1
|
Lin Y, Su R, Huang Y. Thermo-responsive Fluorescent Nanopolymer Delivery Platform for Treatment of Diffuse Large B-cell Lymphoma (DLBCL). J Fluoresc 2025:10.1007/s10895-024-04092-y. [PMID: 39786693 DOI: 10.1007/s10895-024-04092-y] [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: 09/30/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a highly aggressive diffuse malignant proliferative disease of the lymphatic system. Patients usually present with progressive lymph node enlargement and/or extra-lymph node lesions and require early treatment upon diagnosis. Most of the patients are in stage III or IV at the time of diagnosis and about 40% of the patients are difficult to cure. In this study, we developed a novel drug delivery platform using poly(ε-caprolactone)-tetraphenylethylene (PCL-TPE). This fluorescent polymer, combined with a solid-state dye, exhibits temperature-dependent fluorescence. We loaded the synthesized ethyl 2-amino-4-(4-methylthiophen-2-yl)-5,6,7,8-tetrahydro-2 H-chromene-3-carboxylate (compound 1) onto PCL-TPE, creating PCL-TPE@1. By treating human lymphoma cells, we found that PCL-TPE@1 was able to effectively reduce the migratory ability of lymphoma cells by inhibiting the expression of PRMT6. This platform is easy to synthesize, offers thermo-responsive fluorescence, flexibility, and biocompatibility, making it suitable for biomedical drug delivery and smart devices.
Collapse
Affiliation(s)
- Yun Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Rui Su
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Yun Huang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, China.
| |
Collapse
|
2
|
Fan CH, Yeh CK. Theranostic nanomaterials for intervention of the blood–brain barrier. THERANOSTICS NANOMATERIALS IN DRUG DELIVERY 2025:395-410. [DOI: 10.1016/b978-0-443-22044-9.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Epstein JE, Pople CB, Meng Y, Lipsman N. An update on the role of focused ultrasound in neuro-oncology. Curr Opin Neurol 2024; 37:682-692. [PMID: 39498847 DOI: 10.1097/wco.0000000000001314] [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: 11/07/2024]
Abstract
PURPOSE OF REVIEW Brain tumor treatment presents challenges for patients and clinicians, with prognosis for many of the most common brain tumors being poor. Focused ultrasound (FUS) can be deployed in several ways to circumvent these challenges, including the need to penetrate the blood-brain barrier and spare healthy brain tissue. This article reviews current FUS applications within neuro-oncology, emphasizing ongoing or recently completed clinical trials. RECENT FINDINGS Most clinical interest in FUS for neuro-oncology remains focused on exploring BBB disruption to enhance the delivery of standard-of-care therapeutics. More recently, the application of FUS for radiosensitization, liquid biopsy, and sonodynamic therapy is garnering increased clinical attention to assist in tumor ablation, early detection, and phenotypic diagnosis. Preclinical studies show encouraging data for the immunomodulatory effects of FUS, but these findings have yet to be tested clinically. SUMMARY FUS is a burgeoning area of neuro-oncology research. Data from several forthcoming large clinical trials should help clarify its role in neuro-oncology care.
Collapse
Affiliation(s)
- Jordan E Epstein
- Harquail Centre for Neuromodulation
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Christopher B Pople
- Harquail Centre for Neuromodulation
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Ying Meng
- Harquail Centre for Neuromodulation
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Ontario, Canada
| |
Collapse
|
4
|
Lea-Banks H, Chauhan N, Hynynen K. Investigating the hypotensive effect of focused ultrasound neuromodulation and barbiturate-loaded nanodroplets in healthy and hypertensive rats. Brain Stimul 2024; 17:1317-1327. [PMID: 39643112 DOI: 10.1016/j.brs.2024.12.002] [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: 09/06/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Current strategies for reducing blood pressure (BP) are ineffective and unsafe for many patient populations, including drug-resistant hypertension and during pregnancy. Stimulating the periaqueductal grey (PAG) region has shown promise in treating drug-resistant hypertension in patients using deep brain stimulation. OBJECTIVE To develop a minimally invasive neuromodulation technique for the sustained treatment of hypertension. METHODS We have investigated BP reduction using focused ultrasound (FUS) (540 kHz) and anesthetic-loaded ultrasound-responsive nanodroplets to deliver pentobarbital to the PAG in normotensive (N = 27) and hypertensive (N = 20) male and female rats. BP, heart rate and plasma hormone content were collected before and after FUS exposure, and neuronal activity was mapped in the PAG using C-Fos and neuron subtype staining. Cavitation activity was monitored by detecting acoustic emissions from vaporizing nanodroplets, and neuromodulation was verified with immunohistochemistry. RESULTS Systolic and diastolic BP were reduced for 6 h following a single sonication of the PAG (-37/-28 mmHg systolic/diastolic), and the offline effect was extended to 4 days with consecutive sonications combined with systemically injected pentobarbital-loaded nanodroplets. In contrast, FUS applied to the frontal cortex had no effect on BP. Immunohistochemistry revealed stimulation of inhibitory neurons in the PAG region, indicating that the hypotensive effect was associated with a GABAergic pathway. The acoustic emissions from vaporizing droplets were found to correlate with neuron activity and change in BP, offering the potential for real-time treatment monitoring using ultrasound. CONCLUSIONS This work has implications for developing a new treatment for hypertension that has greater safety and broader applicability for vulnerable patient populations.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Imaging, University of Toronto, Toronto, Canada.
| | - Neha Chauhan
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
5
|
Glickstein B, Shaul O, Ilovitsh T. Rationally Designed Acoustic Holograms for Uniform Nanodroplet-Mediated Tissue Ablation. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1606-1615. [PMID: 39352816 DOI: 10.1109/tuffc.2024.3471873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Nanodroplets (NDs) are phase-changing agents that have shown great potential for ultrasound (US) applications. When US is applied, NDs can undergo a phase transition into gas bubbles, enabling cavitation that can be used to reduce the pressure threshold required for mechanical ablation of tissues. Effective tissue fractionation depends on precise vaporization to achieve uniform and predictable bubble formation. This study aimed to optimize ND vaporization using acoustic holograms for improved ND-mediated histotripsy. Tissue ablation was conducted using a two-step approach, where a rotating imaging probe was used for ND vaporization followed by low-frequency US for detonation. We developed and validated three distinct acoustic hologram patterns targeting different regions within a circular area through simulations and experiments. Using custom-made gelatin phantoms designed for optimal ND vaporization imaging, the superpositioned patterns demonstrated significantly more uniform ND vaporization compared to standard single-focus steering, with ND coverage reaching % for the optimized vaporization approach versus % for the single focus steering. Ex vivo chicken liver experiments confirmed the enhanced efficiency of the optimized approach, resulting in significantly larger and more uniform lesion areas. Lesion areas generated by 120 s of treatment reached mm2 compared to mm2 for the standard approach, a 5.1-fold increase. These findings suggest that using acoustic holograms can improve ND vaporization uniformity and enhance the homogeneity of tissue fractionation, thereby potentially enhancing therapeutic outcomes.
Collapse
|
6
|
Dauba A, Spitzlei C, Bautista KJB, Jourdain L, Selingue E, VanTreeck KE, Mattern JA, Denis C, Ouldali M, Arteni AA, Truillet C, Larrat B, Tsuruta J, Durham PG, Papadopoulou V, Dayton PA, Tsapis N, Novell A. Low-boiling-point perfluorocarbon nanodroplets for adaptable ultrasound-induced blood-brain barrier opening. J Control Release 2024; 376:441-456. [PMID: 39419451 DOI: 10.1016/j.jconrel.2024.10.023] [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: 06/26/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Low-boiling point perfluorocarbon nanodroplets (NDs) are valued as effective sonosensitive agents, encapsulating a liquid perfluorocarbon that would instantaneously vaporize at body temperature without the NDs shell. Those NDs have been explored for both therapeutic and diagnostic purposes. Here, phospholipid-shelled nanodroplets containing octafluoropropane (C3F8) or decafluorobutane (C4F10) formed by condensation of microbubbles were thoroughly characterized before blood-brain (BBB) permeabilization. Transmission electron microscopy (TEM) and cryo-TEM were employed to confirm droplet formation while providing high-resolution insights into the droplet surface and lipid arrangement assessed from electron density observation after condensation. The vaporization threshold of NDs was determined with a high-speed camera, and the frequency signal emitted by the freshly vaporized bubbles was analyzed using cavitation detection. C3F8 NDs exhibited vaporization at 0.3 MPa (f0 = 1.5 MHz, 50 cycles), and emitted signals at 2 f0 and 1.5 f0 from 0.45 MPa onwards (f0 = 1.5 MHz, 50 cycles), while broadband noise was measured starting from 0.55 MPa. NDs with the higher boiling point C4F10 vaporized at 1.15 MPa and emitted signals at 2 f0 from 0.65 MPa and 1.5 f0 from 0.9 MPa, while broadband noise was detected starting from 0.95 MPa. Both ND formulations were used to permeabilize the BBB in healthy mice using tailored ultrasound sequences, allowing for the identification of optimal applications for each NDs type. C3F8 NDs proved suitable and safe for permeabilizing a large area, potentially the entire brain, at low acoustic pressure. Meanwhile, C4F10 droplets facilitated very localized (400 μm isotropic) permeabilization at higher pressure. This study prompts a closer examination of the structural rearrangements occurring during the condensation of microbubbles into NDs and highlights the potential to tailor solutions for different brain pathologies by choosing the composition of the NDs and adjusting the ultrasound sequence.
Collapse
Affiliation(s)
- Ambre Dauba
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Claire Spitzlei
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France; Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Kathlyne Jayne B Bautista
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Laurène Jourdain
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Erwan Selingue
- Université Paris-Saclay, CEA, CNRS, Baobab, NeuroSpin, Gif-sur-Yvette 91191, France
| | - Kelly E VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jacob A Mattern
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Caroline Denis
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Malika Ouldali
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Cryo-electron Microscopy Facility, CRYOEM-Gif, 91198 Gif-sur-Yvette, France
| | - Ana-Andreea Arteni
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Cryo-electron Microscopy Facility, CRYOEM-Gif, 91198 Gif-sur-Yvette, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, Baobab, NeuroSpin, Gif-sur-Yvette 91191, France
| | - James Tsuruta
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Phillip G Durham
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA; Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, North Carolina, USA
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91401, France.
| |
Collapse
|
7
|
Wu Q, Choi V, Bau L, Carugo D, Evans ND, Stride E. Investigation of Ultrasound Mediated Extravasation of a Model Drug by Perfluorobutane Nanodroplets. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1573-1584. [PMID: 39060156 DOI: 10.1016/j.ultrasmedbio.2024.06.016] [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: 02/09/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE Perfluorocarbon nanodroplets (NDs) have been widely investigated as both diagnostic and therapeutic agents. There remains, however, a challenge in generating NDs that do not vaporize spontaneously but can be activated at ultrasound pressures that do not produce unwanted bioeffects. In previous work, it has been shown that phospholipid-coated perfluorobutane (PFB) NDs can potentially overcome this challenge. The aim of this study was to investigate whether these NDs can promote drug delivery. METHODS A combination of high-speed optical imaging and passive cavitation detection was used to study the acoustic properties of the PFB-NDs in a tissue mimicking phantom. PFB-NDs were exposed to ultrasound at frequencies from 0.5 to 1.5 MHz and peak negative pressures from 0.5 to 3.5 MPa. In addition, the penetration depth of two model drugs (Nile Red and 200 nm diameter fluorescent polymer spheres) into the phantom was measured. RESULTS PFB NDs were found to be stable in aqueous suspension at both 4°C and 37°C; their size remaining unchanged at 215 ± 11 nm over 24 h. Penetration of both model drugs in the phantom was found to increase with increasing ultrasound peak negative pressure and decreasing frequency and was found to be positively correlated with the energy of acoustic emissions. Extravasation depths >1 mm were observed at 0.5 MHz with pressures <1 MPa. CONCLUSION The results of the study thus suggest that PFB NDs can be used both as drug carriers and as nuclei for cavitation to enhance drug delivery without the need for high intensity ultrasound.
Collapse
Affiliation(s)
- Qiang Wu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Victor Choi
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Dario Carugo
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Nicholas D Evans
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research Group, University of Southampton, Southampton, UK; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK; Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Wilson MG, Riis TS, Kubanek J. Controlled ultrasonic interventions through the human skull. Front Hum Neurosci 2024; 18:1412921. [PMID: 38979100 PMCID: PMC11228146 DOI: 10.3389/fnhum.2024.1412921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Transcranial focused ultrasound enables precise and non-invasive manipulations of deep brain circuits in humans, promising to provide safe and effective treatments of various neurological and mental health conditions. Ultrasound focused to deep brain targets can be used to modulate neural activity directly or localize the release of psychoactive drugs. However, these applications have been impeded by a key barrier-the human skull, which attenuates ultrasound strongly and unpredictably. To address this issue, we have developed an ultrasound-based approach that directly measures and compensates for the ultrasound attenuation by the skull. No additional skull imaging, simulations, assumptions, or free parameters are necessary; the method measures the attenuation directly by emitting a pulse of ultrasound from an array on one side of the head and measuring with an array on the opposite side. Here, we apply this emerging method to two primary future uses-neuromodulation and local drug release. Specifically, we show that the correction enables effective stimulation of peripheral nerves and effective release of propofol from nanoparticle carriers through an ex vivo human skull. Neither application was effective without the correction. Moreover, the effects show the expected dose-response relationship and targeting specificity. This article highlights the need for precise control of ultrasound intensity within the skull and provides a direct and practical approach for addressing this lingering barrier.
Collapse
Affiliation(s)
- Matthew G Wilson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Thomas S Riis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Jan Kubanek
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
9
|
Wilson MG, Parikh A, Dara A, Beaver AS, Kubanek J. Targeted drug release from stable and safe ultrasound-sensitive nanocarriers. Front Mol Biosci 2024; 11:1408767. [PMID: 38962281 PMCID: PMC11219560 DOI: 10.3389/fmolb.2024.1408767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/17/2024] [Indexed: 07/05/2024] Open
Abstract
Targeted delivery of medication has the promise of increasing the effectiveness and safety of current systemic drug treatments. Focused ultrasound is emerging as noninvasive and practical energy for targeted drug release. However, it has yet to be determined which nanocarriers and ultrasound parameters can provide both effective and safe release. Perfluorocarbon nanodroplets have the potential to achieve these goals, but current approaches have either been effective or safe, but not both. We found that nanocarriers with highly stable perfluorocarbon cores mediate effective drug release so long as they are activated by ultrasound of sufficiently low frequency. We demonstrate a favorable safety profile of this formulation in a non-human primate. To facilitate translation of this approach into humans, we provide an optimized method for manufacturing the nanocarriers. This study provides a recipe and release parameters for effective and safe drug release from nanoparticle carriers in the body part specified by focused ultrasonic waves.
Collapse
Affiliation(s)
- Matthew G. Wilson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | | | | | | | | |
Collapse
|
10
|
Meng Y, Kalia LV, Kalia SK, Hamani C, Huang Y, Hynynen K, Lipsman N, Davidson B. Current Progress in Magnetic Resonance-Guided Focused Ultrasound to Facilitate Drug Delivery across the Blood-Brain Barrier. Pharmaceutics 2024; 16:719. [PMID: 38931843 PMCID: PMC11206305 DOI: 10.3390/pharmaceutics16060719] [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: 03/19/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
This review discusses the current progress in the clinical use of magnetic resonance-guided focused ultrasound (MRgFUS) and other ultrasound platforms to transiently permeabilize the blood-brain barrier (BBB) for drug delivery in neurological disorders and neuro-oncology. Safety trials in humans have followed on from extensive pre-clinical studies, demonstrating a reassuring safety profile and paving the way for numerous translational clinical trials in Alzheimer's disease, Parkinson's disease, and primary and metastatic brain tumors. Future directions include improving ultrasound delivery devices, exploring alternative delivery approaches such as nanodroplets, and expanding the application to other neurological conditions.
Collapse
Affiliation(s)
- Ying Meng
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Lorraine V. Kalia
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Suneil K. Kalia
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
- Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), University Health Network, Toronto, ON M5T 1M8, Canada
- KITE Research Institute, University Health Network, Toronto, ON M5G 2A2, Canada
| | - Clement Hamani
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Yuexi Huang
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | | | - Nir Lipsman
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Benjamin Davidson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
11
|
Pan X, Huang W, Nie G, Wang C, Wang H. Ultrasound-Sensitive Intelligent Nanosystems: A Promising Strategy for the Treatment of Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303180. [PMID: 37871967 DOI: 10.1002/adma.202303180] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/26/2023] [Indexed: 10/25/2023]
Abstract
Neurological diseases are a major global health challenge, affecting hundreds of millions of people worldwide. Ultrasound therapy plays an irreplaceable role in the treatment of neurological diseases due to its noninvasive, highly focused, and strong tissue penetration capabilities. However, the complexity of brain and nervous system and the safety risks associated with prolonged exposure to ultrasound therapy severely limit the applicability of ultrasound therapy. Ultrasound-sensitive intelligent nanosystems (USINs) are a novel therapeutic strategy for neurological diseases that bring greater spatiotemporal controllability and improve safety to overcome these challenges. This review provides a detailed overview of therapeutic strategies and clinical advances of ultrasound in neurological diseases, focusing on the potential of USINs-based ultrasound in the treatment of neurological diseases. Based on the physical and chemical effects induced by ultrasound, rational design of USINs is a prerequisite for improving the efficacy of ultrasound therapy. Recent developments of ultrasound-sensitive nanocarriers and nanoagents are systemically reviewed. Finally, the challenges and developing prospects of USINs are discussed in depth, with a view to providing useful insights and guidance for efficient ultrasound treatment of neurological diseases.
Collapse
Affiliation(s)
- Xueting Pan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Wenping Huang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing, 100850, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
12
|
Wilson MG, Webb TD, Odéen H, Kubanek J. Remotely controlled drug release in deep brain regions of non-human primates. J Control Release 2024; 369:775-785. [PMID: 38604386 PMCID: PMC11111335 DOI: 10.1016/j.jconrel.2024.04.013] [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: 10/05/2023] [Revised: 03/18/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
Many areas of science and medicine would benefit from selective release of drugs in specific regions. Nanoparticle drug carriers activated by focused ultrasound-remotely applied, depth-penetrating energy-may provide such selective interventions. Here, we developed stable, ultrasound-responsive nanoparticles that can be used to release drugs effectively and safely in non-human primates. The nanoparticles were used to release propofol in deep brain visual regions. The release reversibly modulated the subjects' visual choice behavior and was specific to the targeted region and to the released drug. Gadolinium-enhanced MR imaging suggested an intact blood-brain barrier. Blood draws showed normal clinical chemistry and hematology. In summary, this study provides a safe and effective approach to release drugs on demand in selected deep brain regions at levels sufficient to modulate behavior.
Collapse
Affiliation(s)
- Matthew G Wilson
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Taylor D Webb
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Henrik Odéen
- Department of Radiology and Imaging Sciences, University of Utah, 729 Arapeen Drive, Salt Lake City, UT 84108, USA
| | - Jan Kubanek
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA.
| |
Collapse
|
13
|
Schwinghamer K, Line S, Tesar DB, Miller DW, Sreedhara A, Siahaan TJ. Selective Uptake of Macromolecules to the Brain in Microfluidics and Animal Models Using the HAVN1 Peptide as a Blood-Brain Barrier Modulator. Mol Pharm 2024; 21:1639-1652. [PMID: 38395041 PMCID: PMC10984760 DOI: 10.1021/acs.molpharmaceut.3c00775] [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] [Indexed: 02/25/2024]
Abstract
Monoclonal antibodies (mAbs) possess favorable pharmacokinetic properties, high binding specificity and affinity, and minimal off-target effects, making them promising therapeutic agents for central nervous system (CNS) disorders. However, their development as effective therapeutic and diagnostic agents for brain disorders is hindered by their limited ability to efficiently penetrate the blood-brain barrier (BBB). Therefore, it is crucial to develop efficient delivery methods that enhance the penetration of antibodies into the brain. Previous studies have demonstrated the potential of cadherin-derived peptides (i.e., ADTC5, HAVN1 peptides) as BBB modulators (BBBMs) to increase paracellular porosities for penetration of molecules across the BBB. Here, we test the effectiveness of the leading BBBM peptide, HAVN1 (Cyclo(1,6)SHAVSS), in enhancing the permeation of various monoclonal antibodies through the BBB using both in vitro and in vivo systems. In vitro, HAVN1 has been shown to increase the permeability of fluorescently labeled macromolecules, such as a 70 kDa dextran, 50 kDa Fab1, and 150 kDa mAb1, by 4- to 9-fold in a three-dimensional blood-brain barrier (3D-BBB) microfluidics model using a human BBB endothelial cell line (i.e., hCMEC/D3). HAVN1 was selective in modulating the BBB endothelial cell, compared to the pulmonary vascular endothelial (PVE) cell barrier. Co-administration of HAVN1 significantly improved brain depositions of mAb1, mAb2, and Fab1 in C57BL/6 mice after 15 min in the systemic circulation. Furthermore, HAVN1 still significantly enhanced brain deposition of mAb2 when it was administered 24 h after the administration of the mAb. Lastly, we observed that multiple doses of HAVN1 may have a cumulative effect on the brain deposition of mAb2 within a 24-h period. These findings offer promising insights into optimizing HAVN1 and mAb dosing regimens to control or modulate mAb brain deposition for achieving desired mAb dose in the brain to provide its therapeutic effects.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, KS 66047, USA
| | - Stacey Line
- Department of Pharmacology and Therapeutics, University of Manitoba, 753 McDermot Avenue Winnipeg, MB, R3E 0T6, Canada
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Donald W. Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, 753 McDermot Avenue Winnipeg, MB, R3E 0T6, Canada
| | - Alavattam Sreedhara
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, 2093 Constant Ave., Lawrence, KS 66047, USA
| |
Collapse
|
14
|
Durham PG, Butnariu A, Alghorazi R, Pinton G, Krishna V, Dayton PA. Current clinical investigations of focused ultrasound blood-brain barrier disruption: A review. Neurotherapeutics 2024; 21:e00352. [PMID: 38636309 PMCID: PMC11044032 DOI: 10.1016/j.neurot.2024.e00352] [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: 12/15/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/20/2024] Open
Abstract
The blood-brain barrier (BBB) presents a formidable challenge in delivering therapeutic agents to the central nervous system. Ultrasound-mediated BBB disruption has emerged as a promising non-invasive technique to enhance drug delivery to the brain. This manuscript reviews fundamental principles of ultrasound-based techniques and their mechanisms of action in temporarily permeabilizing the BBB. Clinical trials employing ultrasound for BBB disruption are discussed, summarizing diverse applications ranging from the treatment of neurodegenerative diseases to targeted drug delivery for brain tumors. The review also addresses safety considerations, outlining the current understanding of potential risks and mitigation strategies associated with ultrasound exposure, including real-time monitoring and assessment of treatment efficacy. Among the large number of studies, significant successes are highlighted thus providing perspective on the future direction of the field.
Collapse
Affiliation(s)
- Phillip G Durham
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA
| | | | - Rizk Alghorazi
- School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Gianmarco Pinton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA
| | - Vibhor Krishna
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA; School of Medicine, University of North Carolina, Chapel Hill, NC, United States.
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Chapel Hill, NC, USA.
| |
Collapse
|
15
|
Wilson MG, Webb TD, Odéen H, Kubanek J. Remotely controlled drug release in deep brain regions of non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.09.561539. [PMID: 37873134 PMCID: PMC10592699 DOI: 10.1101/2023.10.09.561539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Many areas of science and medicine would benefit from selective release of drugs in specific regions of interest. Nanoparticle drug carriers activated by focused ultrasound-remotely applied, depth-penetrating energy-may provide such selective interventions. Here, we developed stable, ultrasound-responsive nanoparticles that can be used to release drugs effectively and safely in non-human primates. The nanoparticles were used to release propofol in deep brain visual regions. The release reversibly modulated the subjects' visual choice behavior and was specific to the targeted region and to the released drug. Gadolinium-enhanced MRI imaging suggested an intact blood-brain barrier. Blood draws showed normal clinical chemistry and hematology. In summary, this study provides a safe and effective approach to release drugs on demand in selected deep brain regions at levels sufficient to modulate behavior.
Collapse
|
16
|
Pellow C, Jafari Sojahrood A, Zhao X, Kolios MC, Exner AA, Goertz DE. Synchronous Intravital Imaging and Cavitation Monitoring of Antivascular Focused Ultrasound in Tumor Microvasculature Using Monodisperse Low Boiling Point Nanodroplets. ACS NANO 2024; 18:410-427. [PMID: 38147452 PMCID: PMC10786165 DOI: 10.1021/acsnano.3c07711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.
Collapse
Affiliation(s)
- Carly Pellow
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
| | - Amin Jafari Sojahrood
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Xiaoxiao Zhao
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| | - Michael C. Kolios
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Agata A. Exner
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - David E. Goertz
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| |
Collapse
|
17
|
Vlatakis S, Zhang W, Thomas S, Cressey P, Moldovan AC, Metzger H, Prentice P, Cochran S, Thanou M. Effect of Phase-Change Nanodroplets and Ultrasound on Blood-Brain Barrier Permeability In Vitro. Pharmaceutics 2023; 16:51. [PMID: 38258062 PMCID: PMC10818572 DOI: 10.3390/pharmaceutics16010051] [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: 11/02/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Phase-change nanodroplets (PCND;NDs) are emulsions with a perfluorocarbon (PFC) core that undergo acoustic vaporisation as a response to ultrasound (US). Nanodroplets change to microbubbles and cavitate while under the effect of US. This cavitation can apply forces on cell connections in biological barrier membranes, such as the blood-brain barrier (BBB), and trigger a transient and reversible increased permeability to molecules and matter. This study aims to present the preparation of lipid-based NDs and investigate their effects on the brain endothelial cell barrier in vitro. The NDs were prepared using the thin-film hydration method, followed by the PFC addition. They were characterised for size, cavitation (using a high-speed camera), and PFC encapsulation (using FTIR). The bEnd.3 (mouse brain endothelial) cells were seeded onto transwell inserts. Fluorescein with NDs and/or microbubbles were applied on the bEND3 cells and the effect of US on fluorescein permeability was measured. The Live/Dead assay was used to assess the BBB integrity after the treatments. Size and PFC content analysis indicated that the NDs were stable while stored. High-speed camera imaging confirmed that the NDs cavitate after US exposure of 0.12 MPa. The BBB cell model experiments revealed a 4-fold increase in cell membrane permeation after the combined application of US and NDs. The Live/Dead assay results indicated damage to the BBB membrane integrity, but this damage was less when compared to the one caused by microbubbles. This in vitro study shows that nanodroplets have the potential to cause BBB opening in a similar manner to microbubbles. Both cavitation agents caused damage on the endothelial cells. It appears that NDs cause less cell damage compared to microbubbles.
Collapse
Affiliation(s)
- Stavros Vlatakis
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Weiqi Zhang
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Sarah Thomas
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Paul Cressey
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Alexandru Corneliu Moldovan
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Hilde Metzger
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Paul Prentice
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Sandy Cochran
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Maya Thanou
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| |
Collapse
|
18
|
Brans V, Gray MD, Sezgin E, Stride EPJ. Protein-Decorated Microbubbles for Ultrasound-Mediated Cell Surface Manipulation. ACS APPLIED BIO MATERIALS 2023; 6:5746-5758. [PMID: 38048163 PMCID: PMC10731656 DOI: 10.1021/acsabm.3c00861] [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: 09/25/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023]
Abstract
Delivering cargo to the cell membranes of specific cell types in the body is a major challenge for a range of treatments, including immunotherapy. This study investigates employing protein-decorated microbubbles (MBs) and ultrasound (US) to "tag" cellular membranes of interest with a specific protein. Phospholipid-coated MBs were produced and functionalized with a model protein using a metallochelating complex through an NTA(Ni) and histidine residue interaction. Successful "tagging" of the cellular membrane was observed using microscopy in adherent cells and was promoted by US exposure. Further modification of the MB surface to enable selective binding to target cells was then achieved by functionalizing the MBs with a targeting protein (transferrin) that specifically binds to a receptor on the target cell membrane. Attachment and subsequent transfer of material from MBs functionalized with transferrin to the target cells significantly increased, even in the absence of US. This work demonstrates the potential of these MBs as a platform for the noninvasive delivery of proteins to the surface of specific cell types.
Collapse
Affiliation(s)
- Veerle
A. Brans
- Department
of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DL, U.K.
| | - Michael D. Gray
- Department
of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DL, U.K.
| | - Erdinc Sezgin
- Science
for Life Laboratory, Department of Women’s and Children’s
Health, Karolinska Institutet, 17165 Solna, Sweden
| | - Eleanor P. J. Stride
- Department
of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DL, U.K.
| |
Collapse
|
19
|
Schwinghamer K, Siahaan TJ. Enhancing Antibody Exposure in the Central Nervous System: Mechanisms of Uptake, Clearance, and Strategies for Improved Brain Delivery. JOURNAL OF NANOTHERANOSTICS 2023; 4:463-479. [PMID: 39897432 PMCID: PMC11784990 DOI: 10.3390/jnt4040020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Antibodies (mAbs) are attractive molecules for their application as a diagnostic and therapeutic agent for diseases of the central nervous system (CNS). mAbs can be generated to have high affinity and specificity to target molecules in the CNS. Unfortunately, only a very small number of mAbs have been specifically developed and approved for neurological indications. This is primarily attributed to their low exposure within the CNS, hindering their ability to reach and effectively engage their potential targets in the brain. This review discusses aspects of various barriers such as the blood-brain barrier (BBB) and blood-cerebrospinal fluid (CSF) barrier (BCSFB) that regulate the entry and clearance of mAbs into and from the brain. The roles of the glymphatic system on brain exposure and clearance are being described. We also discuss the proposed mechanisms of the uptake of mAbs into the brain and for clearance. Finally, several methods of enhancing the exposure of mAbs in the CNS were discussed, including receptor-mediated transcytosis, osmotic BBB opening, focused ultrasound (FUS), BBB-modulating peptides, and enhancement of mAb brain retention.
Collapse
Affiliation(s)
- Kelly Schwinghamer
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS 66046, USA
| |
Collapse
|
20
|
Purohit MP, Roy KS, Xiang Y, Yu BJ, Azadian MM, Muwanga G, Hart AR, Taoube AK, Lopez DG, Airan RD. Acoustomechanically activatable liposomes for ultrasonic drug uncaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563690. [PMID: 37961368 PMCID: PMC10634775 DOI: 10.1101/2023.10.23.563690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Ultrasound-activatable drug-loaded nanocarriers enable noninvasive and spatiotemporally-precise on-demand drug delivery throughout the body. However, most systems for ultrasonic drug uncaging utilize cavitation or heating as the drug release mechanism and often incorporate relatively exotic excipients into the formulation that together limit the drug-loading potential, stability, and clinical translatability and applicability of these systems. Here we describe an alternate strategy for the design of such systems in which the acoustic impedance and osmolarity of the internal liquid phase of a drug-loaded particle is tuned to maximize ultrasound-induced drug release. No gas phase, cavitation, or medium heating is necessary for the drug release mechanism. Instead, a non-cavitation-based mechanical response to ultrasound mediates the drug release. Importantly, this strategy can be implemented with relatively common pharmaceutical excipients, as we demonstrate here by implementing this mechanism with the inclusion of a few percent sucrose into the internal buffer of a liposome. Further, the ultrasound protocols sufficient for in vivo drug uncaging with this system are achievable with current clinical therapeutic ultrasound systems and with intensities that are within FDA and society guidelines for safe transcranial ultrasound application. Finally, this current implementation of this mechanism should be versatile and effective for the loading and uncaging of any therapeutic that may be loaded into a liposome, as we demonstrate for four different drugs in vitro, and two in vivo. These acoustomechanically activatable liposomes formulated with common pharmaceutical excipients promise a system with high clinical translational potential for ultrasonic drug uncaging of myriad drugs of clinical interest.
Collapse
Affiliation(s)
| | - Kanchan Sinha Roy
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Yun Xiang
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Brenda J. Yu
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Biophysics Program, Stanford University, Stanford, CA, 94305 USA
| | - Matine M. Azadian
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Neurosciences Program, Stanford University, Stanford, CA, 94305 USA
| | - Gabriella Muwanga
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Neurosciences Program, Stanford University, Stanford, CA, 94305 USA
| | - Alex R. Hart
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Chemistry, Stanford University, Stanford, CA, 94305 USA
| | - Ali K. Taoube
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Diego Gomez Lopez
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, TN 37235 USA
| | - Raag D. Airan
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305 USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305 USA
| |
Collapse
|
21
|
Shi S, Ren H, Xie Y, Yu M, Chen Y, Yang L. Engineering advanced nanomedicines against central nervous system diseases. MATERIALS TODAY 2023; 69:355-392. [DOI: 10.1016/j.mattod.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
22
|
Zhang G, Liao C, Hu JR, Hu HM, Lei YM, Harput S, Ye HR. Nanodroplet-Based Super-Resolution Ultrasound Localization Microscopy. ACS Sens 2023; 8:3294-3306. [PMID: 37607403 DOI: 10.1021/acssensors.3c00418] [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] [Indexed: 08/24/2023]
Abstract
Over the past decade, super-resolution ultrasound localization microscopy (SR-ULM) has revolutionized ultrasound imaging with its capability to resolve the microvascular structures below the ultrasound diffraction limit. The introduction of this imaging technique enables the visualization, quantification, and characterization of tissue microvasculature. The early implementations of SR-ULM utilize microbubbles (MBs) that require a long image acquisition time due to the requirement of capturing sparsely isolated microbubble signals. The next-generation SR-ULM employs nanodroplets that have the potential to significantly reduce the image acquisition time without sacrificing the resolution. This review discusses various nanodroplet-based ultrasound localization microscopy techniques and their corresponding imaging mechanisms. A summary is given on the preclinical applications of SR-ULM with nanodroplets, and the challenges in the clinical translation of nanodroplet-based SR-ULM are presented while discussing the future perspectives. In conclusion, ultrasound localization microscopy is a promising microvasculature imaging technology that can provide new diagnostic and prognostic information for a wide range of pathologies, such as cancer, heart conditions, and autoimmune diseases, and enable personalized treatment monitoring at a microlevel.
Collapse
Affiliation(s)
- Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan 430080, People's Republic of China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, People's Republic of China
- Physics for Medicine Paris, Inserm U1273, ESPCI Paris, PSL University, CNRS, Paris 75015, France
| | - Chen Liao
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan 430080, People's Republic of China
- Medical College, Wuhan University of Science and Technology, Wuhan 430065, People's Republic of China
| | - Jun-Rui Hu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Hai-Man Hu
- Department of Electrical and Electronic Engineering, Hubei University of Technology, Wuhan 430068, People's Republic of China
| | - Yu-Meng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan 430080, People's Republic of China
| | - Sevan Harput
- Department of Electrical and Electronic Engineering, London South Bank University, London SE1 0AA, U.K
| | - Hua-Rong Ye
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan 430080, People's Republic of China
| |
Collapse
|
23
|
Ting SG, Lea-Banks H, Hynynen K. Physical Characterization to Improve Scalability and Potential of Anesthetic-Loaded Nanodroplets. Pharmaceutics 2023; 15:2077. [PMID: 37631291 PMCID: PMC10457791 DOI: 10.3390/pharmaceutics15082077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/21/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Drug-loaded perfluorocarbon nanodroplets (NDs) can be activated non-invasively by focused ultrasound (FUS) and allow for precise drug-delivery. Anesthetic-loaded NDs and transcranial FUS have previously achieved targeted neuromodulation. To assess the clinical potential of anesthetic-loaded NDs, in depth physical characterization and investigation of storage strategies and triggered-activation is necessary. Pentobarbital-loaded decafluorobutane nanodroplets (PBNDs) with a Definity-derived lipid shell (237 nm; 4.08 × 109 particles/mL) were fabricated and assessed. Change in droplet stability, concentration, and drug-release efficacy were tested for PBNDs frozen at -80 °C over 4 weeks. PBND diameter and the polydispersity index of thawed droplets remained consistent up to 14 days frozen. Cryo-TEM images revealed NDs begin to lose circularity at 7 days, and by 14 days, perfluorocarbon dissolution and lipid fragmentation occurred. The level of acoustic response and drug release decreases through prolonged storage. PBNDs showed no hemolytic activity at clinically relevant concentrations and conditions. At increasing sonication pressures, liquid PBNDs vaporized into gas microbubbles, and acoustic activity at the second harmonic frequency (2 f0) peaked at lower pressures than the subharmonic frequency (1/2 f0). Definity-based PBNDs have been thoroughly characterized, cryo-TEM has been shown to be suitable to image the internal structure of volatile NDs, and PBNDs can be reliably stored at -80 °C for future use up to 7 days without significant degradation, loss of acoustic response, or reduction in ultrasound-triggered drug release.
Collapse
Affiliation(s)
- Siulam Ginni Ting
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
- Institute of Biomedical Imaging, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
24
|
Li Q, Tang Z, Zhang Y, Yuan T, Yuan B, Du L, Jin Y. Application of low-intensity ultrasound by opening blood-brain barrier for enhanced brain-targeted drug delivery. Int J Pharm 2023; 642:123191. [PMID: 37391108 DOI: 10.1016/j.ijpharm.2023.123191] [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: 02/13/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
Brain-targeted drug delivery has been a research hotspot, and substantial amount of related studies were already translated into standard therapy and put into clinical use. However, low effective rate retains a huge challenge for brain disease. Because, the blood-brain barrier (BBB) protects brain from pathogenic molecules and tightly controls the process of molecular transportation, which gives rise to poor-liposoluble drugs or molecules with high molecular weight cannot permeate the barrier to exert treating effect. There is an ongoing process to dig out more methods for efficient brain-targeted drug delivery. Besides modified chemical methods such as prodrugs design and brain-targeted nanotechnology, physical methods as a novel initiative could enhance the treatment effect for brain disease. In our study, the influence of low-intensity ultrasound on transient opening BBB and the related applications were explored. A medical ultrasound therapeutic device (1 MHz) was used on heads of mice at different intensities and for different treating time. Evans blue was used as a model to exhibit the permeability of the BBB after subcutaneous injection. Three types of intensities (0.6, 0.8, and 1.0 W/cm2) and duration times (1, 3, and 5 min) of ultrasound were respectively investigated. It was found that the combinations of 0.6 W/cm2/1 min, 0.6 W/cm2/3 min, 0.6 W/cm2/5 min, 0.8 W/cm2/1 min, and 1.0 W/cm2/1 min could open the BBB sufficiently with significant Evans blue staining in the brain. Brain pathological analysis showed structural change on moderate degree was found on cerebral cortex after ultrasound and could recovered rapidly. There are no obvious changes in the behavior of mice after ultrasound processing. More importantly, the BBB recovered quickly at 12 h after ultrasound application with complete BBB structure and unbroken tight junction, suggesting that ultrasound was safe to apply for brain-targeted drug delivery. Proper use of local ultrasound on the brain is a promising technique to open the BBB and enhance brain-targeted delivery.
Collapse
Affiliation(s)
- Qian Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ziyan Tang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yuanyuan Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tianyu Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; College of Pharmacy, Henan University, Kaifeng 475004, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lina Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; College of Pharmacy, Henan University, Kaifeng 475004, China.
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; College of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
25
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
26
|
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 2023:107-125. [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] [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
|
27
|
Gouveia FV, Lea‐Banks H, Aubert I, Lipsman N, Hynynen K, Hamani C. Anesthetic-loaded nanodroplets with focused ultrasound reduces agitation in Alzheimer's mice. Ann Clin Transl Neurol 2023; 10:507-519. [PMID: 36715553 PMCID: PMC10109287 DOI: 10.1002/acn3.51737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/03/2023] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) is often associated with neuropsychiatric symptoms, including agitation and aggressive behavior. These symptoms increase with disease severity, ranging from 10% in mild cognitive impairment to 50% in patients with moderate-to-severe AD, pose a great risk for self-injury and injury to caregivers, result in high rates of institutionalization and great suffering for patients and families. Current pharmacological therapies have limited efficacy and a high potential for severe side effects. Thus, there is a growing need to develop novel therapeutics tailored to safely and effectively reduce agitation and aggressive behavior in AD. Here, we investigate for the first time the use of focused ultrasound combined with anesthetic-loaded nanodroplets (nanoFUS) targeting the amygdala (key structure in the neurocircuitry of agitation) as a novel minimally invasive tool to modulate local neural activity and reduce agitation and aggressive behavior in the TgCRND8 AD transgenic mice. METHODS Male and female animals were tested in the resident-intruder (i.e., aggressive behavior) and open-field tests (i.e., motor agitation) for baseline measures, followed by treatment with active- or sham-nanoFUS. Behavioral testing was then repeated after treatment. RESULTS Active-nanoFUS neuromodulation reduced aggressive behavior and agitation in male mice, as compared to sham-treated controls. Treatment with active-nanoFUS increased the time male mice spent in social-non-aggressive behaviors. INTERPRETATION Our results show that neuromodulation with active-nanoFUS may be a potential therapeutic tool for the treatment of neuropsychiatric symptoms, with special focus on agitation and aggressive behaviors. Further studies are necessary to establish cellular, molecular and long-term behavioral changes following treatment with nanoFUS.
Collapse
Affiliation(s)
- Flavia Venetucci Gouveia
- Biological Sciences PlatformSunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Neurosciences and Mental HealthThe Hospital for Sick ChildrenTorontoOntarioM5G 1X8Canada
| | - Harriet Lea‐Banks
- Physical Sciences PlatformSunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Isabelle Aubert
- Biological Sciences PlatformSunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Laboratory Medicine & PathobiologyUniversity of TorontoTorontoOntarioM5S 1A1Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences CentreTorontoOntarioM4N 3M5Canada
| | - Nir Lipsman
- Biological Sciences PlatformSunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences CentreTorontoOntarioM4N 3M5Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences CentreTorontoOntarioM4N 3M5Canada
- Division of NeurosurgeryUniversity of TorontoTorontoOntarioM5T 1P5Canada
| | - Kullervo Hynynen
- Physical Sciences PlatformSunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences CentreTorontoOntarioM4N 3M5Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5S 1A1Canada
- Institute of Biomedical Engineering, University of TorontoTorontoOntarioM5S 1A1Canada
| | - Clement Hamani
- Biological Sciences PlatformSunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences CentreTorontoOntarioM4N 3M5Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences CentreTorontoOntarioM4N 3M5Canada
- Division of NeurosurgeryUniversity of TorontoTorontoOntarioM5T 1P5Canada
| |
Collapse
|
28
|
Hu Y, Wei J, Shen Y, Chen S, Chen X. Barrier-breaking effects of ultrasonic cavitation for drug delivery and biomarker release. ULTRASONICS SONOCHEMISTRY 2023; 94:106346. [PMID: 36870921 PMCID: PMC10040969 DOI: 10.1016/j.ultsonch.2023.106346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 05/27/2023]
Abstract
Recently, emerging evidence has demonstrated that cavitation actually creates important bidirectional channels on biological barriers for both intratumoral drug delivery and extratumoral biomarker release. To promote the barrier-breaking effects of cavitation for both therapy and diagnosis, we first reviewed recent technical advances of ultrasound and its contrast agents (microbubbles, nanodroplets, and gas-stabilizing nanoparticles) and then reported the newly-revealed cavitation physical details. In particular, we summarized five types of cellular responses of cavitation in breaking the plasma membrane (membrane retraction, sonoporation, endocytosis/exocytosis, blebbing and apoptosis) and compared the vascular cavitation effects of three different types of ultrasound contrast agents in breaking the blood-tumor barrier and tumor microenvironment. Moreover, we highlighted the current achievements of the barrier-breaking effects of cavitation in mediating drug delivery and biomarker release. We emphasized that the precise induction of a specific cavitation effect for barrier-breaking was still challenged by the complex combination of multiple acoustic and non-acoustic cavitation parameters. Therefore, we provided the cutting-edge in-situ cavitation imaging and feedback control methods and suggested the development of an international cavitation quantification standard for the clinical guidance of cavitation-mediated barrier-breaking effects.
Collapse
Affiliation(s)
- Yaxin Hu
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Jianpeng Wei
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Yuanyuan Shen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Siping Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Xin Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China.
| |
Collapse
|
29
|
Jo S, Sun IC, Ahn CH, Lee S, Kim K. Recent Trend of Ultrasound-Mediated Nanoparticle Delivery for Brain Imaging and Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:120-137. [PMID: 35184560 DOI: 10.1021/acsami.1c22803] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In view of the fact that the blood-brain barrier (BBB) prevents the transport of imaging probes and therapeutic agents to the brain and thus hinders the diagnosis and treatment of brain-related disorders, methods of circumventing this problem (e.g., ultrasound-mediated nanoparticle delivery) have drawn much attention. Among the related techniques, focused ultrasound (FUS) is a favorite means of enhancing drug delivery via transient BBB opening. Photoacoustic brain imaging relies on the conversion of light into heat and the detection of ultrasound signals from contrast agents, offering the benefits of high resolution and large penetration depth. The extensive versatility and adjustable physicochemical properties of nanoparticles make them promising therapeutic agents and imaging probes, allowing for successful brain imaging and treatment through the combined action of ultrasound and nanoparticulate agents. FUS-induced BBB opening enables nanoparticle-based drug delivery systems to efficiently access the brain. Moreover, photoacoustic brain imaging using nanoparticle-based contrast agents effectively visualizes brain morphologies or diseases. Herein, we review the progress in the simultaneous use of nanoparticles and ultrasound in brain research, revealing the potential of ultrasound-mediated nanoparticle delivery for the effective diagnosis and treatment of brain disorders.
Collapse
Affiliation(s)
- SeongHoon Jo
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Republic of Korea
- Research Institute of Advanced Materials (RIAM), Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul08826, Republic of Korea
| | - In-Cheol Sun
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Cheol-Hee Ahn
- Research Institute of Advanced Materials (RIAM), Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul08826, Republic of Korea
| | - Sangmin Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul02447, Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5, Hwarang-ro, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
30
|
Tuncaboylu DC, Wischke C. Opportunities and Challenges of Switchable Materials for Pharmaceutical Use. Pharmaceutics 2022; 14:2331. [PMID: 36365149 PMCID: PMC9696173 DOI: 10.3390/pharmaceutics14112331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 06/27/2024] Open
Abstract
Switchable polymeric materials, which can respond to triggering signals through changes in their properties, have become a major research focus for parenteral controlled delivery systems. They may enable externally induced drug release or delivery that is adaptive to in vivo stimuli. Despite the promise of new functionalities using switchable materials, several of these concepts may need to face challenges associated with clinical use. Accordingly, this review provides an overview of various types of switchable polymers responsive to different types of stimuli and addresses opportunities and challenges that may arise from their application in biomedicine.
Collapse
|
31
|
Li S, Zhang H, Li W, Zhang Y, Gao X, Liu H, Li N, Hu H. Controllable Formation and Real-Time Characterization of Single Microdroplets Using Optical Tweezers. MICROMACHINES 2022; 13:1693. [PMID: 36296046 PMCID: PMC9607241 DOI: 10.3390/mi13101693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
Existing preparation methods for microdroplets usually require offline measurements to characterize single microdroplets. Here, we report an optical method used to facilitate the controllable formation and real-time characterization of single microdroplets. The optical tweezer technique was used to capture and form a microdroplet at the center of the trap. The controllable growth and real-time characterization of the microdroplet was realized, respectively, by adjusting experimental parameters and by resolving the Raman spectra by fitting Mie scattering to the spike positions of the spectra during the controllable growth of microdroplets. The proposed method can be potentially applied in optical microlenses and virus detection.
Collapse
Affiliation(s)
- Shuai Li
- Quantum Sensing Center, Zhejiang Lab., Hangzhou 310000, China
| | - Hanlin Zhang
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wenqiang Li
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yizhou Zhang
- Quantum Sensing Center, Zhejiang Lab., Hangzhou 310000, China
| | - Xiaowen Gao
- Quantum Sensing Center, Zhejiang Lab., Hangzhou 310000, China
| | - Haiqing Liu
- Isvision (Hangzhou) Technology Co., Ltd., Hangzhou 310052, China
| | - Nan Li
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Huizhu Hu
- Quantum Sensing Center, Zhejiang Lab., Hangzhou 310000, China
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
32
|
Zhang S, Gan L, Cao F, Wang H, Gong P, Ma C, Ren L, Lin Y, Lin X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res Bull 2022; 190:69-83. [PMID: 36162603 DOI: 10.1016/j.brainresbull.2022.09.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Three different barriers are formed between the cerebrovascular and the brain parenchyma: the blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the cerebrospinal fluid-brain barrier (CBB). The BBB is the main regulator of blood and central nervous system (CNS) material exchange. The semipermeable nature of the BBB limits the passage of larger molecules and hydrophilic small molecules, Food and Drug Administration (FDA)-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Although the complexity of the BBB affects CNS drug delivery, understanding the composition and function of the BBB can provide a platform for the development of new methods for CNS drug delivery. This review summarizes the classification of the brain barrier, the composition and role of the basic structures of the BBB, and the transport, barrier, and destruction mechanisms of the BBB; discusses the advantages and disadvantages of different drug delivery methods and prospects for future drug delivery strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| | - Lin Gan
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Fengye Cao
- Yiyang The First Hospital of Traditional Chinese Medicine, Yiyang, Hunan Province, 413000, China
| | - Hao Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Peng Gong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Congcong Ma
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Li Ren
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Xianming Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China.
| |
Collapse
|
33
|
Guo H, Chen Q, Li T, Sun D, Xi L. Photoacoustic-triggered nanomedicine delivery to internal organs using a dual-wavelength laparoscope. JOURNAL OF BIOPHOTONICS 2022; 15:e202200116. [PMID: 35661424 DOI: 10.1002/jbio.202200116] [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: 04/15/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Precise drug delivery for internal organs is always an important clinical issue. In this study, we developed a dual-wavelength optical resolution photoacoustic laparoscope, and achieved precise and efficient delivery of nanomedicine to multiple abdominal organs. The laparoscope integrated 532 and 820 nm light to map the vascular network, and visually deliver the nanoparticles to the targeted area using photoacoustic radiation force, respectively. To achieve endoscopic use, we employed a micro-electro-mechanical-system (MEMS) scanner to realize internal two-dimensional raster scanning of the optical beams. Using phantom experiments, the lateral resolutions were measured as 3.75 μm for 532 nm, and 5.25 μm for 820 nm, respectively. Besides, we demonstrated the feasibility of targeted drug delivery using mouse-ear tumor model, normal organs, and colon tumor model. All the experimental results suggested that this strategy can serve as a promising precise drug delivery method for the effective treatment of internal organ diseases.
Collapse
Affiliation(s)
- Heng Guo
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Qian Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Tingting Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Desheng Sun
- Department of Ultrasonic Imaging, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lei Xi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
- Guang Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
34
|
Fan CH, Ho YJ, Lin CW, Wu N, Chiang PH, Yeh CK. State-of-the-art of ultrasound-triggered drug delivery from ultrasound-responsive drug carriers. Expert Opin Drug Deliv 2022; 19:997-1009. [PMID: 35930441 DOI: 10.1080/17425247.2022.2110585] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The development of new tools to locally and non-invasively transferring therapeutic substances at the desired site in deep living tissue has been a long sought-after goal within the drug delivery field. Among the established methods, ultrasound (US) with US-responsive carriers holds great promise and demonstrates on-demand delivery of a variety of functional substances with spatial precision of several millimeters in deep-seated tissues in animal models and humans. These properties have motivated several explorations of US with US responsive carriers as a modality for neuromodulation and the treatment of various diseases, such as stroke and cancer. AREAS COVERED This article briefly discussed three specific mechanisms that enhance in vivo drug delivery via US with US-responsive carriers: 1) permeabilizing cellular membrane, 2) increasing the permeability of vessels, and 3) promoting cellular endocytotic uptake. Besides, a series of US-responsive drug carriers are discussed, with an emphasis on the relation between structural feature and therapeutic outcome. EXPERT OPINION This article summarized current development for each of US-responsive drug carrier, focusing on the routes of enhancing delivery and applications. The mechanisms of interaction between US-responsive carriers and US energy, such as cavitation, hyperthermia, and reactive oxygen species, as well as how these interactions can improve drug delivery into target cell/tissue. It can be expected that there are serval efforts to further identification of US-responsive particles, design of novel US waveform sequence, and survey of optimal combination between US parameters and US-responsive carriers for better controlling the spatiotemporal drug release profile, stability, and safety in vivo. The authors believe these will provide novel tools for precisely designing treatment strategies and significantly benefit the clinical management of several diseases.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Hua Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
35
|
Lea-Banks H, Wu SK, Lee H, Hynynen K. Ultrasound-triggered oxygen-loaded nanodroplets enhance and monitor cerebral damage from sonodynamic therapy. Nanotheranostics 2022; 6:376-387. [PMID: 35795341 PMCID: PMC9254362 DOI: 10.7150/ntno.71946] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/03/2022] [Indexed: 11/05/2022] Open
Abstract
In sonodynamic therapy, cellular toxicity from sonosensitizer drugs, such as 5-aminolevulinic acid hydrochloride (5-ALA), may be triggered with focused ultrasound through the production of reactive oxygen species (ROS). Here we show that by increasing local oxygen during treatment, using oxygen-loaded perfluorocarbon nanodroplets (250 +/- 8 nm), we can increase the damage induced by 5-ALA, and monitor the severity by recording acoustic emissions in the brain. To achieve this, we sonicated the right striatum of 16 healthy rats after an intravenous dose of 5-ALA (200 mg/kg), followed by saline, nanodroplets, or oxygen-loaded nanodroplets. We assessed haemorrhage, edema and cell apoptosis immediately following, 24 hr, and 48 hr after focused ultrasound treatment. The localized volume of damaged tissue was significantly enhanced by the presence of oxygen-loaded nanodroplets, compared to ultrasound with unloaded nanodroplets (3-fold increase), and ultrasound alone (40-fold increase). Sonicating 1 hr following 5-ALA injection was found to be more potent than 2 hr following 5-ALA injection (2-fold increase), and the severity of tissue damage corresponded to the acoustic emissions from droplet vaporization. Enhancing the local damage from 5-ALA with monitored cavitation activity and additional oxygen could have significant implications in the treatment of atherosclerosis and non-invasive ablative surgeries.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Sheng-Kai Wu
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Hannah Lee
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
36
|
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
|
37
|
Rahimi S, Jones RM, Hynynen K. An Acoustic Measurement Library for Non-Invasive Trans-Rodent Skull Ultrasonic Focusing at High Frequency. IEEE Trans Biomed Eng 2021; 69:2184-2191. [PMID: 34951839 DOI: 10.1109/tbme.2021.3138352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To investigate the feasibility of developing an acoustic measurement library for non-invasive trans-rodent skull ultrasonic focusing at high frequency. METHODS A fiber-optic hydrophone (FOH) was positioned at the geometric focus of a spherically-curved phased array (64 elements, 25 mm diameter, 20 mm radius of curvature). Elements were driven sequentially (3.3 MHz driving frequency) and FOH waveforms were recorded with and without intervening ex-vivo rodent skullcaps. Measurements were carried out on 15 skullcaps (Sprague-Dawley rats, 182-209 g) across 3 fixed transmission regions per specimen. An element-wise measurement library of skull-induced phase differences was constructed using mean values across all specimens for each transmission region. Library-based transcranial phase differences were compared with direct FOH-based measurements across 5 additional skullcaps not included in the library. RESULTS Library-based phase corrections deviated less from FOH-based trans-skull phase difference values than those calculated for the water-path case, and restored partial transcranial focal quality relative to that recovered using invasive hydrophone-based corrections. Retrospective analysis suggests comparable performance can be obtained using smaller library sizes. CONCLUSION An acoustic measurement library can facilitate non-invasive transcranial aberration correction in rodents at high frequency. SIGNIFICANCE Library-based focusing represents a practical approach for delivering high-frequency ultrasound brain treatments in small animals.
Collapse
|