1
|
Lian Y, Zeng Y, Zhou S, Zhu H, Li F, Cai X. Deep Beamforming for Real-Time 3-D Passive Acoustic Mapping With Row-Column-Addressed Arrays. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2025; 72:226-237. [PMID: 40030804 DOI: 10.1109/tuffc.2024.3524436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Passive acoustic mapping (PAM) is a promising tool to monitor acoustic cavitation activities for focused ultrasound (FUS) therapies. While 2-D matrix arrays allow 3-D PAM, the high channel count requirement and the complexity of the receiving electronics limit their practical value in real-time imaging applications. In this regard, row-column-addressed (RCA) arrays have shown great potential in addressing the difficulties in real-time 3-D ultrasound imaging. However, currently, there is no applicable method for 3-D PAM with RCA arrays. In this work, we propose a deep beamformer for real-time 3-D PAM with RCA arrays. The deep beamformer leverages a deep neural network (DNN) to map radio frequency (RF) microbubble (MB) cavitation signals acquired with the RCA array to 3-D PAM images, achieving similar image quality to the reconstructions performed using the fully populated 2-D matrix array with the angular spectrum (AS) method. In the simulation, the images reconstructed by the deep beamformer showed less than 13.2% and 1.8% differences in the energy spread volume (ESV) and image signal-to-noise ratio (ISNR), compared with those reconstructed using the matrix array. However, the image reconstruction time was reduced by 11 and 30 times on the CPU and GPU, respectively, achieving 42.4 volumes per second image reconstruction speed on a GPU for a volume sized $128\times 128\times 128$ . Experimental data further validated the capabilities of the deep beamformer to accurately localize MB cavitation activities in 3-D space. These results clearly demonstrated the feasibility of real-time and 3-D monitoring of MB cavitation activities with RCA arrays and neural network-based beamformers.
Collapse
|
2
|
Pakdaman Zangabad R, Lee H, Zhang X, Sait Kilinc M, Arvanitis CD, Levent Degertekin F. A High Sensitivity CMUT-Based Passive Cavitation Detector for Monitoring Microbubble Dynamics During Focused Ultrasound Interventions. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1087-1096. [PMID: 39088497 PMCID: PMC11558552 DOI: 10.1109/tuffc.2024.3436918] [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: 08/03/2024]
Abstract
Tracking and controlling microbubble (MB) dynamics in the human brain through acoustic emission (AE) monitoring during transcranial focused ultrasound (tFUS) therapy are critical for attaining safe and effective treatments. The low-amplitude MB emissions have harmonic and ultra-harmonic components, necessitating a broad bandwidth and low-noise system for monitoring transcranial MB activity. Capacitive micromachined ultrasonic transducers (CMUTs) offer high sensitivity and low noise over a broad bandwidth, especially when they are tightly integrated with electronics, making them a good candidate technology for monitoring the MB activity through human skull. In this study, we designed a 16-channel analog front-end (AFE) electronics with a low-noise transimpedance amplifier (TIA), a band-gap reference circuit, and an output buffer stage. To assess AFE performance and ability to detect MB AE, we combined it with a commercial CMUT array. The integrated system has 12.3 - [Formula: see text] receive sensitivity with 0.085 - [Formula: see text] minimum detectable pressure (MDP) up to 3 MHz for a single element CMUT with 3.78 [Formula: see text] area. Experiments with free MBs in a microfluidic channel demonstrate that our system is able to capture key spectral components of MBs' harmonics when sonicated at clinically relevant frequencies (0.5 MHz) and pressures (250 kPa). Together our results demonstrate that the proposed CMUT system can support the development of novel passive cavitation detectors (PCD) to track MB activity for attaining safe and effective focused ultrasound (FUS) treatments.
Collapse
|
3
|
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
|
4
|
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
|
5
|
Zhu H, Zeng Y, Cai X. Passive Acoustic Mapping for Convex Arrays With the Helical Wave Spectrum Method. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:1923-1933. [PMID: 38198274 DOI: 10.1109/tmi.2024.3352283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Passive acoustic mapping (PAM) has emerged as a valuable imaging modality for monitoring the cavitation activity in focused ultrasound therapies. When it comes to imaging in the human abdomen, convex arrays are preferred due to their large acoustic window. However, existing PAM methods for convex arrays rely on the computationally expensive delay-and-sum (DAS) operation limiting the image reconstruction speed when the field-of-view (FOV) is large. In this work, we propose an efficient and frequency-selective PAM method for convex arrays. This method is based on projecting the helical wave spectrum (HWS) between cylindrical surfaces in the imaging field. Both the in silico and in vitro experiments showed that the HWS method has comparable image quality and similar acoustic cavitation source localization accuracy as the DAS-based methods. Compared to the frequency-domain and time-domain DAS methods, the time-complexity of the HWS method is reduced by one order and two orders of magnitude, respectively. A parallel implementation of the HWS method realized millisecond-level image reconstruction speed. We also show that the HWS method is inherently capable of mapping microbubble (MB) cavitation activity of different status, i.e., no cavitation, stable cavitation, or inertial cavitation. After compensating for the lens effects of the convex array, we further combined PAM formed by the HWS method and B-mode imaging as a real-time dual-mode imaging approach to map the anatomical location where MBs cavitate in a liver phantom experiment. This method may find use in applications where convex arrays are required for cavitation activity monitoring in real time.
Collapse
|
6
|
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
|
7
|
Lu S, Su R, Wan C, Guo S, Wan M. Passive acoustic mapping with absolute time-of-flight information and delay-multiply-sum beamforming. Med Phys 2023; 50:2323-2335. [PMID: 36704970 DOI: 10.1002/mp.16248] [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: 09/27/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Passive acoustic mapping (PAM) is showing increasing application potential in monitoring ultrasound therapy by spatially resolving cavitation activity. PAM with the relative time-of-flight information leads to poor axial resolution when implemented with ultrasound diagnostic transducers. Through utilizing the absolute time-of-flight information preserved by the transmit-receive synchronization and applying the common delay-sum (DS) beamforming algorithm, PAM axial resolution can be greatly improved in the short-pulse excitation scenario, as with active ultrasound imaging. However, PAM with the absolute time-of-flight information (referred as AtPAM) suffers from low imaging resolution and weak interference suppression when the DS algorithm is applied. PURPOSE This study aims to propose an enhanced AtPAM algorithm based on delay-multiply-sum (DMS) beamforming, to address the shortcomings of the DS-based AtPAM algorithm. METHODS In DMS beamforming, the element signals delayed by the absolute time delays are first processed with a signed square-root operation and then multiplied in pairs and finally summed, the resulting beamformed output is further band-pass filtered. The performances of DS- and DMS-based AtPAMs are compared by experiments, in which an ultrasound diagnostic transducer (a linear array) is employed to passively sense the wire signals generated by an unfocused ultrasound transducer and the cavitation signals generated by a focused therapeutic ultrasound transducer in a flow phantom. The AtPAM image quality is assessed by main-lobe width (MLW), intensity valley value (IVV), area of pixels (AOP), signal-to-interference ratio (SIR), and signal-to-noise ratio (SNR). RESULTS The single-wire experimental results show that compared to the DS algorithm, the DMS algorithm leads to an enhanced AtPAM image with a decreased transverse MLW of 0.15 mm and an improved SIR and SNR of 31.50 and 18.77 dB. For the four-wire images, the transverse (axial) IVV is decreased by 18.37 dB (13.11 dB) and the SIR (the SNR) is increased by 26.13 dB (18.47 dB) when using the DMS algorithm. The cavitation activity is better highlighted by DMS-based AtPAM, which decreases the AOP by 0.81 mm2 (-10-dB level) and 4.43 mm2 (-20-dB level) and increases the SIR and SNR by 20.14 and 10.48 dB respectively. The pixel distributions of AtPAM images of both wires and cavitation activity also indicate a better suppression of the DMS algorithm in sidelobe and noise. CONCLUSIONS The experimental results illustrate that the DMS algorithm can improve the image quality of AtPAM compared to the DS algorithm. DMS-based AtPAM is beneficial for detecting cavitation activity during short-pulse ultrasound exposure with high resolution, and further for monitoring short-pulse ultrasound therapy.
Collapse
Affiliation(s)
- Shukuan Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Ruibo Su
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chunye Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Shifang Guo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
8
|
Lee H, Guo Y, Ross JL, Schoen S, Degertekin FL, Arvanitis C. Spatially targeted brain cancer immunotherapy with closed-loop controlled focused ultrasound and immune checkpoint blockade. SCIENCE ADVANCES 2022; 8:eadd2288. [PMID: 36399574 PMCID: PMC9674274 DOI: 10.1126/sciadv.add2288] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/21/2022] [Indexed: 05/28/2023]
Abstract
Despite the challenges in treating glioblastomas (GBMs) with immune adjuvants, increasing evidence suggests that targeting the immune cells within the tumor microenvironment (TME) can lead to improved responses. Here, we present a closed-loop controlled, microbubble-enhanced focused ultrasound (MB-FUS) system and test its abilities to safely and effectively treat GBMs using immune checkpoint blockade. The proposed system can fine-tune the exposure settings to promote MB acoustic emission-dependent expression of the proinflammatory marker ICAM-1 and delivery of anti-PD1 in a mouse model of GBM. In addition to enhanced interaction of proinflammatory macrophages within the PD1-expressing TME and significant improvement in survival (P < 0.05), the combined treatment induced long-lived memory T cell formation within the brain that supported tumor rejection in rechallenge experiments. Collectively, our findings demonstrate the ability of MB-FUS to augment the therapeutic impact of immune checkpoint blockade in GBMs and reinforce the notion of spatially tumor-targeted (loco-regional) brain cancer immunotherapy.
Collapse
Affiliation(s)
- Hohyun Lee
- G.W. School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yutong Guo
- G.W. School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - James L. Ross
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Scott Schoen
- Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - F. Levent Degertekin
- G.W. School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Costas Arvanitis
- G.W. School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Georgia Institute of Technology and Emory University, Department of Biomedical Engineering, Atlanta, GA, USA
| |
Collapse
|
9
|
Chien CY, Yang Y, Gong Y, Yue Y, Chen H. Blood-Brain Barrier Opening by Individualized Closed-Loop Feedback Control of Focused Ultrasound. BME FRONTIERS 2022; 2022:9867230. [PMID: 37850162 PMCID: PMC10521637 DOI: 10.34133/2022/9867230] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/01/2022] [Indexed: 10/19/2023] Open
Abstract
Objective and Impact Statement. To develop an approach for individualized closed-loop feedback control of microbubble cavitation to achieve safe and effective focused ultrasound in combination with microbubble-induced blood-brain barrier opening (FUS-BBBO). Introduction. FUS-BBBO is a promising strategy for noninvasive and localized brain drug delivery with a growing number of clinical studies currently ongoing. Real-time cavitation monitoring and feedback control are critical to achieving safe and effective FUS-BBBO. However, feedback control algorithms used in the past were either open-loop or without consideration of baseline cavitation level difference among subjects. Methods. This study performed feedback-controlled FUS-BBBO by defining the target cavitation level based on the baseline stable cavitation level of an individual subject with "dummy" FUS sonication. The dummy FUS sonication applied FUS with a low acoustic pressure for a short duration in the presence of microbubbles to define the baseline stable cavitation level that took into consideration of individual differences in the detected cavitation emissions. FUS-BBBO was then achieved through two sonication phases: ramping-up phase to reach the target cavitation level and maintaining phase to control the stable cavitation level at the target cavitation level. Results. Evaluations performed in wild-type mice demonstrated that this approach achieved effective and safe trans-BBB delivery of a model drug. The drug delivery efficiency increased as the target cavitation level increased from 0.5 dB to 2 dB without causing vascular damage. Increasing the target cavitation level to 3 dB and 4 dB increased the probability of tissue damage. Conclusions. Safe and effective brain drug delivery was achieved using the individualized closed-loop feedback-controlled FUS-BBBO.
Collapse
Affiliation(s)
- Chih-Yen Chien
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Yan Gong
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, Missouri 63130, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, Missouri 63108, USA
| |
Collapse
|
10
|
Lechpammer M, Rao R, Shah S, Mirheydari M, Bhattacharya D, Koehler A, Toukam DK, Haworth KJ, Pomeranz Krummel D, Sengupta S. Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers. Cancers (Basel) 2022; 14:1627. [PMID: 35406398 PMCID: PMC8997081 DOI: 10.3390/cancers14071627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, or glioblastoma multiforme (GBM, WHO Grade IV), is a highly aggressive adult glioma. Despite extensive efforts to improve treatment, the current standard-of-care (SOC) regimen, which consists of maximal resection, radiotherapy, and temozolomide (TMZ), achieves only a 12-15 month survival. The clinical improvements achieved through immunotherapy in several extracranial solid tumors, including non-small-cell lung cancer, melanoma, and non-Hodgkin lymphoma, inspired investigations to pursue various immunotherapeutic interventions in adult glioblastoma patients. Despite some encouraging reports from preclinical and early-stage clinical trials, none of the tested agents have been convincing in Phase III clinical trials. One, but not the only, factor that is accountable for the slow progress is the blood-brain barrier, which prevents most antitumor drugs from reaching the target in appreciable amounts. Herein, we review the current state of immunotherapy in glioblastoma and discuss the significant challenges that prevent advancement. We also provide thoughts on steps that may be taken to remediate these challenges, including the application of ultrasound technologies.
Collapse
Affiliation(s)
- Mirna Lechpammer
- Foundation Medicine, Inc., Cambridge, MA 02141, USA;
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rohan Rao
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Sanjit Shah
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Mona Mirheydari
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Abigail Koehler
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Donatien Kamdem Toukam
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Kevin J. Haworth
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| |
Collapse
|
11
|
Schoen S, Dash P, Arvanitis CD. Experimental Demonstration of Trans-Skull Volumetric Passive Acoustic Mapping With the Heterogeneous Angular Spectrum Approach. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:534-542. [PMID: 34748486 PMCID: PMC10243207 DOI: 10.1109/tuffc.2021.3125670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Real-time, 3-D, passive acoustic mapping (PAM) of microbubble dynamics during transcranial focused ultrasound (FUS) is essential for optimal treatment outcomes. The angular spectrum approach (ASA) potentially offers a very efficient method to perform PAM, as it can reconstruct specific frequency bands pertinent to microbubble dynamics and may be extended to correct aberrations caused by the skull. Here, we experimentally assess the abilities of heterogeneous ASA (HASA) to perform trans-skull PAM. Our experimental investigations demonstrate that the 3-D PAMs of a known 1-MHz source, constructed with HASA through an ex vivo human skull segment, reduced both the localization error (from 4.7 ± 2.3 to 2.3 ± 1.6 mm) and the number, size, and energy of spurious lobes caused by aberration, with the modest additional computational expense. While further improvements in the localization errors are expected with arrays with denser elements and larger aperture, our analysis revealed that experimental constraints associated with the array pitch and aperture (here, 1.8 mm and 2.5 cm, respectively) can be ameliorated by interpolation and peak finding techniques. Beyond the array characteristics, our analysis also indicated that errors in the registration (translation and rotation of ±5 mm and ±5°, respectively) of the skull segment to the array can lead to peak localization errors of the order of a few wavelengths. Interestingly, errors in the spatially dependent speed of sound in the skull (±20%) caused only subwavelength errors in the reconstructions, suggesting that registration is the most important determinant of point source localization accuracy. Collectively, our findings show that HASA can address source localization problems through the skull efficiently and accurately under realistic conditions, thereby creating unique opportunities for imaging and controlling the microbubble dynamics in the brain.
Collapse
|
12
|
SonoVue ® vs. Sonazoid™ vs. Optison™: Which Bubble Is Best for Low-Intensity Sonoporation of Pancreatic Ductal Adenocarcinoma? Pharmaceutics 2022; 14:pharmaceutics14010098. [PMID: 35056994 PMCID: PMC8777813 DOI: 10.3390/pharmaceutics14010098] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/15/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
The use of ultrasound and microbubbles to enhance therapeutic efficacy (sonoporation) has shown great promise in cancer therapy from in vitro to ongoing clinical studies. The fastest bench-to-bedside translation involves the use of ultrasound contrast agents (microbubbles) and clinical diagnostic scanners. Despite substantial research in this field, it is currently not known which of these microbubbles result in the greatest enhancement of therapy within the applied conditions. Three microbubble formulations-SonoVue®, Sonazoid™, and Optison™-were physiochemically and acoustically characterized. The microbubble response to the ultrasound pulses used in vivo was simulated via a Rayleigh-Plesset type equation. The three formulations were compared in vitro for permeabilization efficacy in three different pancreatic cancer cell lines, and in vivo, using an orthotopic pancreatic cancer (PDAC) murine model. The mice were treated using one of the three formulations exposed to ultrasound from a GE Logiq E9 and C1-5 ultrasound transducer. Characterisation of the microbubbles showed a rapid degradation in concentration, shape, and/or size for both SonoVue® and Optison™ within 30 min of reconstitution/opening. Sonazoid™ showed no degradation after 1 h. Attenuation measurements indicated that SonoVue® was the softest bubble followed by Sonazoid™ then Optison™. Sonazoid™ emitted nonlinear ultrasound at the lowest MIs followed by Optison™, then SonoVue®. Simulations indicated that SonoVue® would be the most effective bubble using the evaluated ultrasound conditions. This was verified in the pre-clinical PDAC model demonstrated by improved survival and largest tumor growth inhibition. In vitro results indicated that the best microbubble formulation depends on the ultrasound parameters and concentration used, with SonoVue® being best at lower intensities and Sonazoid™ at higher intensities.
Collapse
|
13
|
Schoen S, Kilinc MS, Lee H, Guo Y, Degertekin FL, Woodworth GF, Arvanitis C. Towards controlled drug delivery in brain tumors with microbubble-enhanced focused ultrasound. Adv Drug Deliv Rev 2022; 180:114043. [PMID: 34801617 PMCID: PMC8724442 DOI: 10.1016/j.addr.2021.114043] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/27/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
Brain tumors are particularly challenging malignancies, due to their location in a structurally and functionally distinct part of the human body - the central nervous system (CNS). The CNS is separated and protected by a unique system of brain and blood vessel cells which together prevent most bloodborne therapeutics from entering the brain tumor microenvironment (TME). Recently, great strides have been made through microbubble (MB) ultrasound contrast agents in conjunction with ultrasound energy to locally increase the permeability of brain vessels and modulate the brain TME. As we elaborate in this review, this physical method can effectively deliver a wide range of anticancer agents, including chemotherapeutics, antibodies, and nanoparticle drug conjugates across a range of preclinical brain tumors, including high grade glioma (glioblastoma), diffuse intrinsic pontine gliomas, and brain metastasis. Moreover, recent evidence suggests that this technology can promote the effective delivery of novel immunotherapeutic agents, including immune check-point inhibitors and chimeric antigen receptor T cells, among others. With early clinical studies demonstrating safety, and several Phase I/II trials testing the preclinical findings underway, this technology is making firm steps towards shaping the future treatments of primary and metastatic brain cancer. By elaborating on its key components, including ultrasound systems and MB technology, along with methods for closed-loop spatial and temporal control of MB activity, we highlight how this technology can be tuned to enable new, personalized treatment strategies for primary brain malignancies and brain metastases.
Collapse
Affiliation(s)
- Scott Schoen
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - M. Sait Kilinc
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Hohyun Lee
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yutong Guo
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - F. Levent Degertekin
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Graeme F. Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA,Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, College Park, MD 20742, USA,Fischell Department of Bioengineering A. James Clarke School of Engineering, University of Maryland
| | - Costas Arvanitis
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA,Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| |
Collapse
|
14
|
Tan C, Yan B, Han T, Yu ACH, Qin P. Improving temporal stability of stable cavitation activity of circulating microbubbles using a closed-loop controller based on pulse-length regulation. ULTRASONICS SONOCHEMISTRY 2022; 82:105882. [PMID: 34969003 PMCID: PMC8855699 DOI: 10.1016/j.ultsonch.2021.105882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/30/2021] [Accepted: 12/18/2021] [Indexed: 05/16/2023]
Abstract
Stable cavitation (SC) has shown great potential for novel therapeutic applications. The spatiotemporal distribution of the SC activity of microbubbles circulating in a target region is not only correlated with the uniformity of treatment, but also with some undesirable effects. Therefore, it is important to achieve controllable and desirable SC activity in target regions for improved therapeutic efficiency and biosafety. This study proposes a closed-loop feedback controller based on pulse length (PL) regulation to improve the temporal stability of SC activity. Microbubbles circulating in a physiological flowing phantom were exposed to a 1 MHz focused transducer. The SC signals produced were initially received by another 7.5 MHz plane transducer, followed by high-speed signal acquisition and real-time processing. Based on the real-time-measured SC intensity excited by the current acoustic pulse, the proposed closed-loop feedback controller used three proportional coefficients to regulate the peak negative pressure (PNP) and PL of the next acoustic pulse during the acceleration and stable stages, respectively. The results show that the rise time and the temporal stability of the SC intensity of the microbubbles circulating in these two stages were improved significantly by the optimized proportional coefficients used in the proposed controller. Importantly, when compared with the traditional closed-loop feedback controller based on PNP regulation, the proposed closed-loop feedback controller based on PL regulation reduced the probability of a transition between stable and inertial cavitation, thus avoiding the risk of disadvantageous bioeffects in practical applications. These results demonstrate the effectiveness of the proposed PL-based closed-loop feedback controller and provide a feasible strategy for realization of controllable cavitation activity in applications.
Collapse
Affiliation(s)
- Chunjie Tan
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bo Yan
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tao Han
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON N2L3G1, Canada
| | - Peng Qin
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
15
|
Localized blood-brain barrier opening in infiltrating gliomas with MRI-guided acoustic emissions-controlled focused ultrasound. Proc Natl Acad Sci U S A 2021; 118:2103280118. [PMID: 34504017 DOI: 10.1073/pnas.2103280118] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological treatment of gliomas and other brain-infiltrating tumors remains challenging due to limited delivery of most therapeutics across the blood-brain barrier (BBB). Transcranial MRI-guided focused ultrasound (FUS), an emerging technology for noninvasive brain treatments, enables transient opening of the BBB through acoustic activation of circulating microbubbles. Here, we evaluate the safety and utility of transcranial microbubble-enhanced FUS (MB-FUS) for spatially targeted BBB opening in patients with infiltrating gliomas. In this Phase 0 clinical trial (NCT03322813), we conducted comparative and quantitative analyses of FUS exposures (sonications) and their effects on gliomas using MRI, histopathology, microbubble acoustic emissions (harmonic dose [HD]), and fluorescence-guided surgery metrics. Contrast-enhanced MRI and histopathology indicated safe and reproducible BBB opening in all patients. These observations occurred using a power cycling closed feedback loop controller, with the power varying by nearly an order of magnitude on average. This range underscores the need for monitoring and titrating the exposure on a patient-by-patient basis. We found a positive correlation between microbubble acoustic emissions (HD) and MR-evident BBB opening (P = 0.07) and associated interstitial changes (P < 0.01), demonstrating the unique capability to titrate the MB-FUS effects in gliomas. Importantly, we identified a 2.2-fold increase of fluorescein accumulation in MB-FUS-treated compared to untreated nonenhancing tumor tissues (P < 0.01) while accounting for vascular density. Collectively, this study demonstrates the capabilities of MB-FUS for safe, localized, controlled BBB opening and highlights the potential of this technology to improve the surgical and pharmacologic treatment of brain tumors.
Collapse
|
16
|
Najem H, Khasraw M, Heimberger AB. Immune Microenvironment Landscape in CNS Tumors and Role in Responses to Immunotherapy. Cells 2021; 10:2032. [PMID: 34440802 PMCID: PMC8393758 DOI: 10.3390/cells10082032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the important evolution of immunotherapeutic agents, brain tumors remain, in general, refractory to immune therapeutics. Recent discoveries have revealed that the glioma microenvironment includes a wide variety of immune cells in various states that play an important role in the process of tumorigenesis. Anti-tumor immune activity may be occurring or induced in immunogenic hot spots or at the invasive edge of central nervous system (CNS) tumors. Understanding the complex heterogeneity of the immune microenvironment in gliomas will likely be the key to unlocking the full potential of immunotherapeutic strategies. An essential consideration will be the induction of immunological effector responses in the setting of the numerous aspects of immunosuppression and evasion. As such, immune therapeutic combinations are a fundamental objective for clinical studies in gliomas. Through immune profiling conducted on immune competent murine models of glioma and ex vivo human glioma tissue, we will discuss how the frequency, distribution of immune cells within the microenvironment, and immune modulatory processes, may be therapeutically modulated to lead to clinical benefits.
Collapse
Affiliation(s)
- Hinda Najem
- Department of Neurological Surgery and Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC 27710, USA;
| | - Amy B. Heimberger
- Department of Neurological Surgery and Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| |
Collapse
|
17
|
Top CB. A Generalized Split-Step Angular Spectrum Method for Efficient Simulation of Wave Propagation in Heterogeneous Media. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:2687-2696. [PMID: 33891551 DOI: 10.1109/tuffc.2021.3075367] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Angular spectrum (AS) methods enable efficient calculation of wave propagation from one plane to another inside homogeneous media. For wave propagation in heterogeneous media such as biological tissues, AS methods cannot be applied directly. Split-stepping techniques decompose the heterogeneous domain into homogeneous and perturbation parts, and provide a solution for forward wave propagation by propagating the incident wave in both frequency-space and frequency-wavenumber domains. Recently, a split-step hybrid angular spectrum (HAS) method was proposed for plane wave propagation of focused ultrasound beams. In this study, we extend these methods to enable simulation of acoustic pressure field for an arbitrary source distribution, by decomposing the source and reflection spectra into orthogonal propagation direction components, propagating each component separately, and summing all components to get the total field. We show that our method can efficiently simulate the pressure field of arbitrary sources in heterogeneous media. The accuracy of the method was analyzed comparing the resultant pressure field with pseudospectral time domain (PSTD) solution for breast tomography and hemispherical transcranial-focused ultrasound simulation models. Eighty times acceleration was achieved for a 3-D breast simulation model compared to PSTD solution with 0.005 normalized root mean-squared difference (NRMSD) between two solutions. For the hemispherical phased array, aberrations due to skull were accurately calculated in a single simulation run as evidenced by the resultant-focused ultrasound beam simulations, which had 0.001 NRMSD with 40 times acceleration factor compared to the PSTD method.
Collapse
|
18
|
Polichetti M, Varray F, Gilles B, Bera JC, Nicolas B. Use of the Cross-Spectral Density Matrix for Enhanced Passive Ultrasound Imaging of Cavitation. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:910-925. [PMID: 33079648 DOI: 10.1109/tuffc.2020.3032345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Passive ultrasound imaging is of great interest for cavitation monitoring. Spatiotemporal monitoring of cavitation bubbles in therapeutic applications is possible using an ultrasound imaging probe to passively receive the acoustic signals from the bubbles. Fourier-domain (FD) beamformers have been proposed to process the signals received into maps of the spatial localization of cavitation activity, with reduced computing times with respect to the time-domain approach, and to take advantage of frequency selectivity for cavitation regime characterization. The approaches proposed have been mainly nonadaptive, and these have suffered from low resolution and contrast, due to the many reconstruction artifacts. Inspired by the array-processing literature and in the context of passive ultrasound imaging of cavitation, we propose here a robust estimation of the second-order statistics of data through spatial covariance matrices in the FD or cross-spectral density matrices (CSMs). The benefits of such formalism are illustrated using advanced reconstruction algorithms, such as the robust Capon beamformer, the Pisarenko class beamformer, and the multiple signal classification approach. Through both simulations and experiments in a water tank, we demonstrate that enhanced localization of cavitation activity (i.e., improved resolution and contrast with respect to nonadaptive approaches) is compatible with the rapid and frequency-selective approaches of the FD. Robust estimation of the CSM and the derived adaptive beamformers paves the way to the development of powerful passive ultrasound imaging tools.
Collapse
|
19
|
Guo Y, Lee H, Fang Z, Velalopoulou A, Kim J, Thomas MB, Liu J, Abramowitz RG, Kim Y, Coskun AF, Krummel DP, Sengupta S, MacDonald TJ, Arvanitis C. Single-cell analysis reveals effective siRNA delivery in brain tumors with microbubble-enhanced ultrasound and cationic nanoparticles. SCIENCE ADVANCES 2021; 7:7/18/eabf7390. [PMID: 33931452 PMCID: PMC8087400 DOI: 10.1126/sciadv.abf7390] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/12/2021] [Indexed: 05/08/2023]
Abstract
RNA-based therapies offer unique advantages for treating brain tumors. However, tumor penetrance and uptake are hampered by RNA therapeutic size, charge, and need to be "packaged" in large carriers to improve bioavailability. Here, we have examined delivery of siRNA, packaged in 50-nm cationic lipid-polymer hybrid nanoparticles (LPHs:siRNA), combined with microbubble-enhanced focused ultrasound (MB-FUS) in pediatric and adult preclinical brain tumor models. Using single-cell image analysis, we show that MB-FUS in combination with LPHs:siRNA leads to more than 10-fold improvement in siRNA delivery into brain tumor microenvironments of the two models. MB-FUS delivery of Smoothened (SMO) targeting siRNAs reduces SMO protein production and markedly increases tumor cell death in the SMO-activated medulloblastoma model. Moreover, our analysis reveals that MB-FUS and nanoparticle properties can be optimized to maximize delivery in the brain tumor microenvironment, thereby serving as a platform for developing next-generation tunable delivery systems for RNA-based therapy in brain tumors.
Collapse
Affiliation(s)
- Yutong Guo
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hohyun Lee
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Zhou Fang
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anastasia Velalopoulou
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jinhwan Kim
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Midhun Ben Thomas
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jingbo Liu
- Department of Pediatrics, Aflac Cancer and Blood, Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan G Abramowitz
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - YongTae Kim
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Aflac Cancer and Blood, Emory University School of Medicine, Atlanta, GA, USA
| | - Costas Arvanitis
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
20
|
Kamimura HAS, Wu SY, Grondin J, Ji R, Aurup C, Zheng W, Heidmann M, Pouliopoulos AN, Konofagou EE. Real-Time Passive Acoustic Mapping Using Sparse Matrix Multiplication. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:164-177. [PMID: 32746182 PMCID: PMC7770101 DOI: 10.1109/tuffc.2020.3001848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Passive acoustic mapping enables the spatiotemporal monitoring of cavitation with circulating microbubbles during focused ultrasound (FUS)-mediated blood-brain barrier opening. However, the computational load for processing large data sets of cavitation maps or more complex algorithms limit the visualization in real-time for treatment monitoring and adjustment. In this study, we implemented a graphical processing unit (GPU)-accelerated sparse matrix-based beamforming and time exposure acoustics in a neuronavigation-guided ultrasound system for real-time spatiotemporal monitoring of cavitation. The system performance was tested in silico through benchmarking, in vitro using nonhuman primate (NHP) and human skull specimens, and demonstrated in vivo in NHPs. We demonstrated the stability of the cavitation map for integration times longer than 62.5 [Formula: see text]. A compromise between real-time displaying and cavitation map quality obtained from beamformed RF data sets with a size of 2000 ×128 ×30 (axial [Formula: see text]) was achieved for an integration time of [Formula: see text], which required a computational time of 0.27 s (frame rate of 3.7 Hz) and could be displayed in real-time between pulses at PRF = 2 Hz. Our benchmarking tests show that the GPU sparse-matrix algorithm processed the RF data set at a computational rate of [Formula: see text]/pixel/sample, which enables adjusting the frame rate and the integration time as needed. The neuronavigation system with real-time implementation of cavitation mapping facilitated the localization of the cavitation activity and helped to identify distortions due to FUS phase aberration. The in vivo test of the method demonstrated the feasibility of GPU-accelerated sparse matrix computing in a close to a clinical condition, where focus distortions exemplify problems during treatment. These experimental conditions show the need for spatiotemporal monitoring of cavitation with real-time capability that enables the operator to correct or halt the sonication in case substantial aberrations are observed.
Collapse
|
21
|
Sun T, Dasgupta A, Zhao Z, Nurunnabi M, Mitragotri S. Physical triggering strategies for drug delivery. Adv Drug Deliv Rev 2020; 158:36-62. [PMID: 32589905 DOI: 10.1016/j.addr.2020.06.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Physically triggered systems hold promise for improving drug delivery by enhancing the controllability of drug accumulation and release, lowering non-specific toxicity, and facilitating clinical translation. Several external physical stimuli including ultrasound, light, electric fields and magnetic fields have been used to control drug delivery and they share some common features such as spatial targeting, spatiotemporal control, and minimal invasiveness. At the same time, they possess several distinctive features in terms of interactions with biological entities and/or the extent of stimulus response. Here, we review the key advances of such systems with a focus on discussing their physical mechanisms, the design rationales, and translational challenges.
Collapse
Affiliation(s)
- Tao Sun
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anshuman Dasgupta
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, TX 79902, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
22
|
A new safety index based on intrapulse monitoring of ultra-harmonic cavitation during ultrasound-induced blood-brain barrier opening procedures. Sci Rep 2020; 10:10088. [PMID: 32572103 PMCID: PMC7308405 DOI: 10.1038/s41598-020-66994-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/29/2020] [Indexed: 11/25/2022] Open
Abstract
Ultrasound-induced blood-brain barrier (BBB) opening using microbubbles is a promising technique for local delivery of therapeutic molecules into the brain. The real-time control of the ultrasound dose delivered through the skull is necessary as the range of pressure for efficient and safe BBB opening is very narrow. Passive cavitation detection (PCD) is a method proposed to monitor the microbubble activity during ultrasound exposure. However, there is still no consensus on a reliable safety indicator able to predict potential damage in the brain. Current approaches for the control of the beam intensity based on PCD employ a full-pulse analysis and may suffer from a lack of sensitivity and poor reaction time. To overcome these limitations, we propose an intra-pulse analysis to monitor the evolution of the frequency content during ultrasound bursts. We hypothesized that the destabilization of microbubbles exposed to a critical level of ultrasound would result in the instantaneous generation of subharmonic and ultra-harmonic components. This specific signature was exploited to define a new sensitive indicator of the safety of the ultrasound protocol. The approach was validated in vivo in rats and non-human primates using a retrospective analysis. Our results demonstrate that intra-pulse monitoring was able to exhibit a sudden appearance of ultra-harmonics during the ultrasound excitation pulse. The repeated detection of such a signature within the excitation pulse was highly correlated with the occurrence of side effects such as hemorrhage and edema. Keeping the acoustic pressure at levels where no such sign of microbubble destabilization occurred resulted in safe BBB openings, as shown by MR images and gross pathology. This new indicator should be more sensitive than conventional full-pulse analysis and can be used to distinguish between potentially harmful and safe ultrasound conditions in the brain with very short reaction time.
Collapse
|
23
|
Smith CAB, Coussios CC. Spatiotemporal Assessment of the Cellular Safety of Cavitation-Based Therapies by Passive Acoustic Mapping. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1235-1243. [PMID: 32111455 DOI: 10.1016/j.ultrasmedbio.2020.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/09/2019] [Accepted: 01/13/2020] [Indexed: 05/09/2023]
Abstract
Many useful therapeutic bio-effects can be generated using ultrasound-induced cavitation. However, cavitation is also capable of causing unwanted cellular and vascular damage, which should be monitored to ensure treatment safety. In this work, the unique opportunity provided by passive acoustic mapping (PAM) to quantify cavitation dose across an entire volume of interest during therapy is utilised to provide setup-independent measures of spatially localised cavitation dose. This spatiotemporally quantifiable cavitation dose is then related to the level of cellular damage generated. The cavitation-mediated destruction of equine red blood cells mixed with one of two types of cavitation nuclei at a variety of concentrations is investigated. The blood is placed within a 0.5-MHz ultrasound field and exposed to a range of peak rarefactional pressures up to 2 MPa, with 50 to 50,000 cycle pulses maintaining a 5% duty cycle. Two co-planar linear arrays at 90° to each other are used to generate 400-µm-resolution frequency domain robust capon beamforming PAM maps, which are then used to generate estimates of cavitation dose. A relationship between this cavitation dose and the levels of haemolysis generated was found which was comparable regardless of the applied acoustic pressure, pulse length, cavitation agent type or concentration used. PAM was then used to monitor cellular damage in multiple locations within a tissue phantom simultaneously, with the damage-cavitation dose relationship being similar for the two experimental models tested. These results lay the groundwork for this method to be applied to other measures of safety, allowing for improved ultrasound monitoring of cavitation-based therapies.
Collapse
Affiliation(s)
- Cameron A B Smith
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Constantin C Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
24
|
Schoen S, Arvanitis CD. Heterogeneous Angular Spectrum Method for Trans-Skull Imaging and Focusing. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:1605-1614. [PMID: 31751231 PMCID: PMC10710012 DOI: 10.1109/tmi.2019.2953872] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Ultrasound, alone or in concert with circulating microbubble contrast agents, has emerged as a promising modality for therapy and imaging of brain diseases. While this has become possible due to advancements in aberration correction methods, a range of applications, including adaptive focusing and tracking of the microbubble dynamics through the human skull, may benefit from even more computationally efficient methods to account for skull aberrations. Here, we derive a general method for the angular spectrum approach (ASA) in a heterogeneous medium, based on a numerical marching scheme to approximate the full implicit solution. We then demonstrate its functionality with simulations for (human) skull-related aberration correction and trans-skull passive acoustic mapping. Our simulations show that the general solution provides accurate trans-skull focusing as compared to the uncorrected case (error in focal point location of 1.0 ± 0.4 mm vs 2.2 ± 0.7 mm) for clinically relevant frequencies (0.25-1.5MHz), apertures (50-100 mm), and targets, with peak focal pressures approximately 30 ± 17% of the free field case, with the effects of skull attenuation and amplitude shading included. In the case of source localization, our method leads to an average of 75% error reduction (from 2.9 ± 1.8 mm to 0.7 ± 0.5 mm) and 40-60% increase in peak intensity, evaluated over the range of frequencies (0.4-1.2 MHz), apertures (50-100 mm), and point source locations (40 mm by 50 mm grid) as compared to the homogeneous medium ASA. Overall, total computation times for both focusing and point source localization of the order milliseconds (166 ± 37 ms, compared with 44 ± 4 ms for the homogeneous ASA formulation) can be attained with this approach. Collectively our findings indicate that the proposed phase correction method based on the ASA could provide a computationally efficient and accurate method for trans-skull transmit focusing and imaging of point scatterers, potentially opening new possibilities for treatment and diagnosis of brain diseases.
Collapse
|
25
|
Pouliopoulos AN, Wu SY, Burgess MT, Karakatsani ME, Kamimura HAS, Konofagou EE. A Clinical System for Non-invasive Blood-Brain Barrier Opening Using a Neuronavigation-Guided Single-Element Focused Ultrasound Transducer. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:73-89. [PMID: 31668690 PMCID: PMC6879801 DOI: 10.1016/j.ultrasmedbio.2019.09.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 05/07/2023]
Abstract
Focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening is currently being investigated in clinical trials. Here, we describe a portable clinical system with a therapeutic transducer suitable for humans, which eliminates the need for in-line magnetic resonance imaging (MRI) guidance. A neuronavigation-guided 0.25-MHz single-element FUS transducer was developed for non-invasive clinical BBB opening. Numerical simulations and experiments were performed to determine the characteristics of the FUS beam within a human skull. We also validated the feasibility of BBB opening obtained with this system in two non-human primates using U.S. Food and Drug Administration (FDA)-approved treatment parameters. Ultrasound propagation through a human skull fragment caused 44.4 ± 1% pressure attenuation at a normal incidence angle, while the focal size decreased by 3.3 ± 1.4% and 3.9 ± 1.8% along the lateral and axial dimension, respectively. Measured lateral and axial shifts were 0.5 ± 0.4 mm and 2.1 ± 1.1 mm, while simulated shifts were 0.1 ± 0.2 mm and 6.1 ± 2.4 mm, respectively. A 1.5-MHz passive cavitation detector transcranially detected cavitation signals of Definity microbubbles flowing through a vessel-mimicking phantom. T1-weighted MRI confirmed a 153 ± 5.5 mm3 BBB opening in two non-human primates at a mechanical index of 0.4, using Definity microbubbles at the FDA-approved dose for imaging applications, without edema or hemorrhage. In conclusion, we developed a portable system for non-invasive BBB opening in humans, which can be achieved at clinically relevant ultrasound exposures without the need for in-line MRI guidance. The proposed FUS system may accelerate the adoption of non-invasive FUS-mediated therapies due to its fast application, low cost and portability.
Collapse
Affiliation(s)
| | - Shih-Ying Wu
- Department of Biomedical Engineering, Columbia University, New York City, New York, USA
| | - Mark T Burgess
- Department of Biomedical Engineering, Columbia University, New York City, New York, USA
| | | | - Hermes A S Kamimura
- Department of Biomedical Engineering, Columbia University, New York City, New York, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York City, New York, USA; Department of Radiology, Columbia University, New York City, New York, USA.
| |
Collapse
|
26
|
Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer 2020; 20:26-41. [PMID: 31601988 PMCID: PMC8246629 DOI: 10.1038/s41568-019-0205-x] [Citation(s) in RCA: 1017] [Impact Index Per Article: 203.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
For a blood-borne cancer therapeutic agent to be effective, it must cross the blood vessel wall to reach cancer cells in adequate quantities, and it must overcome the resistance conferred by the local microenvironment around cancer cells. The brain microenvironment can thwart the effectiveness of drugs against primary brain tumours as well as brain metastases. In this Review, we highlight the cellular and molecular components of the blood-brain barrier (BBB), a specialized neurovascular unit evolved to maintain brain homeostasis. Tumours are known to compromise the integrity of the BBB, resulting in a vasculature known as the blood-tumour barrier (BTB), which is highly heterogeneous and characterized by numerous distinct features, including non-uniform permeability and active efflux of molecules. We discuss the challenges posed by the BBB and BTB for drug delivery, how multiple cell types dictate BBB function and the role of the BTB in disease progression and treatment. Finally, we highlight emerging molecular, cellular and physical strategies to improve drug delivery across the BBB and BTB and discuss their impact on improving conventional as well as emerging treatments, such as immune checkpoint inhibitors and engineered T cells. A deeper understanding of the BBB and BTB through the application of single-cell sequencing and imaging techniques, and the development of biomarkers of BBB integrity along with systems biology approaches, should enable new personalized treatment strategies for primary brain malignancies and brain metastases.
Collapse
Affiliation(s)
- Costas D Arvanitis
- School of Mechanical Engineering, Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Gino B Ferraro
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|