1
|
Chen LE, Nittayacharn P, Exner AA. Progress and potential of nanobubbles for ultrasound-mediated drug delivery. Expert Opin Drug Deliv 2025:1-24. [PMID: 40353846 DOI: 10.1080/17425247.2025.2505044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Despite much progress, nanomedicine-based drug therapies in oncology remain limited by systemic toxicity and insufficient particle accumulation in the tumor. To address these barriers, formulations responsive to external physical stimuli have emerged. One most promising system is the ultrasound stimulation of drug-loaded, gas-core particles (bubbles). Ultrasound induces bubble cavitation for cell and tissue permeabilization, triggers on-demand drug release, and provides opportunities for real-time imaging of delivery. AREAS COVERED Here, we focus on shell-stabilized, gas-core nanoparticles (also termed nanobubbles or ultrafine bubbles) and their role in ultrasound-mediated therapeutic delivery to tumors. This review frames the advantages of nanobubbles within the ongoing deficits in nanomedicine, describes mechanisms of ultrasound-mediated therapy, and details formulation techniques for nanobubble delivery systems. It then highlights the past decade of research in nanobubble-facilitated drug delivery for cancer therapy and anticipates new directions in the field. EXPERT OPINION Nanobubble ultrasound contrast agents offer a spatiotemporally triggerable therapeutic coupled with a safe, accessible imaging modality. Nanobubbles can be loaded with diverse therapeutic cargoes to treat disease and overcome numerous barriers limiting delivery to solid tumors. Close attention to formulation, characterization methods, acoustic testing parameters, and the biological mechanisms of nanobubble delivery will facilitate preclinical research toward clinical adoption.
Collapse
Affiliation(s)
- Laura E Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
2
|
Sabale A, Rizvi MS, Chinthapenta V, Eranki A. Effect of focused ultrasound on shearwave production in a hyperelastic media. Biomech Model Mechanobiol 2025:10.1007/s10237-025-01967-2. [PMID: 40382754 DOI: 10.1007/s10237-025-01967-2] [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: 08/16/2024] [Accepted: 04/13/2025] [Indexed: 05/20/2025]
Abstract
Focused ultrasound (FUS) is an emerging noninvasive modality for treating various medical conditions. It encompasses both therapeutic and diagnostic applications, utilizing ultrasound waves at different intensities. In diagnostic modalities, ultrasound energy is deposited at the focus to generate acoustic radiation force (ARF), resulting in the generation of shear stress and waves, which are utilized in elastography to evaluate the mechanical properties of tissue. However, therapeutic modalities utilizing higher intensities may lead to elevated shear stress levels. The shear stress induced in the focal region during FUS procedures can potentially affect biological processes, such as cell membrane permeability and gene regulation. To better understand the mechanical stress generated during FUS procedures, we developed a finite element model (FEM) to simulate sonication using a single-element FUS transducer. We modeled soft tissue using a neo-Hookean hyperelastic constitutive behavior, offering a more realistic representation of tissue behavior compared to the linear elasticity assumptions commonly employed in ultrasound-based elastography techniques. Operational parameters were varied to simulate different acoustic powers of the transducer by applying mechanical surface pressure at various operating frequencies. The model depicted FUS wave propagation with amplified surface pressure at the focus, generating relevant focal pressures consistent with clinical setups. The focal beam size within the soft tissue material was characterized and exhibited dependency on the operating frequency of the transducer. As the FUS wave converged at the focus, an ARF was exerted, resulting in displacement and induced shear stress around the focal region, which were quantified. The displacement and shear stress that were analyzed were dependent on the applied transducer surface pressure. These findings deepen the understanding of the mechanics of low-intensity FUS and provide valuable insights into its shear-related effects due to displacement and deformation of the media.
Collapse
Affiliation(s)
- Aniket Sabale
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana, 502284, India
| | - Mohd Suhail Rizvi
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana, 502284, India
| | - Viswanath Chinthapenta
- Department of Mechanical and Aerospace Engineering, Indian Institute of Technology, Hyderabad, Telangana, 502284, India
| | - Avinash Eranki
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Telangana, 502284, India.
| |
Collapse
|
3
|
Heo D, Kim H, Katagiri W, Yoon C, Lim HG, Kim C, Choi HS, Kashiwagi S, Kim HH. Enhanced model antigen retention in tissue through topical high-frequency ultrasound treatment. Biomed Eng Lett 2025; 15:273-282. [PMID: 40026896 PMCID: PMC11871201 DOI: 10.1007/s13534-024-00445-5] [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: 06/24/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 03/05/2025] Open
Abstract
Reproducible control of tissue delivery and retention of a therapeutic agent facilitates the maximization of the efficacy of pharmacotherapy. Despite the proposal of various chemical and physical methods for modulating drug delivery and retention, the choice of the physical modality for this purpose has been limited in clinical practice. Thus, this study proposes a novel strategy for modulating the retention of a model antigen in tissues by using non-tissue-damaging high-frequency ultrasoundAfter the injection of a fluorescently labeled model antigen, followed by brief treatment with high-frequency ultrasound (5-20 MHz), the clearance of the antigen was monitored using a real-time near-infrared (NIR) fluorescence imaging system in vivo. Further, ultrasound treatment increased tissue retention at the site of model antigen administration. The results suggested that high-frequency ultrasound could change the response to a macromolecule injected into the tissue, such as a vaccine, thereby modulating the immune response. The proposed ultrasound-based technology is translatable to clinical settings and benefits from well-established ultrasound technologies that have been employed in various medical applications for decades. Moreover, this approach can be broadly applied to enhance the therapeutic effects of current and future immune-mediated therapeutic systems.
Collapse
Affiliation(s)
- Dasom Heo
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| | - Hyunhee Kim
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| | - Wataru Katagiri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129 USA
| | - Changhan Yoon
- Department of Biomedical Engineering, Inje University, Gimhae, 50834 Republic of Korea
| | - Hae Gyun Lim
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513 Republic of Korea
| | - Chulhong Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Department of Electrical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Graduate School of Convergence Science and Technology, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129 USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129 USA
| | - Hyung Ham Kim
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Department of Electrical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Graduate School of Convergence Science and Technology, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| |
Collapse
|
4
|
Desai O, Köster M, Kloos D, Lachmann N, Hauser H, Poortinga A, Wirth D. Ultrasound-triggered drug release in vivo from antibubble-loaded macrophages. J Control Release 2025; 378:365-376. [PMID: 39653149 DOI: 10.1016/j.jconrel.2024.12.007] [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: 06/18/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/22/2024]
Abstract
Nanoparticles have proven to be attractive carriers in therapeutic drug delivery since they can encapsulate, protect and stabilize a plethora of different drugs, thereby improving therapeutic efficacy and reducing side effects. However, specific targeting of drug-loaded nanoparticles to the tissue of interest and a timely and spatially controlled release of drugs on demand still represent a challenge. Recently, gas-filled microparticles, so-called antibubbles, have been developed which can efficiently encapsulate liquid drug droplets. Here, we show that antibubbles are efficiently taken up by macrophages in vitro and are stably maintained for more than 48 h without compromising antibubble integrity and macrophage viability. We show that application of diagnostic ultrasound induces the disintegration of both antibubbles and carrier cells while not affecting non-loaded macrophages. Using 4-hydroxytamoxifen as a model drug, we show ultrasound-mediated drug release upon adoptive transfer of antibubble-loaded macrophages in mice. Together with the ability of macrophages to accumulate in inflamed tissues, antibubble-loaded macrophages represent an attractive tool for targeted delivery of drugs and its ultrasound-mediated spatial and temporal drug release, highlighting the therapeutic perspective of this strategy.
Collapse
Affiliation(s)
- Omkar Desai
- Model System for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mario Köster
- Model System for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Doreen Kloos
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
| | - Nico Lachmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany; RESIST, Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Biomedical Research in End Stage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany; Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany
| | - Hansjörg Hauser
- Model System for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany; iBET-Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Albert Poortinga
- Polymer Technology, Eindhoven University of Technology, Eindhoven 5612 AZ, The Netherlands
| | - Dagmar Wirth
- Model System for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany; Institute for Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
5
|
Mandal TK. Nanomaterial-Enhanced Hybrid Disinfection: A Solution to Combat Multidrug-Resistant Bacteria and Antibiotic Resistance Genes in Wastewater. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1847. [PMID: 39591087 PMCID: PMC11597552 DOI: 10.3390/nano14221847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
This review explores the potential of nanomaterial-enhanced hybrid disinfection methods as effective strategies for addressing the growing challenge of multidrug-resistant (MDR) bacteria and antibiotic resistance genes (ARGs) in wastewater treatment. By integrating hybrid nanocomposites and nanomaterials, natural biocides such as terpenes, and ultrasonication, this approach significantly enhances disinfection efficiency compared to conventional methods. The review highlights the mechanisms through which hybrid nanocomposites and nanomaterials generate reactive oxygen species (ROS) under blue LED irradiation, effectively disrupting MDR bacteria while improving the efficacy of natural biocides through synergistic interactions. Additionally, the review examines critical operational parameters-such as light intensity, catalyst dosage, and ultrasonication power-that optimize treatment outcomes and ensure the reusability of hybrid nanocomposites and other nanomaterials without significant loss of photocatalytic activity. Furthermore, this hybrid method shows promise in degrading ARGs, thereby addressing both microbial and genetic pollution. Overall, this review underscores the need for innovative wastewater treatment solutions that are efficient, sustainable, and scalable, contributing to the global fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Tapas Kumar Mandal
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
6
|
Sreedasyam R, Wilson BG, Ferrandez PR, Botvinick EL, Venugopalan V. An optical system for cellular mechanostimulation in 3D hydrogels. Acta Biomater 2024; 189:439-448. [PMID: 39368720 DOI: 10.1016/j.actbio.2024.09.050] [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: 01/04/2023] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
We introduce a method utilizing single laser-generated cavitation bubbles to stimulate cellular mechanotransduction in dermal fibroblasts embedded within 3D hydrogels. We demonstrate that fibroblasts embedded in either amorphous or fibrillar hydrogels engage in Ca2+ signaling following exposure to an impulsive mechanical stimulus provided by a single 250 µm diameter laser-generated cavitation bubble. We find that the spatial extent of the cellular signaling is larger for cells embedded within a fibrous collagen hydrogel as compared to those embedded within an amorphous polyvinyl alcohol polymer (SLO-PVA) hydrogel. Additionally, for fibroblasts embedded in collagen, we find an increased range of cellular mechanosensitivity for cells that are polarized relative to the radial axis as compared to the circumferential axis. By contrast, fibroblasts embedded within SLO-PVA did not display orientation-dependent mechanosensitivity. Fibroblasts embedded in hydrogels and cultured in calcium-free media did not show cavitation-induced mechanotransduction; implicating calcium signaling based on transmembrane Ca2+ transport. This study demonstrates the utility of single laser-generated cavitation bubbles to provide local non-invasive impulsive mechanical stimuli within 3D hydrogel tissue models with concurrent imaging using optical microscopy. STATEMENT OF SIGNIFICANCE: Currently, there are limited methods for the non-invasive real-time assessment of cellular sensitivity to mechanical stimuli within 3D tissue scaffolds. We describe an original approach that utilizes a pulsed laser microbeam within a standard laser scanning microscope system to generate single cavitation bubbles to provide impulsive mechanostimulation to cells within 3D fibrillar and amorphous hydrogels. Using this technique, we measure the cellular mechanosensitivity of primary human dermal fibroblasts embedded in amorphous and fibrillar hydrogels, thereby providing a useful method to examine cellular mechanotransduction in 3D biomaterials. Moreover, the implementation of our method within a standard optical microscope makes it suitable for broad adoption by cellular mechanotransduction researchers and opens the possibility of high-throughput evaluation of biomaterials with respect to cellular mechanosignaling.
Collapse
Affiliation(s)
- Rahul Sreedasyam
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Bryce G Wilson
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States
| | - Patricia R Ferrandez
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| | - Vasan Venugopalan
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| |
Collapse
|
7
|
Hoshi R, Gorospe KA, Labouta HI, Azad T, Lee WL, Thu KL. Alternative Strategies for Delivering Immunotherapeutics Targeting the PD-1/PD-L1 Immune Checkpoint in Cancer. Pharmaceutics 2024; 16:1181. [PMID: 39339217 PMCID: PMC11434872 DOI: 10.3390/pharmaceutics16091181] [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: 08/05/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
The programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint constitutes an inhibitory pathway best known for its regulation of cluster of differentiation 8 (CD8)+ T cell-mediated immune responses. Engagement of PD-L1 with PD-1 expressed on CD8+ T cells activates downstream signaling pathways that culminate in T cell exhaustion and/or apoptosis. Physiologically, these immunosuppressive effects exist to prevent autoimmunity, but cancer cells exploit this pathway by overexpressing PD-L1 to facilitate immune escape. Intravenously (IV) administered immune checkpoint inhibitors (ICIs) that block the interaction between PD-1/PD-L1 have achieved great success in reversing T cell exhaustion and promoting tumor regression in various malignancies. However, these ICIs can cause immune-related adverse events (irAEs) due to off-tumor toxicities which limits their therapeutic potential. Therefore, considerable effort has been channeled into exploring alternative delivery strategies that enhance tumor-directed delivery of PD-1/PD-L1 ICIs and reduce irAEs. Here, we briefly describe PD-1/PD-L1-targeted cancer immunotherapy and associated irAEs. We then provide a detailed review of alternative delivery approaches, including locoregional (LDD)-, oncolytic virus (OV)-, nanoparticle (NP)-, and ultrasound and microbubble (USMB)-mediated delivery that are currently under investigation for enhancing tumor-specific delivery to minimize toxic off-tumor effects. We conclude with a commentary on key challenges associated with these delivery methods and potential strategies to mitigate them.
Collapse
Affiliation(s)
- Ryunosuke Hoshi
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Kristyna A. Gorospe
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Hagar I. Labouta
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Leslie Dan Faculty of Pharmacy, University of Toronto, St. George Campus, Toronto, ON M5S 3M2, Canada
- Biomedical Engineering, Faculty of Applied Science and Engineering, University of Toronto, St. George Campus, Toronto, ON M5S 3E2, Canada
| | - Taha Azad
- Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Health Campus, Sherbrooke, QC J1K 2R1, Canada;
- Research Center, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC J1J 3H5, Canada
| | - Warren L. Lee
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Biochemistry, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada
- Medicine and the Interdepartmental Division of Critical Care Medicine, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5B 1T8, Canada
| | - Kelsie L. Thu
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| |
Collapse
|
8
|
Kim HK, Choi Y, Kim KH, Byun Y, Kim TH, Kim JH, An SH, Bae D, Choi MK, Lee M, Kang G, Chung J, Kim S, Kwon K. Scalable production of siRNA-encapsulated extracellular vesicles for the inhibition of KRAS-mutant cancer using acoustic shock waves. J Extracell Vesicles 2024; 13:e12508. [PMID: 39323378 PMCID: PMC11424982 DOI: 10.1002/jev2.12508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 07/16/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as a potential delivery vehicle for nucleic-acid-based therapeutics, but challenges related to their large-scale production and cargo-loading efficiency have limited their therapeutic potential. To address these issues, we developed a novel "shock wave extracellular vesicles engineering technology" (SWEET) as a non-genetic, scalable manufacturing strategy that uses shock waves (SWs) to encapsulate siRNAs in EVs. Here, we describe the use of the SWEET platform to load large quantities of KRASG12C-targeting siRNA into small bovine-milk-derived EVs (sBMEVs), with high efficiency. The siRNA-loaded sBMEVs effectively silenced oncogenic KRASG12C expression in cancer cells; they inhibited tumour growth when administered intravenously in a non-small cell lung cancer xenograft mouse model. Our study demonstrates the potential for the SWEET platform to serve as a novel method that allows large-scale production of cargo-loaded EVs for use in a wide range of therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - DaeHo Bae
- Exollence Co., Ltd.SeoulRepublic of Korea
| | | | | | - Gwansuk Kang
- Division of Gastroenterology and Hepatology, School of MedicineStanford UniversityStanfordCaliforniaUSA
| | | | | | - Kihwan Kwon
- Exollence Co., Ltd.SeoulRepublic of Korea
- Department of Internal Medicine, College of MedicineEwha Womans UniversitySeoulRepublic of Korea
| |
Collapse
|
9
|
Tu B, Li Y, Wen W, Liu J. Bibliometric and visualized analysis of ultrasound combined with microbubble therapy technology from 2009 to 2023. Front Pharmacol 2024; 15:1418142. [PMID: 39119614 PMCID: PMC11306066 DOI: 10.3389/fphar.2024.1418142] [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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
Background In recent years, with the rapid advancement of fundamental ultrasonography research, the application of ultrasound in disease treatment has progressively increased. An increasing body of research indicates that microbubbles serve not only as contrast agents but also in conjunction with ultrasound, enhancing cavitation effects and facilitating targeted drug delivery, thereby augmenting therapeutic efficacy. The objective of this study is to explore the current status and prevailing research trends in this field from 2009 to 2023 through bibliometric analysis and to forecast future developmental trajectories. Methods We selected the Science Citation Index Expanded (SCI-Expanded) from the Web of Science Core Collection (WOSCC) as our primary data source. On 19 January 2024, we conducted a comprehensive search encompassing all articles and reviews published between 2009 and 2023 and utilized the bibliometric online analysis platform, CiteSpace and VOSviewer software to analyze countries/regions, institutions, authors, keywords, and references, used Microsoft Excel 2021 to visualize the trends of the number of articles published by year. Results Between 1 January 2009, and 31 December 2023, 3,326 publications on ultrasound combined with microbubble therapy technology were included. There were a total of 2,846 articles (85.6%) and 480 reviews (14.4%) from 13,062 scholars in 68 countries/regions published in 782 journals. China and the United States emerged as the primary contributors in this domain. In terms of publication output and global institutional collaboration, the University of Toronto in Canada has made the most significant contribution to this field. Professor Kullervo Hynynen has achieved remarkable accomplishments in this area. Ultrasound in Medicine and Biology is at the core of the publishing of research on ultrasound combined with microbubble therapy technology. Keywords such as "sonodynamic therapy," "oxygen," "loaded microbubbles" and "Alzheimer's disease" indicate emerging trends in the field and hold the potential to evolve into significant areas of future investigation. Conclusion This study provides a summary of the key contributions of ultrasound combined with microbubble therapy to the field's development over the past 15 years and delves into the historical underpinnings and contemporary trends of ultrasound combined with microbubble therapy technology, providing valuable guidance for researchers.
Collapse
Affiliation(s)
- Bin Tu
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yan Li
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wen Wen
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jian Liu
- Department of Ultrasound, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
10
|
Nakayama M, Noda A, Akasaka H, Tominaga T, McCorkell G, Geso M, Sasaki R. Ultrasound-stimulated Microbubbles for Treatment of Pancreatic Cancer Cells with Radiation and Nanoparticles: In vitro Study. J Med Phys 2024; 49:326-334. [PMID: 39526155 PMCID: PMC11548062 DOI: 10.4103/jmp.jmp_30_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/10/2024] [Accepted: 08/10/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose This study aims to investigate the radiation enhancement effects of ultrasound-stimulated microbubbles (USMB) with X-rays and nanoparticles on pancreatic cancer cells in vitro. Methods Sonazoid™ microbubbles were used for USMB treatment with a commercially available ultrasound unit. The characterization of the microbubbles before and after ultrasound exposure with different mechanical parameters was evaluated microscopically. Two pancreatic cancer cell lines, MIAPaCa-2 and PANC-1, were treated with different concentrations of microbubbles in combination with 150 kVp X-rays and hydrogen peroxide-modified titanium dioxide nanoparticles. Cell viability was evaluated using a water-soluble tetrazolium dye and a colony formation assay. In addition, intracellular reactive oxygen species (ROS) induced by the combined treatment were assessed. Results The number of burst microbubbles increased with ultrasound's higher mechanical index and the exposure time. A significant radiation enhancement effect with a significant increase in ROS levels was observed in MIAPaCa-2 cells treated with USMB and 6 Gy X-rays, whereas it was not significant in PANC-1 cells treated with the same. When a higher concentration of USMB was applied with X-rays, no radiation enhancement effects were observed in either cell line. Moreover, there was no radiation enhancement effect by USMB between cells treated with and without nanoparticles. Conclusions The results indicate that USMB treatment can additively enhance the therapeutic efficacy of radiation therapy on pancreatic cancer cells, while the synergistic enhancement effects are likely to be cell type and microbubble concentration dependent. In addition, USMB did not improve the efficacy of nanoparticle-induced radiosensitization in the current setting.
Collapse
Affiliation(s)
- Masao Nakayama
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo
- Discipline of Medical Radiations, School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
- Division of Radiation Therapy, Kita-Harima Medical Center, Ono, Hyogo
| | - Ayaha Noda
- Department of Clinical Radiology, Faculty of Health Sciences, Hiroshima International University, Higashihiroshima, Hiroshima, Japan
| | - Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Takahiro Tominaga
- Department of Clinical Radiology, Faculty of Health Sciences, Hiroshima International University, Higashihiroshima, Hiroshima, Japan
| | - Giulia McCorkell
- Discipline of Medical Radiations, School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Moshi Geso
- Discipline of Medical Radiations, School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo
| |
Collapse
|
11
|
Zhou M, Liu L, Zhou W, Yang H, Li M, Yin P, Zhou Y. How Microbubble-Enhanced Shock Waves Promote the Delivery of Lipid-siRNA across Neuronal Plasma Membrane: A Computational Study. J Phys Chem B 2024; 128:2897-2904. [PMID: 38484173 DOI: 10.1021/acs.jpcb.3c07556] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
In this computational study, we examine the potential of microbubble-enhanced shock waves to improve the delivery of lipid-siRNA nanoparticles across neuronal plasma membranes with the ultimate aim of enhancing brain tumor treatment. We critically evaluate several variables related to experiments, including the bubble size, the shock speed and action time, and the amount of siRNA encapsulated in the liposome. Our findings reveal that microbubble-enhanced shock waves are essential for the high delivery of small lipid vesicles (under 30 nm diameter); its corresponding variables significantly impact drug penetration and absorption rates and influence the overall efficacy of the drug delivery system. Long-time recovery simulations further provide valuable insights into the self-healing ability of the plasma membrane following shock wave exposure and the subsequent absorption dynamics of siRNA. This work provides the dynamic process of siRNA released from lipid vesicles with shock wave and nanobubbles, thereby serving as a molecular mechanism support for developing tunable delivery systems for RNA-based therapy in brain tumors.
Collapse
Affiliation(s)
- Mi Zhou
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
- School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Liu Liu
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Wenyu Zhou
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Hong Yang
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Ming Li
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Ping Yin
- School of Materials Science & Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Yang Zhou
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| |
Collapse
|
12
|
Shakya G, Cattaneo M, Guerriero G, Prasanna A, Fiorini S, Supponen O. Ultrasound-responsive microbubbles and nanodroplets: A pathway to targeted drug delivery. Adv Drug Deliv Rev 2024; 206:115178. [PMID: 38199257 DOI: 10.1016/j.addr.2023.115178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Ultrasound-responsive agents have shown great potential as targeted drug delivery agents, effectively augmenting cell permeability and facilitating drug absorption. This review focuses on two specific agents, microbubbles and nanodroplets, and provides a sequential overview of their drug delivery process. Particular emphasis is given to the mechanical response of the agents under ultrasound, and the subsequent physical and biological effects on the cells. Finally, the state-of-the-art in their pre-clinical and clinical implementation are discussed. Throughout the review, major challenges that need to be overcome in order to accelerate their clinical translation are highlighted.
Collapse
Affiliation(s)
- Gazendra Shakya
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Marco Cattaneo
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Giulia Guerriero
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Anunay Prasanna
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Samuele Fiorini
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Outi Supponen
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|
13
|
Huang G, Lin L, Liu Q, Wu S, Chen J, Zhu R, You H, Sun C. Three-dimensional array of microbubbles sonoporation of cells in microfluidics. Front Bioeng Biotechnol 2024; 12:1353333. [PMID: 38419723 PMCID: PMC10899490 DOI: 10.3389/fbioe.2024.1353333] [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: 12/10/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Sonoporation is a popular membrane disruption technique widely applicable in various fields, including cell therapy, drug delivery, and biomanufacturing. In recent years, there has been significant progress in achieving controlled, high-viability, and high-efficiency cell sonoporation in microfluidics. If the microchannels are too small, especially when scaled down to the cellular level, it still remains a challenge to overcome microchannel clogging, and low throughput. Here, we presented a microfluidic device capable of modulating membrane permeability through oscillating three-dimensional array of microbubbles. Simulations were performed to analyze the effective range of action of the oscillating microbubbles to obtain the optimal microchannel size. Utilizing a high-precision light curing 3D printer to fabricate uniformly sized microstructures in a one-step on both the side walls and the top surface for the generation of microbubbles. These microbubbles oscillated with nearly identical amplitudes and frequencies, ensuring efficient and stable sonoporation within the system. Cells were captured and trapped on the bubble surface by the acoustic streaming and secondary acoustic radiation forces induced by the oscillating microbubbles. At a driving voltage of 30 Vpp, the sonoporation efficiency of cells reached 93.9% ± 2.4%.
Collapse
Affiliation(s)
- Guangyong Huang
- School of Mechanical Engineering, Guangxi University, Nanning, China
- School of Mechanical and Automotive Engineering, Guangxi University of Science and Technology, Liuzhou, China
| | - Lin Lin
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Quanhui Liu
- Animal Science and Technology College, Guangxi University, Nanning, China
| | - Shixiong Wu
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Jiapeng Chen
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Rongxing Zhu
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Hui You
- School of Mechanical Engineering, Guangxi University, Nanning, China
| | - Cuimin Sun
- School of Computer, Electronics and Information, Guangxi University, Nanning, China
| |
Collapse
|
14
|
Tran NLH, Lam TQ, Duong PVQ, Doan LH, Vu MP, Nguyen KHP, Nguyen KT. Review on the Significant Interactions between Ultrafine Gas Bubbles and Biological Systems. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:984-996. [PMID: 38153335 DOI: 10.1021/acs.langmuir.3c03223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Having sizes comparable with living cells and high abundance, ultrafine bubbles (UBs) are prone to inevitable interactions with different types of cells and facilitate alterations in physiological properties. The interactions of four typical cell types (e.g., bacterial, fungal, plant, and mammalian cells) with UBs have been studied over recent years. For bacterial cells, UBs have been utilized in creating the capillary force to tear down biofilms. The release of high amounts of heat, pressure, and free radicals during bubble rupture is also found to affect bacterial cell growth. Similarly, the bubble gas core identity plays an important role in the development of fungal cells. By the proposed mechanism of attachment of UBs on hydrophobin proteins in the fungal cell wall, oxygen and ozone gas-filled ultrafine bubbles can either promote or hinder the cell growth rate. On the other hand, reactive oxygen species (ROS) formation and mass transfer facilitation are two means of indirect interactions between UBs and plant cells. Likewise, the use of different gas cores in generating bubbles can produce different physical effects on these cells, for example, hydrogen gas for antioxidation against infections and oxygen for oxidation of toxic metal ions. For mammalian cells, the importance of investigating their interactions with UBs lies in the bubbles' action on cell viability as membrane poration for drug delivery can greatly affect cells' survival. UBs have been utilized and tested in forming the pores by different methods, ranging from bubble oscillation and microstream generation through acoustic cavitation to bubble implosion.
Collapse
Affiliation(s)
- Nguyen Le Hanh Tran
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Thien Quang Lam
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Phuong Vu Quynh Duong
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Linh Hai Doan
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Mai Phuong Vu
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Khang Huy Phuc Nguyen
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| | - Khoi Tan Nguyen
- School of Biotechnology, International University, Vietnam National University, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
15
|
Cui LW, Fan LY, Shen ZY. Application Research Progress of Nanomaterial Graphene and its Derivative Complexes in Tumor Diagnosis and Therapy. Curr Med Chem 2024; 31:6436-6459. [PMID: 38299292 DOI: 10.2174/0109298673251648231106112354] [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: 03/16/2023] [Revised: 08/05/2023] [Accepted: 10/05/2023] [Indexed: 02/02/2024]
Abstract
Functional nanomaterial graphene and its derivatives have attracted considerable attention in many fields because of their unique physical and chemical properties. Most notably, graphene has become a research hotspot in the biomedical field, especially in relation to malignant tumors. In this study, we briefly review relevant research from recent years on graphene and its derivatives in tumor diagnosis and antitumor therapy. The main contents of the study include the graphene-derivative diagnosis of tumors in the early stage, graphene quantum dots, photodynamics, MRI contrast agent, acoustic dynamics, and the effects of ultrasonic cavitation and graphene on tumor therapy. Moreover, the biocompatibility of graphene is briefly described. This review provides a broad overview of the applications of graphene and its derivatives in tumors. Conclusion, graphene and its derivatives play an important role in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Li Wen Cui
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, No. 30, North Tong-yang Road, Pingchao Town, Tongzhou District, Nantong, Jiangsu 226361, China
| | - Lu Yao Fan
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, No. 30, North Tong-yang Road, Pingchao Town, Tongzhou District, Nantong, Jiangsu 226361, China
| | - Zhi Yong Shen
- Department of Radiology, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, No. 30, North Tong-yang Road, Pingchao Town, Tongzhou District, Nantong, Jiangsu 226361, China
| |
Collapse
|
16
|
Shi Y, Weng W, Chen M, Huang H, Chen X, Peng Y, Hu Y. Improving DNA vaccination performance through a new microbubble design and an optimized sonoporation protocol. ULTRASONICS SONOCHEMISTRY 2023; 101:106685. [PMID: 37976565 PMCID: PMC10692915 DOI: 10.1016/j.ultsonch.2023.106685] [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: 08/20/2023] [Revised: 10/23/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
As a non-viral transfection method, ultrasound and microbubble-induced sonoporation can achieve spatially targeted gene delivery with synergistic immunostimulatory effects. Here, we report for the first time the application of sonoporation for improving DNA vaccination performance. This study developed a new microbubble design with nanoscale DNA/PEI complexes loaded onto cationic microbubbles to attain significant increases in DNA-loading capacity (0.25 pg per microbubble) and in vitro transfection efficiency. Using live-cell imaging, we revealed the membrane perforation and cellular delivery characteristics of sonoporation. Using luciferase reporter gene for in vivo transfection, we showed that sonoporation increased the transfection efficiency by 40.9-fold when compared with intramuscular injection. Moreover, we comprehensively optimized the sonoporation protocol and further increased the transfection efficiency by 43.6-fold. Immunofluorescent staining results showed that sonoporation effectively activated the MHC-II+ immune cells. Using a hepatitis B DNA vaccine, sonoporation induced significantly higher serum antibody levels when compared with intramuscular injection, and the antibodies sustained for 56 weeks. In addition, we recorded the longest reported expression period (400 days) of the sonoporation-delivered gene. Whole genome resequencing confirmed that the gene with stable expression existed in an extrachromosomal state without integration. Our results demonstrated the potential of sonoporation for efficient and safe DNA vaccination.
Collapse
Affiliation(s)
- Yuanchao Shi
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Weixiong Weng
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Mengting Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Haoqiang Huang
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Xin Chen
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Yin Peng
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Yaxin Hu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
17
|
Wu H, Zhou C, Li Y, Jin Y, Lai X, Ohl CD, Li D, Yu H. Mechanisms underlying the influence of skin properties on a single cavitation bubble in low-frequency sonophoresis. ULTRASONICS SONOCHEMISTRY 2023; 101:106690. [PMID: 37948892 PMCID: PMC10663890 DOI: 10.1016/j.ultsonch.2023.106690] [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: 09/12/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
As a safe and effective method for systemic transdermal drug delivery (TDD), sonophoresis has drawn much attention from researchers. Despite numerous studies confirming cavitation as the main reason for sonophoresis, the effect skin has on cavitation bubble dynamics remains elusive due to the difficulty of experimental challenges. For a start, we reveal how single cavitation bubble (SCB) dynamics are affected by skin properties, including elasticity, hydrophilicity and texture. We use polydimethylsiloxane (PDMS) to simulate human skin and record the temporary evolution of SCBs with synchronous ultrafast photography. The influences of skin properties on SCBs are concluded: 1) SCBs collapse later near walls with better elasticities and generate microjets with higher speed; 2) SCBs collapse later near hydrophilic walls with slower microjets; and 3) the existence of a texture structure on walls also delays the time of bubble collapse near them and slows the velocities of microjets (v) during collapses.
Collapse
Affiliation(s)
- Hao Wu
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, PR China; Department of Soft Matter, Institute of Physics, Otto-von-Guericke University, Magdeburg 39106, Germany; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Cheng Zhou
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, PR China
| | - Yuanyuan Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yongzhen Jin
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Xiaochen Lai
- School of Automation, Nanjing University of Information Science & Technology, Nanjing 210044, PR China
| | - Claus-Dieter Ohl
- Department of Soft Matter, Institute of Physics, Otto-von-Guericke University, Magdeburg 39106, Germany
| | - Dachao Li
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, PR China
| | - Haixia Yu
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, PR China.
| |
Collapse
|
18
|
Aliabouzar M, Abeid BA, Kripfgans OD, Fowlkes JB, Estrada JB, Fabiilli ML. Real-time spatiotemporal characterization of mechanics and sonoporation of acoustic droplet vaporization in acoustically responsive scaffolds. APPLIED PHYSICS LETTERS 2023; 123:114101. [PMID: 37705893 PMCID: PMC10497320 DOI: 10.1063/5.0159661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/26/2023] [Indexed: 09/15/2023]
Abstract
Phase-shift droplets provide a flexible and dynamic platform for therapeutic and diagnostic applications of ultrasound. The spatiotemporal response of phase-shift droplets to focused ultrasound, via the mechanism termed acoustic droplet vaporization (ADV), can generate a range of bioeffects. Although ADV has been used widely in theranostic applications, ADV-induced bioeffects are understudied. Here, we integrated ultra-high-speed microscopy, confocal microscopy, and focused ultrasound for real-time visualization of ADV-induced mechanics and sonoporation in fibrin-based, tissue-mimicking hydrogels. Three monodispersed phase-shift droplets-containing perfluoropentane (PFP), perfluorohexane (PFH), or perfluorooctane (PFO)-with an average radius of ∼6 μm were studied. Fibroblasts and tracer particles, co-encapsulated within the hydrogel, were used to quantify sonoporation and mechanics resulting from ADV, respectively. The maximum radial expansion, expansion velocity, induced strain, and displacement of tracer particles were significantly higher in fibrin gels containing PFP droplets compared to PFH or PFO. Additionally, cell membrane permeabilization significantly depended on the distance between the droplet and cell (d), decreasing rapidly with increasing d. Significant membrane permeabilization occurred when d was smaller than the maximum radius of expansion. Both ultra-high-speed and confocal images indicate a hyper-local region of influence by an ADV bubble, which correlated inversely with the bulk boiling point of the phase-shift droplets. The findings provide insight into developing optimal approaches for therapeutic applications of ADV.
Collapse
Affiliation(s)
| | - Bachir A. Abeid
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | - Jonathan B. Estrada
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
19
|
Wen Z, Liu C, Teng Z, Jin Q, Liao Z, Zhu X, Huo S. Ultrasound meets the cell membrane: for enhanced endocytosis and drug delivery. NANOSCALE 2023; 15:13532-13545. [PMID: 37548587 DOI: 10.1039/d3nr02562d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Endocytosis plays a crucial role in drug delivery for precision therapy. As a non-invasive and spatiotemporal-controllable stimulus, ultrasound (US) has been utilized for improving drug delivery efficiency due to its ability to enhance cell membrane permeability. When US meets the cell membrane, the well-known cavitation effect generated by US can cause various biophysical effects, facilitating the delivery of various cargoes, especially nanocarriers. The comprehension of recent progress in the biophysical mechanism governing the interaction between ultrasound and cell membranes holds significant implications for the broader scientific community, particularly in drug delivery and nanomedicine. This review will summarize the latest research results on the biological effects and mechanisms of US-enhanced cellular endocytosis. Moreover, the latest achievements in US-related biomedical applications will be discussed. Finally, challenges and opportunities of US-enhanced endocytosis for biomedical applications will be provided.
Collapse
Affiliation(s)
- Zihao Wen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zihao Teng
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Quanyi Jin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
20
|
Pandur Ž, Zevnik J, Podbevšek D, Stojković B, Stopar D, Dular M. Water treatment by cavitation: Understanding it at a single bubble - bacterial cell level. WATER RESEARCH 2023; 236:119956. [PMID: 37087917 DOI: 10.1016/j.watres.2023.119956] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
Cavitation is a potentially useful phenomenon accompanied by extreme conditions, which is one of the reasons for its increased use in a variety of applications, such as surface cleaning, enhanced chemistry, and water treatment. Yet, we are still not able to answer many fundamental questions related to efficacy and effectiveness of cavitation treatment, such as: "Can single bubbles destroy contaminants?" and "What precisely is the mechanism behind bubble's cleaning power?". For these reasons, the present paper addresses cavitation as a tool for eradication and removal of wall-bound bacteria at a fundamental level of a single microbubble and a bacterial cell. We present a method to study bubble-bacteria interaction on a nano- to microscale resolution in both space and time. The method allows for accurate and fast positioning of a single microbubble above the individual wall-bound bacterial cell with optical tweezers and triggering of a violent microscale cavitation event, which either results in mechanical removal or destruction of the bacterial cell. Results on E. coli bacteria show that only cells in the immediate vicinity of the microbubble are affected, and that a very high likelihood of cell detachment and cell death exists for cells located directly under the center of a bubble. Further details behind near-wall microbubble dynamics are revealed by numerical simulations, which demonstrate that a water jet resulting from a near-wall bubble implosion is the primary mechanism of wall-bound cell damage. The results suggest that peak hydrodynamic forces as high as 0.8 μN and 1.2 μN are required to achieve consistent E. coli bacterial cell detachment or death with high frequency mechanical perturbations on a nano- to microsecond time scale. Understanding of the cavitation phenomenon at a fundamental level of a single bubble will enable further optimization of novel water treatment and surface cleaning technologies to provide more efficient and chemical-free processes.
Collapse
Affiliation(s)
- Žiga Pandur
- Faculty of Mechanical Engineering, University of Ljubljana, Askerceva 6, 1000 Ljubljana, Slovenia; Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Jure Zevnik
- Faculty of Mechanical Engineering, University of Ljubljana, Askerceva 6, 1000 Ljubljana, Slovenia
| | - Darjan Podbevšek
- Faculty of Mechanical Engineering, University of Ljubljana, Askerceva 6, 1000 Ljubljana, Slovenia; Advanced Science Research Center at The Graduate Center of the City University of New York, 85 Saint Nicholas Terrace, New York, USA
| | - Biljana Stojković
- Faculty of Mechanical Engineering, University of Ljubljana, Askerceva 6, 1000 Ljubljana, Slovenia
| | - David Stopar
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Matevž Dular
- Faculty of Mechanical Engineering, University of Ljubljana, Askerceva 6, 1000 Ljubljana, Slovenia.
| |
Collapse
|
21
|
Hu S, Zhang X, Melzer A, Landgraf L. Ultrasound-induced cavitation renders prostate cancer cells susceptible to hyperthermia: Analysis of potential cellular and molecular mechanisms. Front Genet 2023; 14:1122758. [PMID: 37152995 PMCID: PMC10154534 DOI: 10.3389/fgene.2023.1122758] [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: 12/13/2022] [Accepted: 03/21/2023] [Indexed: 05/09/2023] Open
Abstract
Background: Focused ultrasound (FUS) has become an important non-invasive therapy for prostate tumor ablation via thermal effects in the clinic. The cavitation effect induced by FUS is applied for histotripsy, support drug delivery, and the induction of blood vessel destruction for cancer therapy. Numerous studies report that cavitation-induced sonoporation could provoke multiple anti-proliferative effects on cancer cells. Therefore, cavitation alone or in combination with thermal treatment is of great interest but research in this field is inadequate. Methods: Human prostate cancer cells (LNCap and PC-3) were exposed to 40 s cavitation using a FUS system, followed by water bath hyperthermia (HT). The clonogenic assay, WST-1 assay, and Transwell® invasion assay, respectively, were used to assess cancer cell clonogenic survival, metabolic activity, and invasion potential. Fluorescence microscopy using propidium iodide (PI) as a probe of cell membrane integrity was used to identify sonoporation. The H2A.X assay and Nicoletti test were conducted in the mechanism investigation to detect DNA double-strand breaks (DSBs) and cell cycle arrest. Immunofluorescence microscopy and flow cytometry were performed to determine the distribution and expression of 5α-reductase (SRD5A). Results: Short FUS shots with cavitation (FUS-Cav) in combination with HT resulted in, respectively, a 2.2, 2.3, and 2.8-fold decrease (LNCap) and a 2.0, 1.5, and 1.6-fold decrease (PC-3) in the clonogenic survival, cell invasiveness and metabolic activity of prostate cancer cells when compared to HT alone. FUS-Cav immediately induced sonoporation in 61.7% of LNCap cells, and the combination treatment led to a 1.4 (LNCap) and 1.6-fold (PC-3) increase in the number of DSBs compared to HT alone. Meanwhile, the combination therapy resulted in 26.68% of LNCap and 31.70% of PC-3 with cell cycle arrest in the Sub-G1 phase and 35.37% of PC-3 with cell cycle arrest in the G2/M phase. Additionally, the treatment of FUS-Cav combined with HT block the androgen receptor (AR) signal pathway by reducing the relative Type I 5α-reductase (SRD5A1) level to 38.28 ± 3.76% in LNCap cells, and decreasing the relative Type III 5α-reductase 3 (SRD5A3) level to 22.87 ± 4.88% in PC-3 cells, in contrast, the relative SRD5A level in untreated groups was set to 100%. Conclusion: FUS-induced cavitation increases the effects of HT by interrupting cancer cell membranes, inducing the DSBs and cell cycle arrest, and blocking the AR signal pathway of the prostate cancer cells, with the potential to be a promising adjuvant therapy in prostate cancer treatment.
Collapse
Affiliation(s)
- Shaonan Hu
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| | - Xinrui Zhang
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| | - Andreas Melzer
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
- Institute for Medical Science and Technology (IMSaT), University of Dundee, Dundee, United Kingdom
| | - Lisa Landgraf
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| |
Collapse
|
22
|
Zhu Y, Li Q, Wang C, Hao Y, Yang N, Chen M, Ji J, Feng L, Liu Z. Rational Design of Biomaterials to Potentiate Cancer Thermal Therapy. Chem Rev 2023. [PMID: 36912061 DOI: 10.1021/acs.chemrev.2c00822] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Cancer thermal therapy, also known as hyperthermia therapy, has long been exploited to eradicate mass lesions that are now defined as cancer. With the development of corresponding technologies and equipment, local hyperthermia therapies such as radiofrequency ablation, microwave ablation, and high-intensity focused ultrasound, have has been validated to effectively ablate tumors in modern clinical practice. However, they still face many shortcomings, including nonspecific damages to adjacent normal tissues and incomplete ablation particularly for large tumors, restricting their wide clinical usage. Attributed to their versatile physiochemical properties, biomaterials have been specially designed to potentiate local hyperthermia treatments according to their unique working principles. Meanwhile, biomaterial-based delivery systems are able to bridge hyperthermia therapies with other types of treatment strategies such as chemotherapy, radiotherapy and immunotherapy. Therefore, in this review, we discuss recent progress in the development of functional biomaterials to reinforce local hyperthermia by functioning as thermal sensitizers to endow more efficient tumor-localized thermal ablation and/or as delivery vehicles to synergize with other therapeutic modalities for combined cancer treatments. Thereafter, we provide a critical perspective on the further development of biomaterial-assisted local hyperthermia toward clinical applications.
Collapse
Affiliation(s)
- Yujie Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Quguang Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Chunjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, Zhejiang, P.R. China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P.R. China
| |
Collapse
|
23
|
Kim J, Bae H, Han HS, Lee J. Ultrasonic Enhancement of Chondrogenesis in Mesenchymal Stem Cells by Bolt-Clamped Langevin Transducers. MICROMACHINES 2023; 14:202. [PMID: 36677263 PMCID: PMC9865917 DOI: 10.3390/mi14010202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 06/17/2023]
Abstract
We recently investigated the design and fabrication of Langevin-type transducers for therapeutic ultrasound. Effect of ultrasonic energy arising from the transducer on biological tissue was examined. In this study, the transducer was set to radiate acoustic energy to mesenchymal stem cells (MSCs) for inducing differentiation into cartilage tissue. The average chondrogenic ratio in area was 20.82% in the control group, for which no external stimulation was given. Shear stress was applied to MSCs as the contrast group, which resulted in 42.66% on average with a 25.92% minimum rate; acoustic pressure from the flat tip causing transient cavitation enhanced chondrogenesis up to 52.96%. For the round tip excited by 20 Vpp, the maximum differentiation value of 69.43% was found, since it delivered relatively high acoustic pressure to MSCs. Hence, the results from this study indicate that ultrasound pressure at the kPa level can enhance MSC chondrogenesis compared to the tens of kHz range by Langevin transducers.
Collapse
Affiliation(s)
- Jinhyuk Kim
- Department of Electronic Engineering, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Hyuncheol Bae
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jungwoo Lee
- Department of Electronic Engineering, Kwangwoon University, Seoul 01897, Republic of Korea
| |
Collapse
|
24
|
Qin Y, Geng X, Sun Y, Zhao Y, Chai W, Wang X, Wang P. Ultrasound nanotheranostics: Toward precision medicine. J Control Release 2023; 353:105-124. [PMID: 36400289 DOI: 10.1016/j.jconrel.2022.11.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022]
Abstract
Ultrasound (US) is a mechanical wave that can penetrate biological tissues and trigger complex bioeffects. The mechanisms of US in different diagnosis and treatment are different, and the functional application of commercial US is also expanding. In particular, recent developments in nanotechnology have led to a wider use of US in precision medicine. In this review, we focus on US in combination with versatile micro and nanoparticles (NPs)/nanovesicles for tumor theranostics. We first introduce US-assisted drug delivery as a stimulus-responsive approach that spatiotemporally regulates the deposit of nanomedicines in target tissues. Multiple functionalized NPs and their US-regulated drug-release curves are analyzed in detail. Moreover, as a typical representative of US therapy, sonodynamic antitumor strategy is attracting researchers' attention. The collaborative efficiency and mechanisms of US and various nano-sensitizers such as nano-porphyrins and organic/inorganic nanosized sensitizers are outlined in this paper. A series of physicochemical processes during ultrasonic cavitation and NPs activation are also discussed. Finally, the new applications of US and diagnostic NPs in tumor-monitoring and image-guided combined therapy are summarized. Diagnostic NPs contain substances with imaging properties that enhance US contrast and photoacoustic imaging. The development of such high-resolution, low-background US-based imaging methods has contributed to modern precision medicine. It is expected that the integration of non-invasive US and nanotechnology will lead to significant breakthroughs in future clinical applications.
Collapse
Affiliation(s)
- Yang Qin
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaorui Geng
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yitong Zhao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Wenyu Chai
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
25
|
Centner CS, Moore JT, Baxter ME, Yaddanapudi K, Bates PJ, Kopechek JA. Comparison of Acoustofluidic and Static Systems for Ultrasound-Mediated Molecular Delivery to T Lymphocytes. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:90-105. [PMID: 36241589 DOI: 10.1016/j.ultrasmedbio.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 07/22/2022] [Accepted: 08/08/2022] [Indexed: 06/16/2023]
Abstract
Continuous-flow acoustofluidic technologies can potentially improve processing of T lymphocytes for cell therapies by addressing the limitations with viral and non-viral delivery methods. The objective of this study was to assess the intracellular delivery efficiency with acoustofluidic treatment compared with that of static ultrasound treatment. Optimization of parameters in acoustofluidic and static configurations was performed by assessing intracellular delivery of a fluorescent compound (calcein) in viable human Jurkat T lymphocytes. Ultrasound pressure and the concentration of cationic phospholipid-coated microbubbles influenced calcein delivery in both systems. In the static system, a treatment time of 45 s increased molecular delivery compared with 0-30 s (p < 0.01). Refined parameters were used to assess molecular delivery of small and large compounds (0.6-kDa calcein and 150-kDa fluorescein isothiocyanate-dextran, respectively) after ultrasound treatment with the acoustofluidic or static systems. Molecular delivery was similar with refined parameters for acoustofluidic treatment and static treatment (p > 0.05), even though acoustofluidic treatment had lower microbubble concentration (24 μg/mL vs. 94 μg/mL) and shorter treatment time (∼2-3 s vs. 45 s). This study indicates that the acoustofluidic system can significantly enhance intracellular molecular delivery, which could potentially enable acoustofluidic cell transfection during continuous flow processing for manufacture of cell therapies or other applications.
Collapse
Affiliation(s)
- Connor S Centner
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA
| | - John T Moore
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA
| | - Mary E Baxter
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA
| | | | - Paula J Bates
- School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jonathan A Kopechek
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA.
| |
Collapse
|
26
|
Zhang M, Zhang S, Shi J, Hu Y, Wu S, Zan Z, Zhao P, Gao C, Du Y, Wang Y, Lin F, Fu X, Li D, Qin P, Fan Z. Cell mechanical responses to subcellular perturbations generated by ultrasound and targeted microbubbles. Acta Biomater 2023; 155:471-481. [PMID: 36400351 DOI: 10.1016/j.actbio.2022.11.017] [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: 08/08/2022] [Revised: 10/19/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
The inherently dynamic and anisotropic microenvironment of cells imposes not only global and slow physical stimulations on cells but also acute and local perturbations. However, cell mechanical responses to transient subcellular physical signals remain unclear. In this study, acoustically activated targeted microbubbles were used to exert mechanical perturbations to single cells. The cellular contractile force was sensed by elastic micropillar arrays, while the pillar deformations were imaged using brightfield high-speed video microscopy at a frame rate of 1k frames per second for the first 10s and then confocal fluorescence microscopy. Cell mechanical responses are accompanied by cell membrane integrity changes. Both processes are determined by the perturbation strength generated by microbubble volumetric oscillations. The instantaneous cellular traction force relaxation exhibits two distinct patterns, correlated with two cell fates (survival or permanent damage). The mathematical modeling unveils that force-induced actomyosin disassembly leads to gradual traction force relaxation in the first few seconds. The perturbation may also influence the far end subcellular regions from the microbubbles and may propagate into connected cells with attenuations and delays. This study carefully characterizes the cell mechanical responses to local perturbations induced by ultrasound and microbubbles, advancing our understanding of the fundamentals of cell mechano-sensing, -responsiveness, and -transduction. STATEMENT OF SIGNIFICANCE: Subcellular physical perturbations commonly exist but haven't been fully explored yet. The subcellular perturbation generated by ultrasound and targeted microbubbles covers a wide range of strength, from mild, intermediate to intense, providing a broad biomedical relevance. With µm2 spatial sensing ability and up to 1ms temporal resolution, we present spatiotemporal details of the instantaneous cellular contractile force changes followed by attenuated and delayed global responses. The correlation between the cell mechanical responses and cell fates highlights the important role of the instantaneous mechanical responses in the entire cellular reactive processes. Supported by mathematical modeling, our work provides new insights into the dynamics and mechanisms of cell mechanics.
Collapse
Affiliation(s)
- Meiru Zhang
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Suyan Zhang
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Jianmin Shi
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yi Hu
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Shuying Wu
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Zhaoguang Zan
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Pu Zhao
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Changkai Gao
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Yanyao Du
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Yulin Wang
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Feng Lin
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Xing Fu
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Dachao Li
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China
| | - Peng Qin
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhenzhen Fan
- Department of Biomedical Engineering, Tianjin University, Tianjin 300072, China; State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
27
|
Cai Y, Fan K, Lin J, Ma L, Li F. Advances in BBB on Chip and Application for Studying Reversible Opening of Blood-Brain Barrier by Sonoporation. MICROMACHINES 2022; 14:112. [PMID: 36677173 PMCID: PMC9861620 DOI: 10.3390/mi14010112] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
The complex structure of the blood-brain barrier (BBB), which blocks nearly all large biomolecules, hinders drug delivery to the brain and drug assessment, thus decelerating drug development. Conventional in vitro models of BBB cannot mimic some crucial features of BBB in vivo including a shear stress environment and the interaction between different types of cells. There is a great demand for a new in vitro platform of BBB that can be used for drug delivery studies. Compared with in vivo models, an in vitro platform has the merits of low cost, shorter test period, and simplicity of operation. Microfluidic technology and microfabrication are good tools in rebuilding the BBB in vitro. During the past decade, great efforts have been made to improve BBB penetration for drug delivery using biochemical or physical stimuli. In particular, compared with other drug delivery strategies, sonoporation is more attractive due to its minimized systemic exposure, high efficiency, controllability, and reversible manner. BBB on chips (BOC) holds great promise when combined with sonoporation. More details and mechanisms such as trans-endothelial electrical resistance (TEER) measurements and dynamic opening of tight junctions can be figured out when using sonoporation stimulating BOC, which will be of great benefit for drug development. Herein, we discuss the recent advances in BOC and sonoporation for BBB disruption with this in vitro platform.
Collapse
Affiliation(s)
- Yicong Cai
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518107, China
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Kexin Fan
- School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Jiawei Lin
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518107, China
| | - Lin Ma
- School of Physics and Optoelectronic Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Fenfang Li
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518107, China
| |
Collapse
|
28
|
Zheng ZY, Feng CH, Xie G, Liu WL, Zhu XL. Proteolysis Degree of Protein Corona Affect Ultrasound-Induced Sublethal Effects on Saccharomyces cerevisiae: Transcriptomics Analysis and Adaptive Regulation of Membrane Homeostasis. Foods 2022; 11:3883. [PMID: 36496692 PMCID: PMC9735630 DOI: 10.3390/foods11233883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/17/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Protein corona (PC) adsorbed on the surface of nanoparticles brings new research perspectives on the interaction between nanoparticles and fermentative microorganisms. Herein, the proteolysis of wheat PC adsorbed on a nano-Se surface using cell-free protease extract from S. cerevisiae was conducted. The proteolysis caused monotonic changes of ζ-potentials and surface hydrophobicity of PC. Notably, the innermost PC layer was difficult to be proteolyzed. Furthermore, when S. cerevisiae was stimulated by ultrasound + 0.1 mg/mL nano-Se@PC, the proportion of lethal and sublethal injured cells increased as a function of the proteolysis time of PC. The transcriptomics analysis revealed that 34 differentially expressed genes which varied monotonically were related to the plasma membrane, fatty acid metabolism, glycerolipid metabolism, etc. Significant declines in the membrane potential and proton motive force disruption of membrane were found with the prolonged proteolysis time; meanwhile, higher membrane permeability, membrane oxidative stress levels, membrane lipid fluidity, and micro-viscosity were triggered.
Collapse
Affiliation(s)
- Zi-Yi Zheng
- School of Material Science and Food Engineering, University of Electronic Science and Technology of China, Zhongshan Institute, Zhongshan 528402, China
| | | | | | | | | |
Collapse
|
29
|
Barzegar-Fallah A, Gandhi K, Rizwan SB, Slatter TL, Reynolds JNJ. Harnessing Ultrasound for Targeting Drug Delivery to the Brain and Breaching the Blood–Brain Tumour Barrier. Pharmaceutics 2022; 14:pharmaceutics14102231. [PMID: 36297666 PMCID: PMC9607160 DOI: 10.3390/pharmaceutics14102231] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant advances in developing drugs to treat brain tumours, achieving therapeutic concentrations of the drug at the tumour site remains a major challenge due to the presence of the blood–brain barrier (BBB). Several strategies have evolved to enhance brain delivery of chemotherapeutic agents to treat tumours; however, most approaches have several limitations which hinder their clinical utility. Promising studies indicate that ultrasound can penetrate the skull to target specific brain regions and transiently open the BBB, safely and reversibly, with a high degree of spatial and temporal specificity. In this review, we initially describe the basics of therapeutic ultrasound, then detail ultrasound-based drug delivery strategies to the brain and the mechanisms by which ultrasound can improve brain tumour therapy. We review pre-clinical and clinical findings from ultrasound-mediated BBB opening and drug delivery studies and outline current therapeutic ultrasound devices and technologies designed for this purpose.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Kushan Gandhi
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Shakila B. Rizwan
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand
| | - Tania L. Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - John N. J. Reynolds
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- Correspondence: ; Tel.: +64-3-479-5781; Fax: +64-3-479-7254
| |
Collapse
|
30
|
Przystupski D, Ussowicz M. Landscape of Cellular Bioeffects Triggered by Ultrasound-Induced Sonoporation. Int J Mol Sci 2022; 23:ijms231911222. [PMID: 36232532 PMCID: PMC9569453 DOI: 10.3390/ijms231911222] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Sonoporation is the process of transient pore formation in the cell membrane triggered by ultrasound (US). Numerous studies have provided us with firm evidence that sonoporation may assist cancer treatment through effective drug and gene delivery. However, there is a massive gap in the body of literature on the issue of understanding the complexity of biophysical and biochemical sonoporation-induced cellular effects. This study provides a detailed explanation of the US-triggered bioeffects, in particular, cell compartments and the internal environment of the cell, as well as the further consequences on cell reproduction and growth. Moreover, a detailed biophysical insight into US-provoked pore formation is presented. This study is expected to review the knowledge of cellular effects initiated by US-induced sonoporation and summarize the attempts at clinical implementation.
Collapse
|
31
|
Zevnik J, Dular M. Cavitation bubble interaction with compliant structures on a microscale: A contribution to the understanding of bacterial cell lysis by cavitation treatment. ULTRASONICS SONOCHEMISTRY 2022; 87:106053. [PMID: 35690044 PMCID: PMC9190065 DOI: 10.1016/j.ultsonch.2022.106053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 05/09/2023]
Abstract
Numerous studies have already shown that the process of cavitation can be successfully used for water treatment and eradication of bacteria. However, most of the relevant studies are being conducted on a macro scale, so the understanding of the processes at a fundamental level remains poor. In attempt to further elucidate the process of cavitation-assisted water treatment on a scale of a single bubble, the present paper numerically addresses interaction between a collapsing microbubble and a nearby compliant structure, that mechanically and structurally resembles a bacterial cell. A fluid-structure interaction methodology is employed, where compressible multiphase flow is considered and the bacterial cell wall is modeled as a multi-layered shell structure. Simulations are performed for two selected model structures, each resembling the main structural features of Gram-negative and Gram-positive bacterial cell envelopes. The contribution of two independent dimensionless geometric parameters is investigated, namely the bubble-cell distance δ and their size ratio ς. Three characteristic modes of bubble collapse dynamics and four modes of spatiotemporal occurrence of peak local stresses in the bacterial cell membrane are identified throughout the parameter space considered. The former range from the development of a weak and thin jet away from the cell to spherical bubble collapses. The results show that local stresses arising from bubble-induced loads can exceed poration thresholds of cell membranes and that bacterial cell damage could be explained solely by mechanical effects in absence of thermal and chemical ones. Based on this, the damage potential of a single microbubble for bacteria eradication is estimated, showing a higher resistance of the Gram-positive model organism to the nearby bubble collapse. Microstreaming is identified as the primary mechanical mechanism of bacterial cell damage, which in certain cases may be enhanced by the occurrence of shock waves during bubble collapse. The results are also discussed in the scope of bacteria eradication by cavitation treatment on a macro scale, where processes of hydrodynamic and ultrasonic cavitation are being employed.
Collapse
Affiliation(s)
- Jure Zevnik
- University of Ljubljana, Faculty of Mechanical Engineering, Aškerčeva cesta 6, Ljubljana, Slovenia.
| | - Matevž Dular
- University of Ljubljana, Faculty of Mechanical Engineering, Aškerčeva cesta 6, Ljubljana, Slovenia
| |
Collapse
|
32
|
Light triggered nanoscale biolistics for efficient intracellular delivery of functional macromolecules in mammalian cells. Nat Commun 2022; 13:1996. [PMID: 35422038 PMCID: PMC9010410 DOI: 10.1038/s41467-022-29713-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Biolistic intracellular delivery of functional macromolecules makes use of dense microparticles which are ballistically fired onto cells with a pressurized gun. While it has been used to transfect plant cells, its application to mammalian cells has met with limited success mainly due to high toxicity. Here we present a more refined nanotechnological approach to biolistic delivery with light-triggered self-assembled nanobombs (NBs) that consist of a photothermal core particle surrounded by smaller nanoprojectiles. Upon irradiation with pulsed laser light, fast heating of the core particle results in vapor bubble formation, which propels the nanoprojectiles through the cell membrane of nearby cells. We show successful transfection of both adherent and non-adherent cells with mRNA and pDNA, outperforming electroporation as the most used physical transfection technology by a factor of 5.5–7.6 in transfection yield. With a throughput of 104-105 cells per second, biolistic delivery with NBs offers scalable and highly efficient transfections of mammalian cells. Ballistic delivery with micro/nano-particles has been successfully used to transfect plant cells, however, has failed in mammalian cells due to toxic effects. Here, the authors report on a self-assembled nano-ballistic delivery system for the delivery of functional macromolecules and demonstrate efficient transfection of mammalian cells.
Collapse
|
33
|
Nance E, Pun SH, Saigal R, Sellers DL. Drug delivery to the central nervous system. NATURE REVIEWS. MATERIALS 2022; 7:314-331. [PMID: 38464996 PMCID: PMC10923597 DOI: 10.1038/s41578-021-00394-w] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 03/12/2024]
Abstract
Despite the rising global incidence of central nervous system (CNS) disorders, CNS drug development remains challenging, with high costs, long pathways to clinical use and high failure rates. The CNS is highly protected by physiological barriers, in particular, the blood-brain barrier and the blood-cerebrospinal fluid barrier, which limit access of most drugs. Biomaterials can be designed to bypass or traverse these barriers, enabling the controlled delivery of drugs into the CNS. In this Review, we first examine the effects of normal and diseased CNS physiology on drug delivery to the brain and spinal cord. We then discuss CNS drug delivery designs and materials that are administered systemically, directly to the CNS, intranasally or peripherally through intramuscular injections. Finally, we highlight important challenges and opportunities for materials design for drug delivery to the CNS and the anticipated clinical impact of CNS drug delivery.
Collapse
Affiliation(s)
- Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Suzie H. Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Rajiv Saigal
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| | - Drew L. Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- These authors contributed equally: Elizabeth Nance, Suzie H. Pun, Rajiv Saigal, Drew L. Sellers
| |
Collapse
|
34
|
Ultrasound-mediated gene delivery of factor VIII plasmids for hemophilia A gene therapy in mice. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 27:916-926. [PMID: 35141050 PMCID: PMC8803955 DOI: 10.1016/j.omtn.2022.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023]
Abstract
Gene therapy offers great promises for a cure of hemophilia A resulting from factor VIII (FVIII) gene deficiency. We have developed and optimized a non-viral ultrasound-mediated gene delivery (UMGD) strategy. UMGD of reporter plasmids targeting mice livers achieved high levels of transgene expression predominantly in hepatocytes. Following UMGD of a plasmid encoding human FVIII driven by a hepatocyte-specific promoter/enhancer (pHP-hF8/N6) into the livers of hemophilia A mice, a partial phenotypic correction was achieved in treated mice. In order to achieve persistent and therapeutic FVIII gene expression, we adopted a plasmid (pHP-hF8-X10) encoding an FVIII variant with significantly increased FVIII secretion. By employing an optimized pulse-train ultrasound condition and immunomodulation, the treated hemophilia A mice achieved 25%–150% of FVIII gene expression on days 1–7 with very mild transient liver damage, as indicated by a small increase of transaminase levels that returned to normal within 3 days. Therapeutic levels of FVIII can be maintained persistently without the generation of inhibitors in mice. These results indicate that UMGD can significantly enhance the efficiency of plasmid DNA transfer into the liver. They also demonstrate the potential of this novel technology to safely and effectively treat hemophilia A.
Collapse
|
35
|
Tu J, Yu ACH. Ultrasound-Mediated Drug Delivery: Sonoporation Mechanisms, Biophysics, and Critical Factors. BME FRONTIERS 2022; 2022:9807347. [PMID: 37850169 PMCID: PMC10521752 DOI: 10.34133/2022/9807347] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/31/2021] [Indexed: 10/19/2023] Open
Abstract
Sonoporation, or the use of ultrasound in the presence of cavitation nuclei to induce plasma membrane perforation, is well considered as an emerging physical approach to facilitate the delivery of drugs and genes to living cells. Nevertheless, this emerging drug delivery paradigm has not yet reached widespread clinical use, because the efficiency of sonoporation is often deemed to be mediocre due to the lack of detailed understanding of the pertinent scientific mechanisms. Here, we summarize the current observational evidence available on the notion of sonoporation, and we discuss the prevailing understanding of the physical and biological processes related to sonoporation. To facilitate systematic understanding, we also present how the extent of sonoporation is dependent on a multitude of factors related to acoustic excitation parameters (ultrasound frequency, pressure, cavitation dose, exposure time), microbubble parameters (size, concentration, bubble-to-cell distance, shell composition), and cellular properties (cell type, cell cycle, biochemical contents). By adopting a science-backed approach to the realization of sonoporation, ultrasound-mediated drug delivery can be more controllably achieved to viably enhance drug uptake into living cells with high sonoporation efficiency. This drug delivery approach, when coupled with concurrent advances in ultrasound imaging, has potential to become an effective therapeutic paradigm.
Collapse
Affiliation(s)
- Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing, China
| | - Alfred C. H. Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
36
|
Liu X, Zhang W, Jing Y, Yi S, Farooq U, Shi J, Pang N, Rong N, Xu L. Non-Cavitation Targeted Microbubble-Mediated Single-Cell Sonoporation. MICROMACHINES 2022; 13:mi13010113. [PMID: 35056278 PMCID: PMC8780975 DOI: 10.3390/mi13010113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023]
Abstract
Sonoporation employs ultrasound accompanied by microbubble (MB) cavitation to induce the reversible disruption of cell membranes and has been exploited as a promising intracellular macromolecular delivery strategy. Due to the damage to cells resulting from strong cavitation, it is difficult to balance efficient delivery and high survival rates. In this paper, a traveling surface acoustic wave (TSAW) device, consisting of a TSAW chip and a polydimethylsiloxane (PDMS) channel, was designed to explore single-cell sonoporation using targeted microbubbles (TMBs) in a non-cavitation regime. A TSAW was applied to precisely manipulate the movement of the TMBs attached to MDA-MB-231 cells, leading to sonoporation at a single-cell level. The impact of input voltage and the number of TMBs on cell sonoporation was investigated. In addition, the physical mechanisms of bubble cavitation or the acoustic radiation force (ARF) for cell sonoporation were analyzed. The TMBs excited by an ARF directly propelled cell membrane deformation, leading to reversible perforation in the cell membrane. When two TMBs adhered to the cell surface and the input voltage was 350 mVpp, the cell sonoporation efficiency went up to 83%.
Collapse
Affiliation(s)
- Xiufang Liu
- College of Medicine and Biological Information Engineering, Northeastern University, 195 Innovation Road, Shenyang 110016, China; (X.L.); (N.P.)
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Wenjun Zhang
- Department of Mechanical and Electrical Engineering, Gannan University of Science and Technology, 156 Kejia Avenue, Ganzhou 341000, China;
| | - Yanshu Jing
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
- Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Shasha Yi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Umar Farooq
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Jingyao Shi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Na Pang
- College of Medicine and Biological Information Engineering, Northeastern University, 195 Innovation Road, Shenyang 110016, China; (X.L.); (N.P.)
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Ning Rong
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
- Correspondence: (N.R.); (L.X.); Tel.: +86-024-83683200 (L.X.)
| | - Lisheng Xu
- College of Medicine and Biological Information Engineering, Northeastern University, 195 Innovation Road, Shenyang 110016, China; (X.L.); (N.P.)
- Neusoft Research of Intelligent Healthcare Technology, Co., Ltd., Shenyang 110167, China
- Correspondence: (N.R.); (L.X.); Tel.: +86-024-83683200 (L.X.)
| |
Collapse
|
37
|
Applications of Ultrasound-Mediated Drug Delivery and Gene Therapy. Int J Mol Sci 2021; 22:ijms222111491. [PMID: 34768922 PMCID: PMC8583720 DOI: 10.3390/ijms222111491] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Gene therapy has continuously evolved throughout the years since its first proposal to develop more specific and effective transfection, capable of treating a myriad of health conditions. Viral vectors are some of the most common and most efficient vehicles for gene transfer. However, the safe and effective delivery of gene therapy remains a major obstacle. Ultrasound contrast agents in the form of microbubbles have provided a unique solution to fulfill the need to shield the vectors from the host immune system and the need for site specific targeted therapy. Since the discovery of the biophysical and biological effects of microbubble sonification, multiple developments have been made to enhance its applicability in targeted drug delivery. The concurrent development of viral vectors and recent research on dual vector strategies have shown promising results. This review will explore the mechanisms and recent advancements in the knowledge of ultrasound-mediated microbubbles in targeting gene and drug therapy.
Collapse
|
38
|
Zevnik J, Dular M. Liposome destruction by a collapsing cavitation microbubble: A numerical study. ULTRASONICS SONOCHEMISTRY 2021; 78:105706. [PMID: 34411844 PMCID: PMC8379499 DOI: 10.1016/j.ultsonch.2021.105706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/16/2021] [Accepted: 07/30/2021] [Indexed: 05/07/2023]
Abstract
Hydrodynamic cavitation poses as a promising new method for wastewater treatment as it has been shown to be able to eradicate bacteria, inactivate viruses, and destroy other biological structures, such as liposomes. Although engineers are already commercializing devices that employ cavitation, we are still not able to answer the fundamental question: What exactly are the damaging mechanisms of hydrodynamic cavitation in various applications? In this light, the present paper numerically addresses the interaction between a single cavitation microbubble and a nearby lipid vesicle of a similar size. A coupled fluid-structure interaction model is employed, from which three critical modes of vesicle deformation are identified and temporally placed in relation to their corresponding driving mechanisms: (a) unilateral stretching at the waist of the liposome during the first bubble collapse and subsequent shock wave propagation, (b) local wrinkling at the tip until the bubble rebounds, and (c) bilateral stretching at the tip of the liposome during the phase of a second bubble contraction. Here, unilateral and bilateral stretching refer to the local in-plane extension of the bilayer in one and both principal directions, respectively. Results are discussed with respect to critical dimensionless distance for vesicle poration and rupture. Liposomes with initially equilibrated envelopes are not expected to be structurally compromised in cases with δ>1.0, when a nearby collapsing bubble is not in their direct contact. However, the critical dimensionless distance for the case of an envelope with pre-existing pores is identified at δ=1.9. Additionally, the influence of liposome-bubble size ratio is addressed, from which a higher potential of larger bubbles for causing stretching-induced liposome destruction can be identified.
Collapse
Affiliation(s)
- Jure Zevnik
- University of Ljubljana, Faculty of Mechanical Engineering, Aškerčeva cesta 6, Ljubljana, Slovenia.
| | - Matevž Dular
- University of Ljubljana, Faculty of Mechanical Engineering, Aškerčeva cesta 6, Ljubljana, Slovenia
| |
Collapse
|
39
|
Sanwal R, Joshi K, Ditmans M, Tsai SSH, Lee WL. Ultrasound and Microbubbles for Targeted Drug Delivery to the Lung Endothelium in ARDS: Cellular Mechanisms and Therapeutic Opportunities. Biomedicines 2021; 9:biomedicines9070803. [PMID: 34356867 PMCID: PMC8301318 DOI: 10.3390/biomedicines9070803] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by increased permeability of the alveolar–capillary membrane, a thin barrier composed of adjacent monolayers of alveolar epithelial and lung microvascular endothelial cells. This results in pulmonary edema and severe hypoxemia and is a common cause of death after both viral (e.g., SARS-CoV-2) and bacterial pneumonia. The involvement of the lung in ARDS is notoriously heterogeneous, with consolidated and edematous lung abutting aerated, less injured regions. This makes treatment difficult, as most therapeutic approaches preferentially affect the normal lung regions or are distributed indiscriminately to other organs. In this review, we describe the use of thoracic ultrasound and microbubbles (USMB) to deliver therapeutic cargo (drugs, genes) preferentially to severely injured areas of the lung and in particular to the lung endothelium. While USMB has been explored in other organs, it has been under-appreciated in the treatment of lung injury since ultrasound energy is scattered by air. However, this limitation can be harnessed to direct therapy specifically to severely injured lungs. We explore the cellular mechanisms governing USMB and describe various permutations of cargo administration. Lastly, we discuss both the challenges and potential opportunities presented by USMB in the lung as a tool for both therapy and research.
Collapse
Affiliation(s)
- Rajiv Sanwal
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kushal Joshi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Mihails Ditmans
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Scott S. H. Tsai
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence: ; Tel.: +416-864-6060 (ext. 77655)
| |
Collapse
|
40
|
Snipstad S, Hanstad S, Bjørkøy A, Mørch Ý, de Lange Davies C. Sonoporation Using Nanoparticle-Loaded Microbubbles Increases Cellular Uptake of Nanoparticles Compared to Co-Incubation of Nanoparticles and Microbubbles. Pharmaceutics 2021; 13:640. [PMID: 33946327 PMCID: PMC8146007 DOI: 10.3390/pharmaceutics13050640] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022] Open
Abstract
Therapeutic agents can benefit from encapsulation in nanoparticles, due to improved pharmacokinetics and biodistribution, protection from degradation, increased cellular uptake and sustained release. Microbubbles in combination with ultrasound have been shown to improve the delivery of nanoparticles and drugs to tumors and across the blood-brain barrier. Here, we evaluate two different microbubbles for enhancing the delivery of polymeric nanoparticles to cells in vitro: a commercially available lipid microbubble (Sonazoid) and a microbubble with a shell composed of protein and nanoparticles. Various ultrasound parameters are applied and confocal microscopy is employed to image cellular uptake. Ultrasound enhanced cellular uptake depending on the pressure and duty cycle. The responsible mechanisms are probably sonoporation and sonoprinting, followed by uptake, and to a smaller degree enhanced endocytosis. The use of commercial Sonazoid microbubbles leads to significantly lower uptake than when using nanoparticle-loaded microbubbles, suggesting that proximity between cells, nanoparticles and microbubbles is important, and that mainly nanoparticles in the shell are taken up, rather than free nanoparticles in solution.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
- Cancer Clinic, St. Olav’s Hospital, Prinsesse Kristinas Gate 1, 7030 Trondheim, Norway
| | - Sigurd Hanstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| |
Collapse
|
41
|
Li Y, Chen Z, Ge S. Sonoporation: Underlying Mechanisms and Applications in Cellular Regulation. BIO INTEGRATION 2021. [DOI: 10.15212/bioi-2020-0028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ultrasound combined with microbubble-mediated sonoporation has been applied to enhance drug or gene intracellular delivery. Sonoporation leads to the formation of openings in the cell membrane, triggered by ultrasound-mediated oscillations and destruction of microbubbles. Multiple mechanisms
are involved in the occurrence of sonoporation, including ultrasonic parameters, microbubbles size, and the distance of microbubbles to cells. Recent advances are beginning to extend applications through the assistance of contrast agents, which allow ultrasound to connect directly to cellular
functions such as gene expression, cellular apoptosis, differentiation, and even epigenetic reprogramming. In this review, we summarize the current state of the art concerning microbubble‐cell interactions and sonoporation effects leading to cellular functions.
Collapse
Affiliation(s)
- Yue Li
- First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhiyi Chen
- First Affiliated Hospital of University of South China, Hengyang, China
| | - Shuping Ge
- Department of Pediatrics, St Christopher’s Hospital for Children, Tower Health and Drexel University, Philadelphia, PA (S.G.)
| |
Collapse
|
42
|
Zevnik J, Dular M. Cavitation bubble interaction with a rigid spherical particle on a microscale. ULTRASONICS SONOCHEMISTRY 2020; 69:105252. [PMID: 32682313 DOI: 10.1016/j.ultsonch.2020.105252] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/27/2020] [Accepted: 07/06/2020] [Indexed: 05/09/2023]
Abstract
Cavitation bubble collapse close to a submerged sphere on a microscale is investigated numerically using a finite volume method in order to determine the likelihood of previously suspected mechanical effects to cause bacterial cell damage, such as impact of a high speed water jet, propagation of bubble emitted shock waves, shear loads, and thermal loads. A grid convergence study and validation of the employed axisymmetric numerical model against the Gilmore's equation is performed for a case of a single microbubble collapse due to a sudden ambient pressure increase. Numerical simulations of bubble-sphere interaction corresponding to different values of nondimensional bubble-sphere standoff distance δ and their size ratio ε are carried out. The obtained results show vastly different bubble collapse dynamics across the considered parameter space, from the development of a fast thin annular jet towards the sphere to an almost spherical bubble collapse. Although some similarities in bubble shape progression to previous studies on larger bubbles exist, it can be noticed that bubble jetting is much less likely to occur on the considered scale due to the cushioning effects of surface tension on the intensity of the collapse. Overall, the results show that the mechanical loads on a spherical particle tend to increase with a sphere-bubble size ratio ε, and decrease with their distance δ. Additionally, the results are discussed with respect to bacteria eradication by hydrodynamic cavitation. Potentially harmful mechanical effects of bubble-sphere interaction on a micro scale are identified, namely the collapse-induced shear loads with peaks of a few megapascals and propagation of bubble emitted shock waves, which could cause spatially highly variable compressive loads with peaks of a few hundred megapascals and gradients of 100 MPa/μm.
Collapse
Affiliation(s)
- Jure Zevnik
- University of Ljubljana, Faculty of Mechanical Engineering, Aškerčeva cesta 6, Ljubljana, Slovenia.
| | - Matevž Dular
- University of Ljubljana, Faculty of Mechanical Engineering, Aškerčeva cesta 6, Ljubljana, Slovenia
| |
Collapse
|
43
|
Harizaj A, De Smedt SC, Lentacker I, Braeckmans K. Physical transfection technologies for macrophages and dendritic cells in immunotherapy. Expert Opin Drug Deliv 2020; 18:229-247. [PMID: 32985919 DOI: 10.1080/17425247.2021.1828340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Dendritic cells (DCs) and macrophages, two important antigen presenting cells (APCs) of the innate immune system, are being explored for the use in cell-based cancer immunotherapy. For this application, the therapeutic potential of patient-derived APCs is increased by delivering different types of functional macromolecules, such as mRNA and pDNA, into their cytosol. Compared to the use of viral and non-viral delivery vectors, physical intracellular delivery techniques are known to be more straightforward, more controllable, faster and generate high delivery efficiencies. AREAS COVERED This review starts with electroporation as the most traditional physical transfection method, before continuing with the more recent technologies such as sonoporation, nanowires and microfluidic cell squeezing. A description is provided of each of those intracellular delivery technologies with their strengths and weaknesses, especially paying attention to delivery efficiency and safety profile. EXPERT OPINION Given the common use of electroporation for the production of therapeutic APCs, it is recommended that more detailed studies are performed on the effect of electroporation on APC fitness, even down to the genetic level. Newer intracellular delivery technologies seem to have less impact on APC functionality but further work is needed to fully uncover their suitability to transfect APCs with different types of macromolecules.
Collapse
Affiliation(s)
- Aranit Harizaj
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| |
Collapse
|
44
|
Yang Y, Li Q, Guo X, Tu J, Zhang D. Mechanisms underlying sonoporation: Interaction between microbubbles and cells. ULTRASONICS SONOCHEMISTRY 2020; 67:105096. [PMID: 32278246 DOI: 10.1016/j.ultsonch.2020.105096] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 05/04/2023]
Abstract
The past several decades have witnessed great progress in "smart drug delivery", an advance technology that can deliver genes or drugs into specific locations of patients' body with enhanced delivery efficiency. Ultrasound-activated mechanical force induced by the interactions between microbubbles and cells, which can stimulate so-called "sonoporation" process, has been regarded as one of the most promising candidates to realize spatiotemporal-controllable drug delivery to selected regions. Both experimental and numerical studies were performed to get in-depth understanding on how the microbubbles interact with cells during sonoporation processes, under different impact parameters. The current work gives an overview of the general mechanism underlying microbubble-mediated sonoporation, and the possible impact factors (e.g., the properties of cavitation agents and cells, acoustical driving parameters and bubble/cell micro-environment) that could affect sonoporation outcomes. Finally, current progress and considerations of sonoporation in clinical applications are reviewed also.
Collapse
Affiliation(s)
- Yanye Yang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Qunying Li
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China.
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China
| |
Collapse
|
45
|
Clinical and Microbiological Effects of Weekly Supragingival Irrigation with Aerosolized 0.5% Hydrogen Peroxide and Formation of Cavitation Bubbles in Gingival Tissues after This Irrigation: A Six-Month Randomized Clinical Trial. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3852431. [PMID: 32802264 PMCID: PMC7415088 DOI: 10.1155/2020/3852431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/08/2020] [Accepted: 07/07/2020] [Indexed: 11/30/2022]
Abstract
Introduction The study investigated the effect of weekly supragingival irrigation with aerosolized 0.5% hydrogen peroxide (H2O2) solution as a maintenance periodontal therapy on clinical and microbiological parameters in patients with chronic periodontitis. The other purpose was to investigate whether cavitation bubbles can penetrate not only into periodontitis-damaged tissues but also into ex vivo porcine healthy periodontal tissues. Materials and Methods The study included 35 systemically healthy patients with chronic periodontitis (CP). After nonsurgical periodontal debridement (NSPD), all patients were randomized into two groups: the Control group (NSDP alone, n = 18) and the Test group (NSDP plus supragingival irrigation, n = 17). Clinical (Approximal Plaque Index (API), Bleeding Index (BI), and Modified Gingival Index (MGI)) and microbiological (Polymerase Chain Reaction technology (using a micro-IDent® kit)) measurements were performed at the initial time point, 3 months, and 6 months after NSPD. The impact of supragingival irrigation on diseased gingival tissues of CP patients (n = 5) and on ex vivo porcine healthy gingival tissue samples (n = 3) was evaluated to estimate morphological changes in healthy and diseased gingival tissues. Results Morphological data revealed that supragingival irrigation caused the formation of cavitation bubbles in diseased gingival tissue of CP patients and in healthy porcine gingival tissues. The decrease in API, BI, and MGI scores after 6 months in the Test group significantly (p ≤ 0.01, p ≤ 0.05, and p ≤ 0.01, respectively) exceeded that in the Control group. Test group patients demonstrated a decrease in periodontal sites showing Pocket Probing Depth > 4 mm and, after 6 months, a statistically significant decrease in the proportion of periopathogenic bacteria. Conclusion The effectiveness of mechanical periodontal treatment combined with weekly supragingival irrigation with aerosolized 0.5% H2O2 solution on clinical and microbiological parameters of periodontal tissues of periodontitis patients is reliably higher than that of mechanical periodontal debridement alone. It has been found that cavitation bubbles as a result of irrigation with the aerosolized 0.5% hydrogen peroxide solution can form not only in periodontal tissues of periodontitis patients but also in ex vivo porcine healthy gingival tissues.
Collapse
|
46
|
The relation of Bleomycin Delivery Efficiency to Microbubble Sonodestruction and Cavitation Spectral Characteristics. Sci Rep 2020; 10:7743. [PMID: 32385397 PMCID: PMC7210292 DOI: 10.1038/s41598-020-64213-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/09/2020] [Indexed: 12/26/2022] Open
Abstract
The concurrent assessment of principal sonoporation factors has been accomplished in a single systemic study. Microbubble sonodestruction dynamics and cavitation spectral characteristics, ultrasound scattering and attenuation, were examined in relation to the intracellular delivery of anticancer drug, bleomycin. Experiments were conducted on Chinese hamster ovary cells coadministered with Sonovue microbubbles. Detailed analysis of the scattering and attenuation temporal functions culminated in quantification of metrics, inertial cavitation dose and attenuation rate, suitable for cavitation control. The exponents, representing microbubble sonodestruction kinetics were exploited to derive dosimetric, microbubble sonodestruction rate. High intracorrelation between empirically-attained metrics defines the relations which indicate deep physical interdependencies within inherent phenomena. Subsequently each quantified metric was validated to be well-applicable to prognosticate the efficacy of bleomycin delivery and cell viability, as indicated by strong overall correlation (R2 > 0.85). Presented results draw valuable insights in sonoporation dosimetry and contribute towards the development of universal sonoporation dosimetry model. Both bleomycin delivery and cell viability reach their respective plateau levels by the time, required to attain total microbubble sonodestruction, which accord with scattering and attenuation decrease to background levels. This suggests a well-defined criterion, feasible through signal-registration, universally employable to set optimal duration of exposure for efficient sonoporation outcome.
Collapse
|
47
|
Liu Q, Jiang J, Tang L, Chen M. The effect of low frequency and low intensity ultrasound combined with microbubbles on the sonoporation efficiency of MDA-MB-231 cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:298. [PMID: 32355742 PMCID: PMC7186677 DOI: 10.21037/atm.2020.02.155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Ultrasound can produce certain biophysical effects including thermal and non-thermal effects on cells. Sonoporation, the most widely studied non-thermal biological effect of ultrasound, is considered to be the basis for new therapeutic applications. Ultrasound irradiation can increase the permeability of cell membranes through sonoporous effect, which makes molecules like those of drugs, protein, and DNA that normally cannot pass through the cell membranes be able to enter cells. Considering the poor therapeutic effect and poor prognosis of triple negative breast cancer, we aimed to explore the experimental conditions and find the optimal parameters to improve the therapeutic efficacy of chemotherapeutic drugs for MDA-MB-231 cells. Methods By establishing an experimental and control group, our study investigated the effect of low frequency and low intensity ultrasound combined with microbubbles on MDA-MB-231 cell membrane permeability at different times. We conducted factorial cross-design and set 3 levels of ultrasound intensity: 230, 300, and 370 mW/cm2; 3 levels of irradiation time: 1, 2, and 3 minutes; and 6 levels of microbubble doses: 0, 0.2, 0.4, 0.6, 0.8, and 1 mL. Results Results show that ultrasound intensity, time of irradiation, and microbubbles concentration are not only related to but also have interactive effects on the sonoporation efficiency of MDA-MB-231 cells, with the rank order being sound intensity, irradiation time, and microbubble concentration. The average positive rates (%) of FD4 staining in sound intensities of 230, 300, and 370 mW/cm2 levels were 1.20±0.71, 13.80±5.86, and 10.71±4.36, respectively; and in irradiated times of 1, 2, and 3 min they were 7.54±5.95, 9.74±8.42, and 8.59±5.80, respectively. When the microbubbles increased according to the gradient of 0, 0.2, 0.4, 0.6, 0.8, and 1 mL, the positive rates (%) of FD4 staining were 7.32±5.89, 9.26±7.39, 8.31±5.67, 10.12±8.42, 8.67±7.23, and 7.72±6.24. Conclusions In our study, the optimal parameters of the sonoporous effect for MDA-MB-231 cells were 300 mW/cm2 of ultrasound intensity, 2 minutes of irradiation time, and 20% microbubbles concentration.
Collapse
Affiliation(s)
- Qi Liu
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Jianwei Jiang
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Lei Tang
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Man Chen
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| |
Collapse
|
48
|
Opening of endothelial cell-cell contacts due to sonoporation. J Control Release 2020; 322:426-438. [PMID: 32246975 DOI: 10.1016/j.jconrel.2020.03.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 01/06/2023]
Abstract
Ultrasound insonification of microbubbles can locally increase vascular permeability to enhance drug delivery. To control and optimize the therapeutic potential, we need to better understand the underlying biological mechanisms of the drug delivery pathways. The aim of this in vitro study was to elucidate the microbubble-endothelial cell interaction using the Brandaris 128 ultra-high-speed camera (up to 25 Mfps) coupled to a custom-built Nikon confocal microscope, to visualize both microbubble oscillation and the cellular response. Sonoporation and opening of cell-cell contacts by single αVβ3-targeted microbubbles (n = 152) was monitored up to 4 min after ultrasound insonification (2 MHz, 100-400 kPa, 10 cycles). Sonoporation occurred when microbubble excursion amplitudes exceeded 0.7 μm. Quantification of the influx of the fluorescent model drug propidium iodide upon sonoporation showed that the size of the created pore increased for larger microbubble excursion amplitudes. Microbubble-mediated opening of cell-cell contacts occurred as a cellular response upon sonoporation and did not correlate with the microbubble excursion amplitude itself. The initial integrity of the cell-cell contacts affected the susceptibly to drug delivery, since cell-cell contacts opened more often when cells were only partially attached to their neighbors (48%) than when fully attached (14%). The drug delivery outcomes were independent of nonlinear microbubble behavior, microbubble location, and cell size. In conclusion, by studying the microbubble-cell interaction at nanosecond and nanometer resolution the relationship between drug delivery pathways and their underlying mechanisms was further unraveled. These novel insights will aid the development of safe and efficient microbubble-mediated drug delivery.
Collapse
|
49
|
Lin YC, Chen HC, Chen HK, Lin YY, Kuo CY, Wang H, Hung CL, Shih CP, Wang CH. Ultrastructural Changes Associated With the Enhanced Permeability of the Round Window Membrane Mediated by Ultrasound Microbubbles. Front Pharmacol 2020; 10:1580. [PMID: 32047431 PMCID: PMC6997169 DOI: 10.3389/fphar.2019.01580] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022] Open
Abstract
The round window membrane (RWM) is the most common entryway for local drug and gene delivery into the inner ear, but its permeability can change the treatment outcome. We previously demonstrated a feasible and highly efficient approach using ultrasound-aided microbubble (USMB) cavitation to enhance the permeability of the RWM. Here, we investigated the safety of USMB exposure and the association between temporal changes in RWM permeability and ultrastructure. Experimental guinea pigs were divided into two treatment groups: a control group receiving round window soaking (RWS) with MBs and treatment (USM) groups undergoing 3 (USM-3) or 5 (USM-5) consecutive USMB exposures (1 min/exposure) at an acoustic intensity of 3 W/cm2 and 1 MHz frequency. The trans-RWM delivery efficiency of biotin-fluorescein isothiocyanate conjugates, used as permeability tracers, revealed a greater than 7-fold higher delivery efficiency for the USM groups immediately after 3 or 5 exposures than for the RWS group. After 24 h, the delivery efficiency was 2.4-fold higher for the USM-3 group but was 6.6-fold higher for the USM-5 group (and 3.7-fold higher after 48 h), when compared to the RWS group. Scanning electron microscopy images of the RWM ultrastructure revealed USMB-induced sonoporation effects that could include the formation of heterogeneous pore-like openings with perforation diameters from 100 nm to several micrometers, disruption of the continuity of the outer epithelial surface layer, and loss of microvilli. These ultrastructural features were associated with differential permeability changes that depended on the USMB exposure course. Fourteen days after treatment, the pore-like openings had significantly decreased in number and the epithelial defects were healed either by cell expansion or by repair by newly migrated epithelial cells. The auditory brainstem response recordings of the animals following the 5-exposure USMB treatment indicated no deterioration in the hearing thresholds at a 2-month follow-up and no significant hair cell damage or apoptosis, based on scanning electron microscopy, surface preparations, and TUNEL assays. USMBs therefore appear to be safe and effective for inner ear drug delivery. The mechanism of enhanced permeability may involve a disruption of the continuity of the outer RWM epithelial layer, which controls transmembrane transport of various substances.
Collapse
Affiliation(s)
- Yi-Chun Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Chien Chen
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hang-Kang Chen
- Teaching and Research Section, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Yuan-Yung Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yin Kuo
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hao Wang
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Lien Hung
- Teaching and Research Section, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Hung Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Taichung Armed Forces General Hospital, Taichung, Taiwan
| |
Collapse
|
50
|
Duan L, Yang L, Jin J, Yang F, Liu D, Hu K, Wang Q, Yue Y, Gu N. Micro/nano-bubble-assisted ultrasound to enhance the EPR effect and potential theranostic applications. Theranostics 2020; 10:462-483. [PMID: 31903132 PMCID: PMC6929974 DOI: 10.7150/thno.37593] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/11/2019] [Indexed: 12/23/2022] Open
Abstract
Drug delivery for tumor theranostics involves the extensive use of the enhanced permeability and retention (EPR) effect. Previously, various types of nanomedicines have been demonstrated to accumulate in solid tumors via the EPR effect. However, EPR is a highly variable phenomenon because of tumor heterogeneity, resulting in low drug delivery efficacy in clinical trials. Because ultrasonication using micro/nanobubbles as contrast agents can disrupt blood vessels and enhance the specific delivery of drugs, it is an effective approach to improve the EPR effect for the passive targeting of tumors. In this review, the basic thermal effect, acoustic streaming, and cavitation mechanisms of ultrasound, which are characteristics that can be utilized to enhance the EPR effect, are briefly introduced. Second, micro/nanobubble-enhanced ultrasound imaging is discussed to understand the validity and variability of the EPR effect. Third, because the tumor microenvironment is complicated owing to elevated interstitial fluid pressure and the deregulated extracellular matrix components, which may be unfavorable for the EPR effect, few new trends in smart bubble drug delivery systems, which may improve the accuracy of EPR-mediated passive drug targeting, are summarized. Finally, the challenging and major concerns that should be considered in the next generation of micro/nanobubble-contrast-enhanced ultrasound theranostics for EPR-mediated passive drug targeting are also discussed.
Collapse
Affiliation(s)
- Lei Duan
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Li Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Juan Jin
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Dong Liu
- West Anhui University, Lu'an, P.R. China
- Anhui Engineering Laboratory for Conservation and Sustainable Utilization of Traditional Chinese Medicine Resources, P. R. China
| | - Ke Hu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qinxin Wang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuanbin Yue
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ning Gu
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|