and 3-bit phasing was designed and experimentally validated, to demonstrate high-frequency phased-array operation. The measurement results verify the phasing functionality of the ASIC with a maximum DNL of 0.35 LSB. The CMOS chip consumes 130 mW and 26.6 mW average power during the continuous pulsing and delivering 200-pulse bursts with a PRF of 1 kHz, respectively.
Collapse
|
28
|
Karakatsani ME, Ji R, Murillo MF, Kugelman T, Kwon N, Lao YH, Liu K, Pouliopoulos AN, Honig LS, Duff KE, Konofagou EE. Focused ultrasound mitigates pathology and improves spatial memory in Alzheimer's mice and patients. Theranostics 2023; 13:4102-4120. [PMID: 37554284 PMCID: PMC10405840 DOI: 10.7150/thno.79898] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/12/2023] [Indexed: 08/10/2023] Open
Abstract
Rationale: Bilateral sonication with focused ultrasound (FUS) in conjunction with microbubbles has been shown to separately reduce amyloid plaques and hyperphosphorylated tau protein in the hippocampal formation and the entorhinal cortex in different mouse models of Alzheimer's disease (AD) without any therapeutic agents. However, the two pathologies are expressed concurrently in human disease. Therefore, the objective of this study is to investigate the effects of repeated bilateral sonications in the presence of both pathologies. Methods: Herein, we investigate its functional and morphological outcomes on brains bearing both pathologies simultaneously. Eleven transgenic mice of the 3xTg-AD line (14 months old) expressing human amyloid beta and human tau and eleven age-matched wild-type littermates received four weekly bilateral sonications covering the hippocampus followed by working memory testing. Afterwards, immunohistochemistry and immunoassays (western blot and ELISA) were employed to assess any changes in amyloid beta and human tau. Furthermore, we present preliminary data from our clinical trial using a neuronavigation-guided FUS system for sonications in AD patients (NCT04118764). Results: Interestingly, both wild-type and transgenic animals that received FUS experienced improved working memory and spent significantly more time in the escape platform-quadrant, with wild-type animals spending 43.2% (sham: 37.7%) and transgenic animals spending 35.3% (sham: 31.0%) of the trial in the target quadrant. Furthermore, this behavioral amelioration in the transgenic animals correlated with a 58.3% decrease in the neuronal length affected by tau and a 27.2% reduction in total tau levels. Amyloid plaque population, volume and overall load were also reduced overall. Consistently, preliminary data from a clinical trial involving AD patients showed a 1.8% decrease of amyloid PET signal 3-weeks after treatment in the treated hemisphere compared to baseline. Conclusion: For the first time, it is shown that bilateral FUS-induced BBB opening significantly and simultaneously ameliorates both coexistent pathologies, which translated to improvements in spatial memory of transgenic animals with complex AD, the human mimicking phenotype. The level of cognitive improvement was significantly correlated with the volume of BBB opening. Non-transgenic animals were also shown to exhibit similar memory amelioration for the first time, indicating that BBB opening results into benefits in the neuronal function regardless of the existence of AD pathology. A potential mechanism of action for the reduction of the both pathologies investigated was the cholesterol metabolism, specifically the LRP1b receptor, which exhibited increased expression levels in transgenic mice following FUS-induced BBB opening. Initial clinical evidence supported that the beta amyloid reduction shown in rodents could be translatable to humans with significant amyloid reduction shown in the treated hemisphere.
Collapse
Affiliation(s)
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Maria F. Murillo
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Tara Kugelman
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Keyu Liu
- Department of Biomedical Engineering, Columbia University, New York, USA
| | | | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Karen E. Duff
- UK Dementia Research Institute, University College London, London, UK
| | - Elisa E. Konofagou
- Department of Radiology, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
29
|
Liu D, Munoz F, Sanatkhani S, Pouliopoulos AN, Konofagou EE, Grinband J, Ferrera VP. Alteration of functional connectivity in the cortex and major brain networks of non-human primates following focused ultrasound exposure in the dorsal striatum. Brain Stimul 2023; 16:1196-1204. [PMID: 37558125 PMCID: PMC10530553 DOI: 10.1016/j.brs.2023.08.003] [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: 03/09/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Focused ultrasound (FUS) is a non-invasive neuromodulation technology that is being investigated for potential treatment of neurological and psychiatric disorders. FUS combined with microbubbles can temporarily open the intact blood-brain barrier (BBB) of animals and humans, and facilitate drug delivery. FUS exposure, either with or without microbubbles, has been demonstrated to alter the behavior of non-human primates (NHP), and previous studies have demonstrated the transient and long-term effects of FUS neuromodulation on functional connectivity using resting state functional MRI. The behavioral effects of FUS vary depending on whether or not it is applied in conjunction with microbubbles to open the BBB, but it is unknown whether opening the BBB affects functional connectivity differently than FUS alone. OBJECTIVE To compare the effects of applying FUS alone (FUS neuromodulation) and FUS with microbubbles (FUS-BBB opening) on changes of resting state functional connectivity in NHP. METHODS We applied 2 min FUS exposure without (neuromodulation) and with microbubbles (BBB opening) in the dorsal striatum of lightly anesthetized non-human primates, and acquired resting state functional MRI 40 min respectively after FUS exposure. The functional connectivity (FC) in the cortex and major brain networks between the two approaches were measured and compared. RESULTS When applying FUS exposure to the caudate nucleus of NHP, we found that both FUS neuromodulation can activate FC between caudate and insular cortex, while inhibiting the FC between caudate and motor cortex. FUS-BBB opening can activate FC between the caudate and medial prefrontal cortex, and within the frontotemporal network (FTN). We also found both FUS and FUS-BBB opening can significantly activate FC within the default mode network (DMN). CONCLUSION The results suggest applying FUS to a deep brain structure can alter functional connectivity in the DMN and FTN, and that FUS neuromodulation and FUS-mediated BBB opening can have different effects on patterns of functional connectivity.
Collapse
Affiliation(s)
- Dong Liu
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA.
| | - Fabian Munoz
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA
| | - Soroosh Sanatkhani
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA
| | - Antonios N Pouliopoulos
- Department of Surgical & Interventional Engineering, School of Biomedical Engineering & Imaging Science, King's College London, UK
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, USA; Department of Radiology, Columbia University, USA
| | - Jack Grinband
- Department of Radiology, Columbia University, USA; Department of Psychiatry, Columbia University, USA
| | - Vincent P Ferrera
- Department of Neuroscience, Columbia University, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, USA; Department of Psychiatry, Columbia University, USA
| |
Collapse
|
30
|
Antoniou A, Nikolaou A, Georgiou A, Evripidou N, Damianou C. Development of an US, MRI, and CT imaging compatible realistic mouse phantom for thermal ablation and focused ultrasound evaluation. ULTRASONICS 2023; 131:106955. [PMID: 36854247 DOI: 10.1016/j.ultras.2023.106955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/09/2022] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Tissue mimicking phantoms (TMPs) play an essential role in modern biomedical research as cost-effective quality assurance and training tools, simultaneously contributing to the reduction of animal use. Herein, we present the development and evaluation of an anatomically accurate mouse phantom intended for image-guided thermal ablation and Focused Ultrasound (FUS) applications. The proposed mouse model consists of skeletal and soft tissue mimics, whose design was based on the Computed tomography (CT) scans data of a live mouse. Advantageously, it is compatible with US, CT, and Magnetic Resonance Imaging (MRI). The compatibility assessment was focused on the radiological behavior of the phantom due to the lack of relevant literature. The X-ray linear attenuation coefficient of candidate materials was estimated to assess the one that matches best the radiological behavior of living tissues. The bone part was manufactured by Fused Deposition Modeling (FDM) printing using Acrylonitrile styrene acrylate (ASA) material. For the soft-tissue mimic, a special mold was 3D printed having a cavity with the unique shape of the mouse body and filled with an agar-based silica-doped gel. The mouse phantom accurately matched the size and reproduced the body surface of the imaged mouse. Tissue-equivalency in terms of X-ray attenuation was demonstrated for the agar-based soft-tissue mimic. The phantom demonstrated excellent MRI visibility of the skeletal and soft-tissue mimics. Good radiological contrast between the skeletal and soft-tissue models was also observed in the CT scans. The model was also able to reproduce realistic behavior during trans-skull sonication as proved by thermocouple measurements. Overall, the proposed phantom is inexpensive, ergonomic, and realistic. It could constitute a powerful tool for image-guided thermal ablation and FUS studies in terms of testing and optimizing the performance of relevant equipment and protocols. It also possess great potential for use in transcranial FUS applications, including the emerging topic of FUS-mediated blood brain barrier (BBB) disruption.
Collapse
Affiliation(s)
- Anastasia Antoniou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus.
| | - Anastasia Nikolaou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus.
| | - Andreas Georgiou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus.
| | - Nikolas Evripidou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus.
| | - Christakis Damianou
- Department of Electrical Engineering, Computer Engineering, and Informatics, Cyprus University of Technology, Limassol, Cyprus.
| |
Collapse
|
31
|
Xu L, Pacia CP, Gong Y, Hu Z, Chien CY, Yang L, Gach HM, Hao Y, Comron H, Huang J, Leuthardt EC, Chen H. Characterization of the Targeting Accuracy of a Neuronavigation-Guided Transcranial FUS System In Vitro, In Vivo, and In Silico. IEEE Trans Biomed Eng 2023; 70:1528-1538. [PMID: 36374883 PMCID: PMC10176741 DOI: 10.1109/tbme.2022.3221887] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Focused ultrasound (FUS)-enabled liquid biopsy (sonobiopsy) is an emerging technique for the noninvasive and spatiotemporally controlled diagnosis of brain cancer by inducing blood-brain barrier (BBB) disruption to release brain tumor-specific biomarkers into the blood circulation. The feasibility, safety, and efficacy of sonobiopsy were demonstrated in both small and large animal models using magnetic resonance-guided FUS devices. However, the high cost and complex operation of magnetic resonance-guided FUS devices limit the future broad application of sonobiopsy in the clinic. In this study, a neuronavigation-guided sonobiopsy device is developed and its targeting accuracy is characterized in vitro, in vivo, and in silico. The sonobiopsy device integrated a commercially available neuronavigation system (BrainSight) with a nimble, lightweight FUS transducer. Its targeting accuracy was characterized in vitro in a water tank using a hydrophone. The performance of the device in BBB disruption was verified in vivo using a pig model, and the targeting accuracy was quantified by measuring the offset between the target and the actual locations of BBB opening. The feasibility of the FUS device in targeting glioblastoma (GBM) tumors was evaluated in silico using numerical simulation by the k-Wave toolbox in glioblastoma patients. It was found that the targeting accuracy of the neuronavigation-guided sonobiopsy device was 1.7 ± 0.8 mm as measured in the water tank. The neuronavigation-guided FUS device successfully induced BBB disruption in pigs with a targeting accuracy of 3.3 ± 1.4 mm. The targeting accuracy of the FUS transducer at the GBM tumor was 5.5 ± 4.9 mm. Age, sex, and incident locations were found to be not correlated with the targeting accuracy in GBM patients. This study demonstrated that the developed neuronavigation-guided FUS device could target the brain with a high spatial targeting accuracy, paving the foundation for its application in the clinic.
Collapse
|
32
|
Johansen PM, Hansen PY, Mohamed AA, Girshfeld SJ, Feldmann M, Lucke-Wold B. Focused ultrasound for treatment of peripheral brain tumors. EXPLORATION OF DRUG SCIENCE 2023; 1:107-125. [PMID: 37171968 PMCID: PMC10168685 DOI: 10.37349/eds.2023.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/14/2023]
Abstract
Malignant brain tumors are the leading cause of cancer-related death in children and remain a significant cause of morbidity and mortality throughout all demographics. Central nervous system (CNS) tumors are classically treated with surgical resection and radiotherapy in addition to adjuvant chemotherapy. However, the therapeutic efficacy of chemotherapeutic agents is limited due to the blood-brain barrier (BBB). Magnetic resonance guided focused ultrasound (MRgFUS) is a new and promising intervention for CNS tumors, which has shown success in preclinical trials. High-intensity focused ultrasound (HIFU) has the capacity to serve as a direct therapeutic agent in the form of thermoablation and mechanical destruction of the tumor. Low-intensity focused ultrasound (LIFU) has been shown to disrupt the BBB and enhance the uptake of therapeutic agents in the brain and CNS. The authors present a review of MRgFUS in the treatment of CNS tumors. This treatment method has shown promising results in preclinical trials including minimal adverse effects, increased infiltration of the therapeutic agents into the CNS, decreased tumor progression, and improved survival rates.
Collapse
Affiliation(s)
| | - Payton Yerke Hansen
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Ali A. Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Sarah J. Girshfeld
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Marc Feldmann
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
33
|
Lim Kee Chang W, Chan TG, Raguseo F, Mishra A, Chattenton D, de Rosales RTM, Long NJ, Morse SV. Rapid short-pulses of focused ultrasound and microbubbles deliver a range of agent sizes to the brain. Sci Rep 2023; 13:6963. [PMID: 37117169 PMCID: PMC10147927 DOI: 10.1038/s41598-023-33671-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023] Open
Abstract
Focused ultrasound and microbubbles can non-invasively and locally deliver therapeutics and imaging agents across the blood-brain barrier. Uniform treatment and minimal adverse bioeffects are critical to achieve reliable doses and enable safe routine use of this technique. Towards these aims, we have previously designed a rapid short-pulse ultrasound sequence and used it to deliver a 3 kDa model agent to mouse brains. We observed a homogeneous distribution in delivery and blood-brain barrier closing within 10 min. However, many therapeutics and imaging agents are larger than 3 kDa, such as antibody fragments and antisense oligonucleotides. Here, we evaluate the feasibility of using rapid short-pulses to deliver higher-molecular-weight model agents. 3, 10 and 70 kDa dextrans were successfully delivered to mouse brains, with decreasing doses and more heterogeneous distributions with increasing agent size. Minimal extravasation of endogenous albumin (66.5 kDa) was observed, while immunoglobulin (~ 150 kDa) and PEGylated liposomes (97.9 nm) were not detected. This study indicates that rapid short-pulses are versatile and, at an acoustic pressure of 0.35 MPa, can deliver therapeutics and imaging agents of sizes up to a hydrodynamic diameter between 8 nm (70 kDa dextran) and 11 nm (immunoglobulin). Increasing the acoustic pressure can extend the use of rapid short-pulses to deliver agents beyond this threshold, with little compromise on safety. This study demonstrates the potential for deliveries of higher-molecular-weight therapeutics and imaging agents using rapid short-pulses.
Collapse
Affiliation(s)
- William Lim Kee Chang
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Tiffany G Chan
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Federica Raguseo
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Aishwarya Mishra
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SW1 7EH, UK
| | - Dani Chattenton
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, London, SM2 5NG, UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SW1 7EH, UK
| | - Nicholas J Long
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Sophie V Morse
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK.
| |
Collapse
|
34
|
Noel RL, Batts AJ, Ji R, Pouliopoulos AN, Bae S, Kline-Schoder AR, Konofagou EE. Natural aging and Alzheimer's disease pathology increase susceptibility to focused ultrasound-induced blood-brain barrier opening. Sci Rep 2023; 13:6757. [PMID: 37185578 PMCID: PMC10130033 DOI: 10.1038/s41598-023-30466-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 02/23/2023] [Indexed: 05/17/2023] Open
Abstract
Focused Ultrasound (FUS) paired with systemically-injected microbubbles (μB) is capable of transiently opening the blood-brain barrier (BBBO) for noninvasive and targeted drug delivery to the brain. FUS-BBBO is also capable of modulating the neuroimmune system, further qualifying its therapeutic potential for neurodegenerative diseases like Alzheimer's disease (AD). Natural aging and AD impose significant strain on the brain and particularly the BBB, modifying its structure and subsequently, its functionality. The emerging focus on treating neurodegenerative diseases with FUS-BBBO necessitates an investigation into the extent that age and AD affect the BBB's response to FUS. FUS-BBBO was performed with a 1.5-MHz, geometrically focused transducer operated at 450 kPa and paired with a bolus microbubble injection of 8 × 108 μB/mL. Here we quantify the BBBO, BBB closing (BBBC) timeline, and BBB permeability (BBBP) following FUS-BBBO in male mice with and without AD pathology, aged 10 weeks, one year, or two years. The data presented herein indicates that natural aging and AD pathology may increase initial BBBO volume by up to 34.4% and 40.7% respectively, extend BBBC timeline by up to 1.3 and 1.5 days respectively, and increase BBBP as measured by average Ktrans values up to 80% and 86.1% respectively in male mice. This characterization of the BBB response to FUS-BBBO with age and AD further clarifies the nature and extent of the functional impact of these factors and may offer new considerations for planning FUS-BBBO interventions in aged and AD populations.
Collapse
Affiliation(s)
- R L Noel
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
| | - A J Batts
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - R Ji
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - A N Pouliopoulos
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - S Bae
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - A R Kline-Schoder
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - E E Konofagou
- Department of Biological Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
- Department of Radiology, Columbia University, 622 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
35
|
Datta MS, Chen Y, Chauhan S, Zhang J, De La Cruz ED, Gong C, Tomer R. Whole-brain mapping reveals the divergent impact of ketamine on the dopamine system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536506. [PMID: 37090584 PMCID: PMC10120808 DOI: 10.1101/2023.04.12.536506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Ketamine is a multifunctional drug with clinical applications as an anesthetic, as a pain management medication and as a transformative fast-acting antidepressant. It is also abused as a recreational drug due to its dissociative property. Recent studies in rodents are revealing the neuronal mechanisms that mediate the complex actions of ketamine, however, its long-term impact due to prolonged exposure remains much less understood with profound scientific and clinical implications. Here, we develop and utilize a high-resolution whole-brain phenotyping approach to show that repeated ketamine administration leads to a dosage-dependent decrease of dopamine (DA) neurons in the behavior state-related midbrain regions and, conversely, an increase within the hypothalamus. Congruently, we show divergently altered innervations of prefrontal cortex, striatum, and sensory areas. Further, we present supporting data for the post-transcriptional regulation of ketamine-induced structural plasticity. Overall, through an unbiased whole-brain analysis, we reveal the divergent brain-wide impact of chronic ketamine exposure on the association and sensory pathways.
Collapse
Affiliation(s)
- Malika S. Datta
- Department of Biological Sciences, Columbia University
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University
| | - Yannan Chen
- Department of Biological Sciences, Columbia University
- Department of Biomedical Engineering, Columbia University
| | | | - Jing Zhang
- Department of Biological Sciences, Columbia University
| | | | - Cheng Gong
- Department of Biological Sciences, Columbia University
- Department of Biomedical Engineering, Columbia University
| | - Raju Tomer
- Department of Biological Sciences, Columbia University
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University
- Department of Biomedical Engineering, Columbia University
| |
Collapse
|
36
|
Blackmore DG, Razansky D, Götz J. Ultrasound as a versatile tool for short- and long-term improvement and monitoring of brain function. Neuron 2023; 111:1174-1190. [PMID: 36917978 DOI: 10.1016/j.neuron.2023.02.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/19/2023] [Accepted: 02/13/2023] [Indexed: 03/15/2023]
Abstract
Treating the brain with focused ultrasound (FUS) at low intensities elicits diverse responses in neurons, astroglia, and the extracellular matrix. In combination with intravenously injected microbubbles, FUS also opens the blood-brain barrier (BBB) and facilitates focal drug delivery. However, an incompletely understood cellular specificity and a wide parameter space currently limit the optimal application of FUS in preclinical and human studies. In this perspective, we discuss how different FUS modalities can be utilized to achieve short- and long-term improvements, thereby potentially treating brain disorders. We review the ongoing efforts to determine which parameters induce neuronal inhibition versus activation and how mechanoreceptors and signaling cascades are activated to induce long-term changes, including memory improvements. We suggest that optimal FUS treatments may require different FUS modalities and devices, depending on the targeted brain area or local pathology, and will be greatly enhanced by new techniques for monitoring FUS efficacy.
Collapse
Affiliation(s)
- Daniel G Blackmore
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel Razansky
- Institute for Biomedical Engineering, Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, 8057 Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, 8093 Zurich, Switzerland
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
37
|
Wear KA, Shah A. Nominal Versus Actual Spatial Resolution: Comparison of Directivity and Frequency-Dependent Effective Sensitive Element Size for Membrane, Needle, Capsule, and Fiber-Optic Hydrophones. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2023; 70:112-119. [PMID: 36178990 DOI: 10.1109/tuffc.2022.3211183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Frequency-dependent effective sensitive element radius [Formula: see text] is a key parameter for elucidating physical mechanisms of hydrophone operation. In addition, it is essential to know [Formula: see text] to correct for hydrophone output voltage reduction due to spatial averaging across the hydrophone sensitive element surface. At low frequencies, [Formula: see text] is greater than geometrical sensitive element radius ag . Consequently, at low frequencies, investigators can overrate their hydrophone spatial resolution. Empirical models for [Formula: see text] for membrane, needle, and fiber-optic hydrophones have been obtained previously. In this article, an empirical model for [Formula: see text] for capsule hydrophones is presented, so that models are now available for the four most common hydrophone types used in biomedical ultrasound. The [Formula: see text] value was estimated from directivity measurements (over the range from 1 to 20 MHz) for five capsule hydrophones (three with [Formula: see text] and two with [Formula: see text]). The results suggest that capsule hydrophones behave according to a "rigid piston" model for k a g ≥ 0.7 ( k = 2π /wavelength). Comparing the four hydrophone types, the low-frequency discrepancy between [Formula: see text] and ag was found to be greatest for membrane hydrophones, followed by capsule hydrophones, and smallest for needle and fiber-optic hydrophones. Empirical models for [Formula: see text] are helpful for choosing an appropriate hydrophone for an experiment and for correcting for spatial averaging (over the sensitive element surface) in pressure and beamwidth measurements. When reporting hydrophone-based pressure measurements, investigators should specify [Formula: see text] at the center frequency (which may be estimated from the models presented here) in addition to ag .
Collapse
|
38
|
Past, present and future of Focused Ultrasound as an adjunct or complement to DIPG/DMG therapy: A consensus of the 2021 FUSF DIPG meeting. Neoplasia 2023; 37:100876. [PMID: 36709715 PMCID: PMC9900434 DOI: 10.1016/j.neo.2023.100876] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 01/29/2023]
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG), now known as Diffuse Midline Glioma (DMG) is a devastating pediatric brain tumor with limited treatment options and a very poor prognosis. Despite more than 250 clinical trials aimed to treat children diagnosed with DMG, no curative therapies currently exist for this patient population. A major obstacle has been the intact blood brain barrier (BBB) which prevents most therapeutics from crossing into the tumor bed. Focused Ultrasound (FUS) is an emerging, noninvasive medical technology which has been shown in both preclinical and clinical research to disrupt the blood brain barrier safely and temporarily. FUS blood brain barrier opening has been studied in combination with chemotherapies in preclinical DMG models, and this technology is now being investigated in clinical trials for the treatment of pediatric brain tumors. Focused ultrasound has additional mechanisms of action, including sonodynamic therapy and radiation sensitization, that hold promise as future DMG therapies as well. This paper, largely based off the proceedings from a workshop held by the Focused Ultrasound Foundation in October of 2021, summarizes the current state of the field of focused ultrasound for DIPG/DMG, including preclinical, technical, and clinical summaries in addition to recommended next steps for continued advancement of the game changing technology of Focused Ultrasound.
Collapse
|
39
|
Leong K, Lao YH, Ji R, Zhou J, Snow K, Kwon N, Saville E, He S, Chauhan S, Chi CW, Datta M, Zhang H, Quek CH, Cai S, Li M, Gaitan Y, Bechtel L, Wu SY, Lutz C, Tomer R, Murray S, Chavez A, Konofagou E. Focused ultrasound-mediated brain genome editing. RESEARCH SQUARE 2023:rs.3.rs-2365576. [PMID: 36712096 PMCID: PMC9882596 DOI: 10.21203/rs.3.rs-2365576/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Gene editing in the mammalian brain has been challenging because of the restricted transport imposed by the blood-brain barrier (BBB). Current approaches rely on local injection to bypass the BBB. However, such administration is highly invasive and not amenable to treating certain delicate regions of the brain. We demonstrate a safe and effective gene editing technique by using focused ultrasound (FUS) to transiently open the BBB for the transport of intravenously delivered CRISPR/Cas9 machinery to the brain.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Stephen Murray
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609
| | | | | |
Collapse
|
40
|
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: 14] [Impact Index Per Article: 4.7] [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
|
41
|
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: 43] [Impact Index Per Article: 14.3] [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
|
42
|
Barzegar-Fallah A, Gandhi K, Rizwan SB, Slatter TL, Reynolds JNJ. Harnessing Ultrasound for Targeting Drug Delivery to the Brain and Breaching the Blood–Brain Tumour Barrier. Pharmaceutics 2022; 14:pharmaceutics14102231. [PMID: 36297666 PMCID: PMC9607160 DOI: 10.3390/pharmaceutics14102231] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant advances in developing drugs to treat brain tumours, achieving therapeutic concentrations of the drug at the tumour site remains a major challenge due to the presence of the blood–brain barrier (BBB). Several strategies have evolved to enhance brain delivery of chemotherapeutic agents to treat tumours; however, most approaches have several limitations which hinder their clinical utility. Promising studies indicate that ultrasound can penetrate the skull to target specific brain regions and transiently open the BBB, safely and reversibly, with a high degree of spatial and temporal specificity. In this review, we initially describe the basics of therapeutic ultrasound, then detail ultrasound-based drug delivery strategies to the brain and the mechanisms by which ultrasound can improve brain tumour therapy. We review pre-clinical and clinical findings from ultrasound-mediated BBB opening and drug delivery studies and outline current therapeutic ultrasound devices and technologies designed for this purpose.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Kushan Gandhi
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Shakila B. Rizwan
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand
| | - Tania L. Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - John N. J. Reynolds
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- Correspondence: ; Tel.: +64-3-479-5781; Fax: +64-3-479-7254
| |
Collapse
|
43
|
Opportunities and challenges in delivering biologics for Alzheimer's disease by low-intensity ultrasound. Adv Drug Deliv Rev 2022; 189:114517. [PMID: 36030018 DOI: 10.1016/j.addr.2022.114517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/24/2023]
Abstract
Low-intensity ultrasound combined with intravenously injected microbubbles (US+MB) is a novel treatment modality for brain disorders, including Alzheimer's disease (AD), safely and transiently allowing therapeutic agents to overcome the blood-brain barrier (BBB) that constitutes a major barrier for therapeutic agents. Here, we first provide an update on immunotherapies in AD and how US+MB has been applied to AD mouse models and in clinical trials, considering the ultrasound and microbubble parameter space. In the second half of the review, we compare different in vitro BBB models and discuss strategies for combining US+MB with BBB modulators (targeting molecules such as claudin-5), and highlight the insight provided by super-resolution microscopy. Finally, we conclude with a short discussion on how in vitro findings can inform the design of animal studies, and how the insight gained may aid treatment optimization in the clinical ultrasound space.
Collapse
|
44
|
Arsiwala TA, Sprowls SA, Blethen KE, Fladeland RA, Wolford CP, Kielkowski BN, Glass MJ, Wang P, Wilson O, Carpenter JS, Ranjan M, Finomore V, Rezai A, Lockman PR. Characterization of passive permeability after low intensity focused ultrasound mediated blood-brain barrier disruption in a preclinical model. Fluids Barriers CNS 2022; 19:72. [PMID: 36076213 PMCID: PMC9461249 DOI: 10.1186/s12987-022-00369-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Systemic drug delivery to the central nervous system is limited by presence of the blood-brain barrier (BBB). Low intensity focused ultrasound (LiFUS) is a non-invasive technique to disrupt the BBB, though there is a lack of understanding of the relationship between LiFUS parameters, such as cavitation dose, time of sonication, microbubble dose, and the time course and magnitude of BBB disruption. Discrepancies in these data arise from experimentation with modified, clinically untranslatable transducers and inconsistent parameters for sonication. In this report, we characterize microbubble and cavitation doses as LiFUS variables as they pertain to the time course and size of BBB opening with a clinical Insightec FUS system. METHODS Female Nu/Nu athymic mice were exposed to LiFUS using the ExAblate Neuro system (v7.4, Insightec, Haifa, Israel) following target verification with magnetic resonance imaging (MRI). Microbubble and cavitation doses ranged from 4-400 μL/kg, and 0.1-1.5 cavitation dose, respectively. The time course and magnitude of BBB opening was evaluated using fluorescent tracers, ranging in size from 105-10,000 Da, administered intravenously at different times pre- or post-LiFUS. Quantitative autoradiography and fluorescence microscopy were used to quantify tracer accumulation in brain. RESULTS We observed a microbubble and cavitation dose dependent increase in tracer uptake within brain after LiFUS. Tracer accumulation was size dependent, with 14C-AIB (100 Da) accumulating to a greater degree than larger markers (~ 625 Da-10 kDa). Our data suggest opening of the BBB via LiFUS is time dependent and biphasic. Accumulation of solutes was highest when administered prior to LiFUS mediated disruption (2-fivefold increases), but was also significantly elevated at 6 h post treatment for both 14C-AIB and Texas Red. CONCLUSION The magnitude of LiFUS mediated BBB opening correlates with concentration of microbubbles, cavitation dose as well as time of tracer administration post-sonication. These data help define the window of maximal BBB opening and applicable sonication parameters on a clinically translatable and commercially available FUS system that can be used to improve passive permeability and accumulation of therapeutics targeting the brain.
Collapse
Affiliation(s)
- Tasneem A. Arsiwala
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA
| | - Samuel A. Sprowls
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA ,grid.239578.20000 0001 0675 4725Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106 USA
| | - Kathryn E. Blethen
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA
| | - Ross A. Fladeland
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA
| | - Cullen P. Wolford
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA
| | - Brooke N. Kielkowski
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA
| | - Morgan J. Glass
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA
| | - Peng Wang
- grid.268154.c0000 0001 2156 6140Rockefeller Neuroscience Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| | - Olivia Wilson
- grid.268154.c0000 0001 2156 6140Rockefeller Neuroscience Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| | - Jeffrey S. Carpenter
- grid.268154.c0000 0001 2156 6140Rockefeller Neuroscience Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA ,grid.268154.c0000 0001 2156 6140Departments of Neuroscience, Neuroradiology, and Neurosurgery, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| | - Manish Ranjan
- grid.268154.c0000 0001 2156 6140Rockefeller Neuroscience Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA ,grid.268154.c0000 0001 2156 6140Departments of Neuroscience, Neuroradiology, and Neurosurgery, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| | - Victor Finomore
- grid.268154.c0000 0001 2156 6140Departments of Neuroscience, Neuroradiology, and Neurosurgery, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| | - Ali Rezai
- grid.268154.c0000 0001 2156 6140Rockefeller Neuroscience Institute, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA ,grid.268154.c0000 0001 2156 6140Departments of Neuroscience, Neuroradiology, and Neurosurgery, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| | - Paul R. Lockman
- grid.268154.c0000 0001 2156 6140Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, HSC, Morgantown, 1 Medical Center Dr, Morgantown, WV 26506 USA ,grid.268154.c0000 0001 2156 6140Departments of Neuroscience, Neuroradiology, and Neurosurgery, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26505 USA
| |
Collapse
|
45
|
Aryal M, Azadian MM, Hart AR, Macedo N, Zhou Q, Rosenthal EL, Airan RD. Noninvasive ultrasonic induction of cerebrospinal fluid flow enhances intrathecal drug delivery. J Control Release 2022; 349:434-442. [PMID: 35798095 DOI: 10.1016/j.jconrel.2022.06.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Intrathecal drug delivery is routinely used in the treatment and prophylaxis of varied central nervous system conditions, as doing so allows drugs to directly bypass the blood-brain barrier. However, the utility of this route of administration is limited by poor brain and spinal cord parenchymal drug uptake from the cerebrospinal fluid. We demonstrate that a simple noninvasive transcranial ultrasound protocol can significantly increase influx of cerebrospinal fluid into the perivascular spaces of the brain, to enhance the uptake of intrathecally administered drugs. Specifically, we administered small (~1 kDa) and large (~155 kDa) molecule agents into the cisterna magna of rats and then applied low, diagnostic-intensity focused ultrasound in a scanning protocol throughout the brain. Using real-time magnetic resonance imaging and ex vivo histologic analyses, we observed significantly increased uptake of small molecule agents into the brain parenchyma, and of both small and large molecule agents into the perivascular space from the cerebrospinal fluid. Notably, there was no evidence of brain parenchymal damage following this intervention. The low intensity and noninvasive approach of transcranial ultrasound in this protocol underscores the ready path to clinical translation of this technique. In this manner, this protocol can be used to directly bypass the blood-brain barrier for whole-brain delivery of a variety of agents. Additionally, this technique can potentially be used as a means to probe the causal role of the glymphatic system in the variety of disease and physiologic processes to which it has been correlated.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Departments of Engineering and Radiation Oncology, Loyola University Chicago, Chicago, IL, United States
| | - Matine M Azadian
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Alex R Hart
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Nicholas Macedo
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Zhou
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Stanford Cancer Center, Stanford Medical Center, Stanford, CA, United States; Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Raag D Airan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Department of Materials Science and Engineering, Stanford University School of Medicine, Stanford, CA, United States; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
46
|
Balbi M, Blackmore DG, Padmanabhan P, Götz J. Ultrasound-Mediated Bioeffects in Senescent Mice and Alzheimer's Mouse Models. Brain Sci 2022; 12:775. [PMID: 35741660 PMCID: PMC9221310 DOI: 10.3390/brainsci12060775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/01/2023] Open
Abstract
Ultrasound is routinely used for a wide range of diagnostic imaging applications. However, given that ultrasound can operate over a wide range of parameters that can all be modulated, its applicability extends far beyond the bioimaging field. In fact, the modality has emerged as a hybrid technology that effectively assists drug delivery by transiently opening the blood-brain barrier (BBB) when combined with intravenously injected microbubbles, and facilitates neuromodulation. Studies in aged mice contributed to an insight into how low-intensity ultrasound brings about its neuromodulatory effects, including increased synaptic plasticity and improved cognitive functions, with a potential role for neurogenesis and the modulation of NMDA receptor-mediated neuronal signalling. This work is complemented by studies in mouse models of Alzheimer's disease (AD), a form of pathological ageing. Here, ultrasound was mainly employed as a BBB-opening tool that clears protein aggregates via microglial activation and neuronal autophagy, thereby restoring cognition. We discuss the currently available ultrasound approaches and how studies in senescent mice are relevant for AD and can accelerate the application of low-intensity ultrasound in the clinic.
Collapse
Affiliation(s)
- Matilde Balbi
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
| | - Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jürgen Götz
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
47
|
Phipps MA, Jonathan S, Yang PF, Chen LM, Grissom W, Caskey CF. A reduced aperture allows for transcranial focus localization at lower pressure. JASA EXPRESS LETTERS 2022; 2:062001. [PMID: 35782333 PMCID: PMC9245740 DOI: 10.1121/10.0011695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Localizing the focus during transcranial focused ultrasound procedures is important to ensure accurate targeting of specific brain regions and interpretation of results. Magnetic resonance acoustic radiation force imaging uses the displacement induced by the ultrasound focus in the brain to localize the beam, but the high pressure required to displace brain tissue may cause damage or confounds during subsequent neuromodulatory experiments. Here, reduced apertures were applied to a phased array transducer to generate comparable displacement to the full aperture but with 20% lower free field pressure.
Collapse
Affiliation(s)
- M Anthony Phipps
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Sumeeth Jonathan
- Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, USA , , , , ,
| | - Pai-Feng Yang
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Li Min Chen
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - William Grissom
- Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37232, USA , , , , ,
| | - Charles F Caskey
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
48
|
Mungur R, Zheng J, Wang B, Chen X, Zhan R, Tong Y. Low-Intensity Focused Ultrasound Technique in Glioblastoma Multiforme Treatment. Front Oncol 2022; 12:903059. [PMID: 35677164 PMCID: PMC9169875 DOI: 10.3389/fonc.2022.903059] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma is one of the central nervous system most aggressive and lethal cancers with poor overall survival rate. Systemic treatment of glioblastoma remains the most challenging aspect due to the low permeability of the blood-brain barrier (BBB) and blood-tumor barrier (BTB), limiting therapeutics extravasation mainly in the core tumor as well as in its surrounding invading areas. It is now possible to overcome these barriers by using low-intensity focused ultrasound (LIFU) together with intravenously administered oscillating microbubbles (MBs). LIFU is a non-invasive technique using converging ultrasound waves which can alter the permeability of BBB/BTB to drug delivery in a specific brain/tumor region. This emerging technique has proven to be both safe and repeatable without causing injury to the brain parenchyma including neurons and other structures. Furthermore, LIFU is also approved by the FDA to treat essential tremors and Parkinson's disease. It is currently under clinical trial in patients suffering from glioblastoma as a drug delivery strategy and liquid biopsy for glioblastoma biomarkers. The use of LIFU+MBs is a step-up in the world of drug delivery, where onco-therapeutics of different molecular sizes and weights can be delivered directly into the brain/tumor parenchyma. Initially, several potent drugs targeting glioblastoma were limited to cross the BBB/BTB; however, using LIFU+MBs, diverse therapeutics showed significantly higher uptake, improved tumor control, and overall survival among different species. Here, we highlight the therapeutic approach of LIFU+MBs mediated drug-delivery in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Rajneesh Mungur
- Department of Neurosurgery of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiesheng Zheng
- Department of Neurosurgery of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ben Wang
- Key Laboratory of Cancer Prevention and Intervention, Key Laboratory of Molecular Biology in Medical Sciences, National Ministry of Education, Cancer Institute, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Xinhua Chen
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Renya Zhan
- Department of Neurosurgery of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Tong
- Department of Neurosurgery of the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
49
|
Andrés D, Jiménez N, Benlloch JM, Camarena F. Numerical Study of Acoustic Holograms for Deep-Brain Targeting through the Temporal Bone Window. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:872-886. [PMID: 35221196 DOI: 10.1016/j.ultrasmedbio.2022.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 06/14/2023]
Abstract
Acoustic holograms can encode complex wavefronts to compensate the aberrations of a therapeutical ultrasound beam propagating through heterogeneous tissues such as the skull, and simultaneously, they can generate diffraction-limited acoustic images, that is, arbitrary shaped focal spots. In this work, we numerically study the performance of acoustic holograms focusing at the thalamic nuclei when the source is located at the temporal bone window. The temporal window is the thinnest area of the lateral skull and it is mainly hairless, so it is a desirable area through which to transmit ultrasonic waves to the deep brain. However, in targeting from this area the bilateral thalamic nuclei are not aligned with the elongated focal spots of conventional focused transducers, and in addition, skull aberrations can distort the focal spot. We found that by using patient-specific holographic lenses coupled to a single-element 650-kHz-frequency 65-mm-aperture source, the focal spot can be sharply adapted to the thalamic nuclei in a bilateral way while skull aberrations are mitigated. Furthermore, the performance of these holograms was studied under misalignment errors between the source and the skull, concluding that for misalignments up to 5°, acoustic images are correctly restored. This work paves the way to designing clinical applications of transcranial ultrasound such as blood-brain barrier opening for drug delivery or deep-brain neuromodulation using this low-cost and personalized technology, presenting desirable aspects for long-term treatments because the patient's head does not need to be shaved completely and skull heating is low.
Collapse
Affiliation(s)
- Diana Andrés
- Instituto de Instrumentación para Imagen Molecular (i3M), Universitat Politècnica de València, Consejo Superior de Investigaciones Científicas (CSIC), València, Spain
| | - Noé Jiménez
- Instituto de Instrumentación para Imagen Molecular (i3M), Universitat Politècnica de València, Consejo Superior de Investigaciones Científicas (CSIC), València, Spain.
| | - José M Benlloch
- Instituto de Instrumentación para Imagen Molecular (i3M), Universitat Politècnica de València, Consejo Superior de Investigaciones Científicas (CSIC), València, Spain
| | - Francisco Camarena
- Instituto de Instrumentación para Imagen Molecular (i3M), Universitat Politècnica de València, Consejo Superior de Investigaciones Científicas (CSIC), València, Spain
| |
Collapse
|
50
|
Gandhi K, Barzegar-Fallah A, Banstola A, Rizwan SB, Reynolds JNJ. Ultrasound-Mediated Blood-Brain Barrier Disruption for Drug Delivery: A Systematic Review of Protocols, Efficacy, and Safety Outcomes from Preclinical and Clinical Studies. Pharmaceutics 2022; 14:pharmaceutics14040833. [PMID: 35456667 PMCID: PMC9029131 DOI: 10.3390/pharmaceutics14040833] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Ultrasound-mediated blood-brain barrier (BBB) disruption has garnered focus as a method of delivering normally impenetrable drugs into the brain. Numerous studies have investigated this approach, and a diverse set of ultrasound parameters appear to influence the efficacy and safety of this approach. An understanding of these findings is essential for safe and reproducible BBB disruption, as well as in identifying the limitations and gaps for further advancement of this drug delivery approach. We aimed to collate and summarise protocols and parameters for achieving ultrasound-mediated BBB disruption in animal and clinical studies, as well as the efficacy and safety methods and outcomes associated with each. A systematic search of electronic databases helped in identifying relevant, included studies. Reference lists of included studies were further screened to identify supplemental studies for inclusion. In total, 107 articles were included in this review, and the following parameters were identified as influencing efficacy and safety outcomes: microbubbles, transducer frequency, peak-negative pressure, pulse characteristics, and the dosing of ultrasound applications. Current protocols and parameters achieving ultrasound-mediated BBB disruption, as well as their associated efficacy and safety outcomes, are identified and summarised. Greater standardisation of protocols and parameters in future preclinical and clinical studies is required to inform robust clinical translation.
Collapse
Affiliation(s)
- Kushan Gandhi
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (K.G.); (A.B.-F.); (A.B.)
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand;
| | - Anita Barzegar-Fallah
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (K.G.); (A.B.-F.); (A.B.)
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand;
| | - Ashik Banstola
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (K.G.); (A.B.-F.); (A.B.)
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand;
| | - Shakila B. Rizwan
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand;
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand
| | - John N. J. Reynolds
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand; (K.G.); (A.B.-F.); (A.B.)
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand;
- Correspondence: ; Tel.: +64-3479-5781; Fax: +64-3479-7254
| |
Collapse
|