1
|
Lin BZ, Fan AC, Wang Y, Lowerison MR, Dong Z, You Q, Sekaran NVC, Llano D, Borden M, Song P. Combined Nanodrops Imaging and Ultrasound Localization Microscopy for Detecting Intracerebral Hemorrhage. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:707-714. [PMID: 39837748 DOI: 10.1016/j.ultrasmedbio.2025.01.002] [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: 08/26/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Advanced imaging methods are crucial for understanding stroke mechanisms and discovering effective treatments to reduce bleeding and enhance recovery. In pre-clinical in vivo stroke imaging, MRI, CT and optical imaging are commonly used to evaluate stroke outcomes in rodent models. However, MRI and CT have limited spatial resolution for rodent brains, and optical imaging is hindered by limited imaging depth of penetration. Here we introduce a novel contrast-enhanced ultrasound imaging method to overcome these challenges and characterize intracerebral hemorrhage with unique insights. METHODS We combined microbubble-based ultrasound localization microscopy (ULM) and nanodrop (ND)-based vessel leakage imaging to achieve simultaneous microvascular imaging and hemorrhage detection. ULM maps brain-wide cerebral vasculature with high spatial resolution and identifies microvascular impairments around hemorrhagic areas. NDs are sub-micron liquid-core particles that can extravasate due to blood-brain barrier breakdown, serving as positive contrast agents to detect hemorrhage sites. RESULTS Our findings demonstrate that NDs could effectively accumulate in the hemorrhagic site and reveal the location of the bleeding areas upon activation by focused ultrasound beams. ULM further reveals the microvascular damage manifested in the form of reduced vascularity and decreased blood flow velocity across areas affected by the hemorrhagic stroke. CONCLUSION The results demonstrate that sequential ULM combined with ND imaging is a useful imaging tool for basic in vivo research in stroke with rodent models where brain-wide detection of active bleeding and microvascular impairment are essential.
Collapse
Affiliation(s)
- Bing-Ze Lin
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Yike Wang
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Matthew R Lowerison
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Zhijie Dong
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Qi You
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Nathiya Vaithiyalingam Chandra Sekaran
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Daniel Llano
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Mark Borden
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Pengfei Song
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA; Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
2
|
Hulme J. Harnessing Ultrasonic Technologies to Treat Staphylococcus Aureus Skin Infections. Molecules 2025; 30:512. [PMID: 39942617 PMCID: PMC11819699 DOI: 10.3390/molecules30030512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The rise of antibiotic-resistant Staphylococcus aureus strains, particularly MRSA, complicates the management of skin and soft tissue infections. This review highlights ultrasonic methodologies as adjunctive therapies to combat S. aureus-driven skin infections and prevent progression to biofilm formation and chronic wounds. Low- and high-frequency ultrasound (LFU and HFU) demonstrate potential in disrupting biofilms, enhancing drug delivery, and promoting tissue repair through cavitation and microbubble activity. These approaches integrate ultrasonic frequencies with microbubbles and therapeutics, such as antibiotics and affimers, to minimize resistance and improve healing. Tailoring the bioeffects of ultrasound on skin structures through localized delivery technologies, including microneedle patches and piezoelectric systems, presents promising solutions for early intervention in skin and soft structure infections (SSSIs).
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
3
|
Yildiz YO, Ruan JL, Gray MD, Bau L, Browning RJ, Mannaris C, Kiltie AE, Vojnovic B, Stride E. Combined drug delivery and treatment monitoring using a single high frequency ultrasound system. Int J Hyperthermia 2024; 41:2430330. [PMID: 39592132 DOI: 10.1080/02656736.2024.2430330] [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: 08/29/2024] [Revised: 10/19/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Ultrasound-mediated drug delivery is typically performed using transducers with center frequencies ≤ 1 MHz to promote acoustic cavitation. Such frequencies are not commonly used for diagnostic ultrasound due to limited spatial resolution. Therefore, delivery and monitoring of therapeutic ultrasound typically requires two transducers to enable both treatment and imaging. This study investigates the feasibility of using a single commercial ultrasound imaging transducer operating at 5 MHz for both drug delivery and real-time imaging. We compared a single-transducer system (STS) at 5 MHz with a conventional dual-transducer system (DTS) using a 1.1 MHz therapeutic transducer and an imaging probe. in vitro experiments demonstrated that the STS could achieve comparable extravasation depth and area as the DTS, with higher drug deposition observed at 5 MHz. Additionally, extravasation patterns were influenced by peak negative pressure (PNP) and duty cycle, with the narrower beam width at 5 MHz offering potential advantages for targeted drug delivery. in vivo experiments in a murine bladder cancer model confirmed the efficacy of the STS for real-time imaging and drug delivery, with cavitation dose correlating with drug deposition. The results suggest that a single-transducer approach may enhance the precision and efficiency of ultrasound-mediated drug delivery, potentially reducing system complexity and cost.
Collapse
Affiliation(s)
- Yesna O Yildiz
- Department of Oncology, University of Oxford, Oxford, UK
| | - Jia-Ling Ruan
- Department of Oncology, University of Oxford, Oxford, UK
| | - Michael D Gray
- Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Luca Bau
- Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | | | - Christophoros Mannaris
- Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Anne E Kiltie
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Eleanor Stride
- Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Wu Q, Choi V, Bau L, Carugo D, Evans ND, Stride E. Investigation of Ultrasound Mediated Extravasation of a Model Drug by Perfluorobutane Nanodroplets. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1573-1584. [PMID: 39060156 DOI: 10.1016/j.ultrasmedbio.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/14/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024]
Abstract
OBJECTIVE Perfluorocarbon nanodroplets (NDs) have been widely investigated as both diagnostic and therapeutic agents. There remains, however, a challenge in generating NDs that do not vaporize spontaneously but can be activated at ultrasound pressures that do not produce unwanted bioeffects. In previous work, it has been shown that phospholipid-coated perfluorobutane (PFB) NDs can potentially overcome this challenge. The aim of this study was to investigate whether these NDs can promote drug delivery. METHODS A combination of high-speed optical imaging and passive cavitation detection was used to study the acoustic properties of the PFB-NDs in a tissue mimicking phantom. PFB-NDs were exposed to ultrasound at frequencies from 0.5 to 1.5 MHz and peak negative pressures from 0.5 to 3.5 MPa. In addition, the penetration depth of two model drugs (Nile Red and 200 nm diameter fluorescent polymer spheres) into the phantom was measured. RESULTS PFB NDs were found to be stable in aqueous suspension at both 4°C and 37°C; their size remaining unchanged at 215 ± 11 nm over 24 h. Penetration of both model drugs in the phantom was found to increase with increasing ultrasound peak negative pressure and decreasing frequency and was found to be positively correlated with the energy of acoustic emissions. Extravasation depths >1 mm were observed at 0.5 MHz with pressures <1 MPa. CONCLUSION The results of the study thus suggest that PFB NDs can be used both as drug carriers and as nuclei for cavitation to enhance drug delivery without the need for high intensity ultrasound.
Collapse
Affiliation(s)
- Qiang Wu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Victor Choi
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Dario Carugo
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Nicholas D Evans
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research Group, University of Southampton, Southampton, UK; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK; Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Fuenteslópez CV, Gray M, Bahcevanci S, Martin A, Smith CAB, Coussios C, Cui Z, Ye H, Patrulea V. Mesenchymal stem cell cryopreservation with cavitation-mediated trehalose treatment. COMMUNICATIONS ENGINEERING 2024; 3:129. [PMID: 39251849 PMCID: PMC11385975 DOI: 10.1038/s44172-024-00265-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Dimethylsulfoxide (DMSO) has conventionally been used for cell cryopreservation both in research and in clinical applications, but has long-term cytotoxic effects. Trehalose, a natural disaccharide, has been proposed as a non-toxic cryoprotectant. However, the lack of specific cell membrane transporter receptors inhibits transmembrane transport and severely limits its cryoprotective capability. This research presents a method to successfully deliver trehalose into mesenchymal stem cells (MSCs) using ultrasound in the presence of microbubbles. The optimised trehalose concentration was shown to be able to not only preserve membrane integrity and cell viability but also the multipotency of MSCs, which are essential for stem cell therapy. Confocal imaging revealed that rhodamine-labelled trehalose was transported into cells rather than simply attached to the membrane. Additionally, the membranes were successfully preserved in lyophilised cells. This study demonstrates that ultrasonication with microbubbles facilitated trehalose delivery, offering promising cryoprotective capability without the cytotoxicity associated with DMSO-based methods.
Collapse
Affiliation(s)
- Carla V Fuenteslópez
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Simge Bahcevanci
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Alexander Martin
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Cameron A B Smith
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| | - Viorica Patrulea
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
6
|
Liu B, Du F, Feng Z, Xiang X, Guo R, Ma L, Zhu B, Qiu L. Ultrasound-augmented cancer immunotherapy. J Mater Chem B 2024; 12:3636-3658. [PMID: 38529593 DOI: 10.1039/d3tb02705h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Cancer is a growing worldwide health problem with the most broadly studied treatments, in which immunotherapy has made notable advancements in recent years. However, innumerable patients have presented a poor response to immunotherapy and simultaneously experienced immune-related adverse events, with failed therapeutic results and increased mortality rates. Consequently, it is crucial to develop alternate tactics to boost therapeutic effects without producing negative side effects. Ultrasound is considered to possess significant therapeutic potential in the antitumor field because of its inherent characteristics, including cavitation, pyrolysis, and sonoporation. Herein, this timely review presents the comprehensive and systematic research progress of ultrasound-enhanced cancer immunotherapy, focusing on the various ultrasound-related mechanisms and strategies. Moreover, this review summarizes the design and application of current sonosensitizers based on sonodynamic therapy, with an attempt to provide guidance on new directions for future cancer therapy.
Collapse
Affiliation(s)
- Bingjie Liu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Fangxue Du
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ziyan Feng
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xi Xiang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruiqian Guo
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bihui Zhu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Li Qiu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Park D, Lee SJ, Park JW. Aptamer-Based Smart Targeting and Spatial Trigger-Response Drug-Delivery Systems for Anticancer Therapy. Biomedicines 2024; 12:187. [PMID: 38255292 PMCID: PMC10813750 DOI: 10.3390/biomedicines12010187] [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: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
In recent years, the field of drug delivery has witnessed remarkable progress, driven by the quest for more effective and precise therapeutic interventions. Among the myriad strategies employed, the integration of aptamers as targeting moieties and stimuli-responsive systems has emerged as a promising avenue, particularly in the context of anticancer therapy. This review explores cutting-edge advancements in targeted drug-delivery systems, focusing on the integration of aptamers and stimuli-responsive platforms for enhanced spatial anticancer therapy. In the aptamer-based drug-delivery systems, we delve into the versatile applications of aptamers, examining their conjugation with gold, silica, and carbon materials. The synergistic interplay between aptamers and these materials is discussed, emphasizing their potential in achieving precise and targeted drug delivery. Additionally, we explore stimuli-responsive drug-delivery systems with an emphasis on spatial anticancer therapy. Tumor microenvironment-responsive nanoparticles are elucidated, and their capacity to exploit the dynamic conditions within cancerous tissues for controlled drug release is detailed. External stimuli-responsive strategies, including ultrasound-mediated, photo-responsive, and magnetic-guided drug-delivery systems, are examined for their role in achieving synergistic anticancer effects. This review integrates diverse approaches in the quest for precision medicine, showcasing the potential of aptamers and stimuli-responsive systems to revolutionize drug-delivery strategies for enhanced anticancer therapy.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|
8
|
Vlatakis S, Zhang W, Thomas S, Cressey P, Moldovan AC, Metzger H, Prentice P, Cochran S, Thanou M. Effect of Phase-Change Nanodroplets and Ultrasound on Blood-Brain Barrier Permeability In Vitro. Pharmaceutics 2023; 16:51. [PMID: 38258062 PMCID: PMC10818572 DOI: 10.3390/pharmaceutics16010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Phase-change nanodroplets (PCND;NDs) are emulsions with a perfluorocarbon (PFC) core that undergo acoustic vaporisation as a response to ultrasound (US). Nanodroplets change to microbubbles and cavitate while under the effect of US. This cavitation can apply forces on cell connections in biological barrier membranes, such as the blood-brain barrier (BBB), and trigger a transient and reversible increased permeability to molecules and matter. This study aims to present the preparation of lipid-based NDs and investigate their effects on the brain endothelial cell barrier in vitro. The NDs were prepared using the thin-film hydration method, followed by the PFC addition. They were characterised for size, cavitation (using a high-speed camera), and PFC encapsulation (using FTIR). The bEnd.3 (mouse brain endothelial) cells were seeded onto transwell inserts. Fluorescein with NDs and/or microbubbles were applied on the bEND3 cells and the effect of US on fluorescein permeability was measured. The Live/Dead assay was used to assess the BBB integrity after the treatments. Size and PFC content analysis indicated that the NDs were stable while stored. High-speed camera imaging confirmed that the NDs cavitate after US exposure of 0.12 MPa. The BBB cell model experiments revealed a 4-fold increase in cell membrane permeation after the combined application of US and NDs. The Live/Dead assay results indicated damage to the BBB membrane integrity, but this damage was less when compared to the one caused by microbubbles. This in vitro study shows that nanodroplets have the potential to cause BBB opening in a similar manner to microbubbles. Both cavitation agents caused damage on the endothelial cells. It appears that NDs cause less cell damage compared to microbubbles.
Collapse
Affiliation(s)
- Stavros Vlatakis
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Weiqi Zhang
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Sarah Thomas
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Paul Cressey
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| | - Alexandru Corneliu Moldovan
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Hilde Metzger
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Paul Prentice
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Sandy Cochran
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; (A.C.M.); (H.M.); (P.P.); (S.C.)
| | - Maya Thanou
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (S.V.); (W.Z.); (S.T.); (P.C.)
| |
Collapse
|
9
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
10
|
Zhang W, Metzger H, Vlatakis S, Claxton A, Carbajal MA, Fung LF, Mason J, Chan KLA, Pouliopoulos AN, Fleck RA, Prentice P, Thanou M. Characterising the chemical and physical properties of phase-change nanodroplets. ULTRASONICS SONOCHEMISTRY 2023; 97:106445. [PMID: 37257208 PMCID: PMC10241977 DOI: 10.1016/j.ultsonch.2023.106445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
Phase-change nanodroplets have attracted increasing interest in recent years as ultrasound theranostic nanoparticles. They are smaller compared to microbubbles and they may distribute better in tissues (e.g. in tumours). They are composed of a stabilising shell and a perfluorocarbon core. Nanodroplets can vaporise into echogenic microbubbles forming cavitation nuclei when exposed to ultrasound. Their perfluorocarbon core phase-change is responsible for the acoustic droplet vaporisation. However, methods to quantify the perfluorocarbon core in nanodroplets are lacking. This is an important feature that can help explain nanodroplet phase change characteristics. In this study, we fabricated nanodroplets using lipids shell and perfluorocarbons. To assess the amount of perfluorocarbon in the core we used two methods, 19F NMR and FTIR. To assess the cavitation after vaporisation we used an ultrasound transducer (1.1 MHz) and a high-speed camera. The 19F NMR based method showed that the fluorine signal correlated accurately with the perfluorocarbon concentration. Using this correlation, we were able to quantify the perfluorocarbon core of nanodroplets. This method was used to assess the content of the perfluorocarbon of the nanodroplets in solutions over time. It was found that perfluoropentane nanodroplets lost their content faster and at higher ratio compared to perfluorohexane nanodroplets. The high-speed imaging indicates that the nanodroplets generate cavitation comparable to that from commercial contrast agent microbubbles. Nanodroplet characterisation should include perfluorocarbon concentration assessment as critical information for their development.
Collapse
Affiliation(s)
- Weiqi Zhang
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Hilde Metzger
- School of Engineering, University of Glasgow, United Kingdom
| | - Stavros Vlatakis
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Amelia Claxton
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | | | - Leong Fan Fung
- Department of Surgical & Interventional Engineering, King's College London, United Kingdom
| | - James Mason
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - K L Andrew Chan
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | | | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, United Kingdom
| | - Paul Prentice
- School of Engineering, University of Glasgow, United Kingdom
| | - Maya Thanou
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom.
| |
Collapse
|
11
|
Vince J, Lewis A, Stride E. High-Speed Imaging of Microsphere Transport by Cavitation Activity in a Tissue-Mimicking Phantom. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1415-1421. [PMID: 36931999 DOI: 10.1016/j.ultrasmedbio.2023.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE Ultrasound-mediated cavitation has been harnessed to improve the delivery of various therapeutics, including the extravasation of small molecule drugs and nanoparticles (<1 µm) into soft tissue. This study investigated whether cavitation could also enhance the extravasation of larger (>10 µm) therapeutic particles, representative of radio- or chemo-embolic particles, in a tissue-mimicking phantom. METHODS High-speed (103-106 frames/s) optical imaging was used to observe the motion of glass microspheres with diameters of 15-32 or 105-107 µm in an agar phantom under exposure to high-intensity focused ultrasound (0.5 MHz) at a range of peak negative pressures (1.9-2.8 MPa) in the presence of SonoVue microbubbles. RESULTS In contrast to the microstreaming reported to be responsible for nanoparticle transport, the formation and translation of bubble clouds were found to be primarily responsible for the motion of glass microspheres. The bubble clouds were seen both to create channels in the phantom and to travel along them under the action of primary acoustic radiation force, either propelling or entraining microspheres with them. Collisions between microspheres were also seen to promote cloud formation and cavitation activity. CONCLUSION Ultrasound-mediated cavitation can promote the transport of solid microparticles in tissue-mimicking material. Further work is needed to understand the influence of tissue mechanical properties and ultrasound exposure parameters on the extent and uniformity of particle distribution that can be achieved.
Collapse
Affiliation(s)
- Jonathan Vince
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Andrew Lewis
- Alchemed Bioscience Consulting Ltd., Stable Cottage, Farnham, Surrey, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Alcaraz PE, Davidson SJ, Shreeve E, Meuschke R, Romanowski M, Witte RS, Porter TR, Matsunaga TO. Thermal and Acoustic Stabilization Of Volatile Phase-Change Contrast Agents Via Layer-By-Layer Assembly. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1058-1069. [PMID: 36797095 PMCID: PMC10050125 DOI: 10.1016/j.ultrasmedbio.2022.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 05/11/2023]
Abstract
OBJECTIVE Phase-change contrast agents (PCCAs) are perfluorocarbon nanodroplets (NDs) that have been widely studied for ultrasound imaging in vitro, pre-clinical studies, and most recently incorporated a variant of PCCAs, namely a microbubble-conjugated microdroplet emulsion, into the first clinical studies. Their properties also make them attractive candidates for a variety of diagnostic and therapeutic applications including drug-delivery, diagnosis and treatment of cancerous and inflammatory diseases, as well as tumor-growth tracking. However, control over the thermal and acoustic stability of PCCAs both in vivo and in vitro has remained a challenge for expanding the potential utility of these agents in novel clinical applications. As such, our objective was to determine the stabilizing effects of layer-by-layer assemblies and its effect on both thermal and acoustic stability. METHODS We utilized layer-by-layer (LBL) assemblies to coat the outer PCCA membrane and characterized layering by measuring zeta potential and particle size. Stability studies were conducted by; 1) incubating the LBL-PCCAs at atmospheric pressure at 37∘C and 45∘C followed by; 2) ultrasound-mediated activation at 7.24 MHz and peak-negative pressures ranging from 0.71 - 5.48 MPa to ascertain nanodroplet activation and resultant microbubble persistence. The thermal and acoustic properties of decafluorobutane gas-condensed nanodroplets (DFB-NDs) layered with 6 and 10 layers of charge-alternating biopolymers, (LBL6NDs and LBL10NDs) respectively, were studied and compared to non-layered DFB-NDs. Half-life determinations were conducted at both 37∘C and 45∘C with acoustic droplet vaporization (ADV) measurements occurring at 23∘C. DISCUSSION Successful application of up to 10 layers of alternating positive and negatively charged biopolymers onto the surface membrane of DFB-NDs was demonstrated. Two major claims were substantiated in this study; namely, (1) biopolymeric layering of DFB-NDs imparts a thermal stability up to an extent; and, (2) both LBL6NDs and LBL10NDs did not appear to alter particle acoustic vaporization thresholds, suggesting that the thermal stability of the particle may not necessarily be coupled with particle acoustic vaporization thresholds. CONCLUSION Results demonstrate that the layered PCCAs had higher thermal stability, where the half-lifes of the LBLxNDs are significantly increased after incubation at 37∘C and 45∘C. Furthermore, the acoustic vaporization profiles the DFB-NDs, LBL6NDs, and LBL10NDs show that there is no statistically significant difference between the acoustic vaporization energy required to initiate acoustic droplet vaporization.
Collapse
Affiliation(s)
- Pedro Enrique Alcaraz
- College of Optical Sciences, University of Arizona, 1630 E University Blvd., Tucson, AZ 85721 United States; Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Medical Imaging, University of Arizona, Tucson, AZ. 85719 United States
| | - Skylar J Davidson
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Evan Shreeve
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Rainee Meuschke
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Marek Romanowski
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Russell S Witte
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Medical Imaging, University of Arizona, Tucson, AZ. 85719 United States
| | - Thomas R Porter
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Terry O Matsunaga
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Medical Imaging, University of Arizona, Tucson, AZ. 85719 United States.
| |
Collapse
|
13
|
Edwards IA, De Carlo F, Sitta J, Varner W, Howard CM, Claudio PP. Enhancing Targeted Therapy in Breast Cancer by Ultrasound-Responsive Nanocarriers. Int J Mol Sci 2023; 24:ijms24065474. [PMID: 36982548 PMCID: PMC10053544 DOI: 10.3390/ijms24065474] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Currently, the response to cancer treatments is highly variable, and severe side effects and toxicity are experienced by patients receiving high doses of chemotherapy, such as those diagnosed with triple-negative breast cancer. The main goal of researchers and clinicians is to develop new effective treatments that will be able to specifically target and kill tumor cells by employing the minimum doses of drugs exerting a therapeutic effect. Despite the development of new formulations that overall can increase the drugs’ pharmacokinetics, and that are specifically designed to bind overexpressed molecules on cancer cells and achieve active targeting of the tumor, the desired clinical outcome has not been reached yet. In this review, we will discuss the current classification and standard of care for breast cancer, the application of nanomedicine, and ultrasound-responsive biocompatible carriers (micro/nanobubbles, liposomes, micelles, polymeric nanoparticles, and nanodroplets/nanoemulsions) employed in preclinical studies to target and enhance the delivery of drugs and genes to breast cancer.
Collapse
Affiliation(s)
- Isaiah A. Edwards
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Flavia De Carlo
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Juliana Sitta
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William Varner
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Candace M. Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Pier Paolo Claudio
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Correspondence:
| |
Collapse
|
14
|
Hu Y, Wei J, Shen Y, Chen S, Chen X. Barrier-breaking effects of ultrasonic cavitation for drug delivery and biomarker release. ULTRASONICS SONOCHEMISTRY 2023; 94:106346. [PMID: 36870921 PMCID: PMC10040969 DOI: 10.1016/j.ultsonch.2023.106346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 05/27/2023]
Abstract
Recently, emerging evidence has demonstrated that cavitation actually creates important bidirectional channels on biological barriers for both intratumoral drug delivery and extratumoral biomarker release. To promote the barrier-breaking effects of cavitation for both therapy and diagnosis, we first reviewed recent technical advances of ultrasound and its contrast agents (microbubbles, nanodroplets, and gas-stabilizing nanoparticles) and then reported the newly-revealed cavitation physical details. In particular, we summarized five types of cellular responses of cavitation in breaking the plasma membrane (membrane retraction, sonoporation, endocytosis/exocytosis, blebbing and apoptosis) and compared the vascular cavitation effects of three different types of ultrasound contrast agents in breaking the blood-tumor barrier and tumor microenvironment. Moreover, we highlighted the current achievements of the barrier-breaking effects of cavitation in mediating drug delivery and biomarker release. We emphasized that the precise induction of a specific cavitation effect for barrier-breaking was still challenged by the complex combination of multiple acoustic and non-acoustic cavitation parameters. Therefore, we provided the cutting-edge in-situ cavitation imaging and feedback control methods and suggested the development of an international cavitation quantification standard for the clinical guidance of cavitation-mediated barrier-breaking effects.
Collapse
Affiliation(s)
- Yaxin Hu
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Jianpeng Wei
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Yuanyuan Shen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Siping Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Xin Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China.
| |
Collapse
|
15
|
Armenia I, Cuestas Ayllón C, Torres Herrero B, Bussolari F, Alfranca G, Grazú V, Martínez de la Fuente J. Photonic and magnetic materials for on-demand local drug delivery. Adv Drug Deliv Rev 2022; 191:114584. [PMID: 36273514 DOI: 10.1016/j.addr.2022.114584] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 10/16/2022] [Indexed: 02/06/2023]
Abstract
Nanomedicine has been considered a promising tool for biomedical research and clinical practice in the 21st century because of the great impact nanomaterials could have on human health. The generation of new smart nanomaterials, which enable time- and space-controlled drug delivery, improve the limitations of conventional treatments, such as non-specific targeting, poor biodistribution and permeability. These smart nanomaterials can respond to internal biological stimuli (pH, enzyme expression and redox potential) and/or external stimuli (such as temperature, ultrasound, magnetic field and light) to further the precision of therapies. To this end, photonic and magnetic nanoparticles, such as gold, silver and iron oxide, have been used to increase sensitivity and responsiveness to external stimuli. In this review, we aim to report the main and most recent systems that involve photonic or magnetic nanomaterials for external stimulus-responsive drug release. The uniqueness of this review lies in highlighting the versatility of integrating these materials within different carriers. This leads to enhanced performance in terms of in vitro and in vivo efficacy, stability and toxicity. We also point out the current regulatory challenges for the translation of these systems from the bench to the bedside, as well as the yet unresolved matter regarding the standardization of these materials.
Collapse
Affiliation(s)
- Ilaria Armenia
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain.
| | - Carlos Cuestas Ayllón
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Beatriz Torres Herrero
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Francesca Bussolari
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Gabriel Alfranca
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Valeria Grazú
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain; Centro de Investigación Biomédica em Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Jesús Martínez de la Fuente
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain; Centro de Investigación Biomédica em Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
16
|
Sarwar U, Naeem M, Nurjis F, Karim S, Raza A. Ultrasound-mediated in vivo biodistribution of coumarin-labeled sorafenib-loaded liposome-based nanotheranostic system. Nanomedicine (Lond) 2022; 17:1909-1927. [PMID: 36695214 DOI: 10.2217/nnm-2022-0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aim: This study aimed to synthesize folate-conjugated sorafenib-loaded (FCSL) liposomes for theranostic application using ultrasound (US). Materials & methods: US parameter optimization, in vitro release, anticancer effect, in vivo biodistribution, optical imaging and biocompatibility of liposomes were studied. Results: With 84% in vitro release after 4 min of US exposure at 3 MHz (1.2 mechanical index), FCSL liposomes showed lower IC50 (8.70 μM) versus sorafenib (9.34 μM) against HepG2 cells. In vivo biodistribution of FCSL liposomes versus sorafenib after 9 mg/kg injection in the liver (8.63 vs 0.55) > intestine (8.45 vs 1.07) > stomach (5.62 vs 0.57) > kidney (5.46 vs 0.91) showed longer circulation time in plasma and can be tracked in mice. Conclusion: A threefold higher drug concentration in the liver in US-exposed mice makes this a successful nanotheranostic approach.
Collapse
Affiliation(s)
- Usama Sarwar
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers & Optronics College (NILOP-C), Pakistan Institute of Engineering & Applied Sciences, Nilore, Islamabad, 45650, Pakistan.,Department of Biotechnology, Medical Genetics Research Laboratory, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Naeem
- Department of Biotechnology, Medical Genetics Research Laboratory, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Farwa Nurjis
- NILOP Nanomedicine Research Laboratories, National Institute of Lasers & Optronics College (NILOP-C), Pakistan Institute of Engineering & Applied Sciences, Nilore, Islamabad, 45650, Pakistan
| | - Shafqat Karim
- Nano Materials Research Group, Pakistan Institute of Nuclear Science & Technology (PINSTECH), Nilore, Islamabad, 45650, Pakistan
| | - Abida Raza
- National Center of Industrial Biotechnology, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, 46000, Pakistan
| |
Collapse
|
17
|
Zhang W, Shi Y, Abd Shukor S, Vijayakumaran A, Vlatakis S, Wright M, Thanou M. Phase-shift nanodroplets as an emerging sonoresponsive nanomaterial for imaging and drug delivery applications. NANOSCALE 2022; 14:2943-2965. [PMID: 35166273 DOI: 10.1039/d1nr07882h] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanodroplets - emerging phase-changing sonoresponsive materials - have attracted substantial attention in biomedical applications for both tumour imaging and therapeutic purposes due to their unique response to ultrasound. As ultrasound is applied at different frequencies and powers, nanodroplets have been shown to cavitate by the process of acoustic droplet vapourisation (ADV), causing the development of mechanical forces which promote sonoporation through cellular membranes. This allows drugs to be delivered efficiently into deeper tissues where tumours are located. Recent reviews on nanodroplets are mostly focused on the mechanism of cavitation and their applications in biomedical fields. However, the chemistry of the nanodroplet components has not been discussed or reviewed yet. In this review, the commonly used materials and preparation methods of nanodroplets are summarised. More importantly, this review provides examples of variable chemistry components in nanodroplets which link them to their efficiency as ultrasound-multimodal imaging agents to image and monitor drug delivery. Finally, the drawbacks of current research, future development, and future direction of nanodroplets are discussed.
Collapse
Affiliation(s)
- Weiqi Zhang
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | - Yuhong Shi
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | | | | | - Stavros Vlatakis
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | - Michael Wright
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | - Maya Thanou
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| |
Collapse
|
18
|
Sabuncu S, Yildirim A. Gas-stabilizing nanoparticles for ultrasound imaging and therapy of cancer. NANO CONVERGENCE 2021; 8:39. [PMID: 34851458 PMCID: PMC8636532 DOI: 10.1186/s40580-021-00287-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/05/2021] [Indexed: 05/06/2023]
Abstract
The use of ultrasound in the clinic has been long established for cancer detection and image-guided tissue biopsies. In addition, ultrasound-based methods have been widely explored to develop more effective cancer therapies such as localized drug delivery, sonodynamic therapy, and focused ultrasound surgery. Stabilized fluorocarbon microbubbles have been in use as contrast agents for ultrasound imaging in the clinic for several decades. It is also known that microbubble cavitation could generate thermal, mechanical, and chemical effects in the tissue to improve ultrasound-based therapies. However, the large size, poor stability, and short-term cavitation activity of microbubbles limit their applications in cancer imaging and therapy. This review will focus on an alternative type of ultrasound responsive material; gas-stabilizing nanoparticles, which can address the limitations of microbubbles with their nanoscale size, robustness, and high cavitation activity. This review will be of interest to researchers who wish to explore new agents to develop improved methods for molecular ultrasound imaging and therapy of cancer.
Collapse
Affiliation(s)
- Sinan Sabuncu
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Adem Yildirim
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health & Science University, Portland, OR, 97201, USA.
| |
Collapse
|
19
|
Lea-Banks H, Hynynen K. Sub-millimetre precision of drug delivery in the brain from ultrasound-triggered nanodroplets. J Control Release 2021; 338:731-741. [PMID: 34530050 DOI: 10.1016/j.jconrel.2021.09.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/17/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022]
Abstract
Drug-loaded nanoscale cavitation agents, called nanodroplets, are an attractive solution to enhance and localize drug delivery, offering increased stability and prolonged half-life in circulation compared to microbubbles. However, the spatial precision with which drug can be released and delivered into brain tissue from such agents has not been directly mapped. Decafluorobutane lipid-shell droplets (206 +/- 6 nm) were loaded with a fluorescent blood-brain barrier (BBB)-penetrating dye (Nile Blue) and vaporized with ultrasound (1.66 MHz, 10 ms pulse length, 1 Hz pulse repetition frequency), generating transient echogenic microbubbles and delivering the encapsulated dye. The distribution and intensity of released fluorophore was mapped in a tissue-mimicking phantom, and in the brain of rats (Sprague Dawley, N = 4, n = 16). The release and distribution of dye was found to be pressure-dependent (0.2-3.5 MPa) and to occur only above the vaporization threshold of the nanodroplets (1.5 +/- 0.25 MPa in vitro, 2.4 +/- 0.05 MPa in vivo). Dye delivery was achieved with sub-millimetre spatial precision, covering an area of 0.4 to 1.5 mm in diameter, determined by the sonication pressure. The distribution and intensity of dye released at depth in the brain followed the axial pressure profile of the ultrasound beam. Nile Blue (354 Da, LogP 2.7) was compared to Nile Red (318 Da, LogP 3.8) and Quantum Dots (CdSe/ZnS, 5 nm diameter) to visualize the role of molecule size and lipophilicity in crossing the intact BBB following triggered release. Acoustic emissions were shown to predict the successful delivery of the BBB-penetrating dye and the extent of the distribution, demonstrating the theranostic capabilities of nanoscale droplets to precisely localize drug delivery in the brain.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
20
|
Vidallon MLP, Giles LW, Pottage MJ, Butler CSG, Crawford SA, Bishop AI, Tabor RF, de Campo L, Teo BM. Tracking the heat-triggered phase change of polydopamine-shelled, perfluorocarbon emulsion droplets into microbubbles using neutron scattering. J Colloid Interface Sci 2021; 607:836-847. [PMID: 34536938 DOI: 10.1016/j.jcis.2021.08.162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/15/2021] [Accepted: 08/24/2021] [Indexed: 01/12/2023]
Abstract
Perfluorocarbon emulsion droplets are hybrid colloidal materials with vast applications, ranging from imaging to drug delivery, due to their controllable phase transition into microbubbles via heat application or acoustic droplet vapourisation. The current work highlights the application of small- and ultra-small-angle neutron scattering (SANS and USANS), in combination with contrast variation techniques, in observing the in situ phase transition of polydopamine-shelled, perfluorocarbon (PDA/PFC) emulsion droplets with controlled polydispersity into microbubbles upon heating. We correlate these measurements with optical and transmission electron microscopy imaging, dynamic light scattering, and thermogravimetric analysis to characterise these emulsions, and observe their phase transition into microbubbles. Results show that the phase transition of PDA/PFC droplets with perfluorohexane (PFH), perfluoropentane (PFP), and PFH-PFP mixtures occur at temperatures that are around 30-40 °C higher than the boiling points of pure liquid PFCs, and this is influenced by the specific PFC compositions (perfluorohexane, perfluoropentane, and mixtures of these PFCs). Analysis and model fitting of neutron scattering data allowed us to monitor droplet size distributions at different temperatures, giving valuable insights into the transformation of these polydisperse, emulsion droplet systems.
Collapse
Affiliation(s)
| | - Luke W Giles
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - Matthew J Pottage
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - Calum S G Butler
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - Simon A Crawford
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, VIC 3800, Australia
| | - Alexis I Bishop
- School of Physics and Astronomy, Monash University, Clayton, VIC 3800, Australia
| | - Rico F Tabor
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia.
| | - Liliana de Campo
- Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Rd, Lucas Heights, NSW 2234, Australia.
| | - Boon Mian Teo
- School of Chemistry, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
21
|
Krafft MP, Riess JG. Therapeutic oxygen delivery by perfluorocarbon-based colloids. Adv Colloid Interface Sci 2021; 294:102407. [PMID: 34120037 DOI: 10.1016/j.cis.2021.102407] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
After the protocol-related indecisive clinical trial of Oxygent, a perfluorooctylbromide/phospholipid nanoemulsion, in cardiac surgery, that often unduly assigned the observed untoward effects to the product, the development of perfluorocarbon (PFC)-based O2 nanoemulsions ("blood substitutes") has come to a low. Yet, significant further demonstrations of PFC O2-delivery efficacy have continuously been reported, such as relief of hypoxia after myocardial infarction or stroke; protection of vital organs during surgery; potentiation of O2-dependent cancer therapies, including radio-, photodynamic-, chemo- and immunotherapies; regeneration of damaged nerve, bone or cartilage; preservation of organ grafts destined for transplantation; and control of gas supply in tissue engineering and biotechnological productions. PFC colloids capable of augmenting O2 delivery include primarily injectable PFC nanoemulsions, microbubbles and phase-shift nanoemulsions. Careful selection of PFC and other colloid components is critical. The basics of O2 delivery by PFC nanoemulsions will be briefly reminded. Improved knowledge of O2 delivery mechanisms has been acquired. Advanced, size-adjustable O2-delivering nanoemulsions have been designed that have extended room-temperature shelf-stability. Alternate O2 delivery options are being investigated that rely on injectable PFC-stabilized microbubbles or phase-shift PFC nanoemulsions. The latter combine prolonged circulation in the vasculature, capacity for penetrating tumor tissues, and acute responsiveness to ultrasound and other external stimuli. Progress in microbubble and phase-shift emulsion engineering, control of phase-shift activation (vaporization), understanding and control of bubble/ultrasound/tissue interactions is discussed. Control of the phase-shift event and of microbubble size require utmost attention. Further PFC-based colloidal systems, including polymeric micelles, PFC-loaded organic or inorganic nanoparticles and scaffolds, have been devised that also carry substantial amounts of O2. Local, on-demand O2 delivery can be triggered by external stimuli, including focused ultrasound irradiation or tumor microenvironment. PFC colloid functionalization and targeting can help adjust their properties for specific indications, augment their efficacy, improve safety profiles, and expand the range of their indications. Many new medical and biotechnological applications involving fluorinated colloids are being assessed, including in the clinic. Further uses of PFC-based colloidal nanotherapeutics will be briefly mentioned that concern contrast diagnostic imaging, including molecular imaging and immune cell tracking; controlled delivery of therapeutic energy, as for noninvasive surgical ablation and sonothrombolysis; and delivery of drugs and genes, including across the blood-brain barrier. Even when the fluorinated colloids investigated are designed for other purposes than O2 supply, they will inevitably also carry and deliver a certain amount of O2, and may thus be considered for O2 delivery or co-delivery applications. Conversely, O2-carrying PFC nanoemulsions possess by nature a unique aptitude for 19F MR imaging, and hence, cell tracking, while PFC-stabilized microbubbles are ideal resonators for ultrasound contrast imaging and can undergo precise manipulation and on-demand destruction by ultrasound waves, thereby opening multiple theranostic opportunities.
Collapse
Affiliation(s)
- Marie Pierre Krafft
- University of Strasbourg, Institut Charles Sadron (CNRS), 23 rue du Loess, 67034 Strasbourg, France.
| | - Jean G Riess
- Harangoutte Institute, 68160 Ste Croix-aux-Mines, France
| |
Collapse
|
22
|
Wu Q, Mannaris C, May JP, Bau L, Polydorou A, Ferri S, Carugo D, Evans ND, Stride E. Investigation of the Acoustic Vaporization Threshold of Lipid-Coated Perfluorobutane Nanodroplets Using Both High-Speed Optical Imaging and Acoustic Methods. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1826-1843. [PMID: 33820668 DOI: 10.1016/j.ultrasmedbio.2021.02.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
A combination of ultrahigh-speed optical imaging (5 × 106 frames/s), B-mode ultrasound and passive cavitation detection was used to study the vaporization process and determine both the acoustic droplet vaporization (ADV) and inertial cavitation (IC) thresholds of phospholipid-coated perfluorobutane nanodroplets (PFB NDs, diameter = 237 ± 16 nm). PFB NDs have not previously been studied with ultrahigh-speed imaging and were observed to form individual microbubbles (1-10 μm) within two to three cycles and subsequently larger bubble clusters (10-50 μm). The ADV and IC thresholds did not statistically significantly differ and decreased with increasing pulse length (20-20,000 cycles), pulse repetition frequency (1-100 Hz), concentration (108-1010 NDs/mL), temperature (20°C-45°C) and decreasing frequency (1.5-0.5 MHz). Overall, the results indicate that at frequencies of 0.5, 1.0 and 1.5 MHz, PFB NDs can be vaporized at moderate peak negative pressures (<2.0 MPa), pulse lengths and pulse repetition frequencies. This finding is encouraging for the use of PFB NDs as cavitation agents, as these conditions are comparable to those required to achieve therapeutic effects with microbubbles, unlike those reported for higher-boiling-point NDs. The differences between the optically and acoustically determined ADV thresholds, however, suggest that application-specific thresholds should be defined according to the biological/therapeutic effect of interest.
Collapse
Affiliation(s)
- Qiang Wu
- Institute of Biomedical Engineering, Department of Engineering Science, Old Road Campus Research Building, University of Oxford, Oxford, United Kingdom
| | - Christophoros Mannaris
- Institute of Biomedical Engineering, Department of Engineering Science, Old Road Campus Research Building, University of Oxford, Oxford, United Kingdom
| | - Jonathan P May
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom; Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Science, Old Road Campus Research Building, University of Oxford, Oxford, United Kingdom
| | - Anastasia Polydorou
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom; Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Sara Ferri
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom; Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Dario Carugo
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom; Department of Pharmaceutics, UCL School of Pharmacy, University College London, London, UK
| | - Nicholas D Evans
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom; Bone and Joint Research Group, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, Old Road Campus Research Building, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
23
|
Ruan JL, Browning RJ, Yildiz YO, Bau L, Kamila S, Gray MD, Folkes L, Hampson A, McHale AP, Callan JF, Vojnovic B, Kiltie AE, Stride E. Evaluation of Loading Strategies to Improve Tumor Uptake of Gemcitabine in a Murine Orthotopic Bladder Cancer Model Using Ultrasound and Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1596-1615. [PMID: 33707089 DOI: 10.1016/j.ultrasmedbio.2021.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
In this study we compared three different microbubble-based approaches to the delivery of a widely used chemotherapy drug, gemcitabine: (i) co-administration of gemcitabine and microbubbles (Gem+MB); (ii) conjugates of microbubbles and gemcitabine-loaded liposomes (GemlipoMB); and (iii) microbubbles with gemcitabine directly bound to their surfaces (GembioMB). Both in vitro and in vivo investigations were carried out, respectively, in the RT112 bladder cancer cell line and in a murine orthotopic muscle-invasive bladder cancer model. The in vitro (in vivo) ultrasound exposure conditions were a 1 (1.1) MHz centre frequency, 0.07 (1.0) MPa peak negative pressure, 3000 (20,000) cycles and 100 (0.5) Hz pulse repetition frequency. Ultrasound exposure produced no significant increase in drug uptake either in vitro or in vivo compared with the drug-only control for co-administered gemcitabine and microbubbles. In vivo, GemlipoMB prolonged the plasma circulation time of gemcitabine, but only GembioMB produced a statistically significant increase in cleaved caspase 3 expression in the tumor, indicative of gemcitabine-induced apoptosis.
Collapse
Affiliation(s)
- Jia-Ling Ruan
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Richard J Browning
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Yesna O Yildiz
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Sukanta Kamila
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Michael D Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Lisa Folkes
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Alix Hampson
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anthony P McHale
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - John F Callan
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Borivoj Vojnovic
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anne E Kiltie
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
24
|
Ferri S, Wu Q, De Grazia A, Polydorou A, May JP, Stride E, Evans ND, Carugo D. Tailoring the size of ultrasound responsive lipid-shelled nanodroplets by varying production parameters and environmental conditions. ULTRASONICS SONOCHEMISTRY 2021; 73:105482. [PMID: 33588208 PMCID: PMC7901031 DOI: 10.1016/j.ultsonch.2021.105482] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/08/2021] [Accepted: 01/27/2021] [Indexed: 05/06/2023]
Abstract
Liquid perfluorocarbon nanodroplets (NDs) are an attractive alternative to microbubbles (MBs) for ultrasound-mediated therapeutic and diagnostic applications. ND size and size distribution have a strong influence on their behaviour in vivo, including extravasation efficiency, circulation time, and response to ultrasound stimulation. Thus, it is desirable to identify ways to tailor the ND size and size distribution during manufacturing. In this study phospholipid-coated NDs, comprising a perfluoro-n-pentane (PFP) core stabilised by a DSPC/PEG40s (1,2-distearoyl-sn-glycero-3-phosphocholine and polyoxyethylene(40)stearate, 9:1 molar ratio) shell, were produced in phosphate-buffered saline (PBS) by sonication. The effect of the following production-related parameters on ND size was investigated: PFP concentration, power and duration of sonication, and incorporation of a lipophilic fluorescent dye. ND stability was also assessed at both 4 °C and 37 °C. When a sonication pulse of 6 s and 15% duty cycle was employed, increasing the volumetric concentration of PFP from 5% to 15% v/v in PBS resulted in an increase in ND diameter from 215.8 ± 16.8 nm to 408.9 ± 171.2 nm. An increase in the intensity of sonication from 48 to 72 W (with 10% PFP v/v in PBS) led to a decrease in ND size from 354.6 ± 127.2 nm to 315.0 ± 100.5 nm. Increasing the sonication time from 20 s to 40 s (using a pulsed sonication with 30% duty cycle) did not result in a significant change in ND size (in the range 278-314 nm); however, when it was increased to 60 s, the average ND diameter reduced to 249.7 ± 9.7 nm, which also presented a significantly lower standard deviation compared to the other experimental conditions investigated (i.e., 9.7 nm vs. > 49.4 nm). The addition of the fluorescent dye DiI at different molar ratios did not affect the ND size distribution. NDs were stable at 4 °C for up to 6 days and at 37 °C for up to 110 min; however, some evidence of ND-to-MB phase transition was observed after 40 min at 37 °C. Finally, phase transition of NDs into MBs was demonstrated using a tissue-mimicking flow phantom under therapeutic ultrasound exposure conditions (ultrasound frequency: 0.5 MHz, acoustic pressure: 2-4 MPa, and pulse repetition frequency: 100 Hz).
Collapse
Affiliation(s)
- Sara Ferri
- Faculty of Engineering and Physical Sciences, Department of Mechanical Engineering, University of Southampton, UK; Centre for Human Development, Stem Cells and Regeneration, Bioengineering Sciences, Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, UK
| | - Qiang Wu
- Department of Engineering Science, University of Oxford, UK
| | - Antonio De Grazia
- Faculty of Engineering and Physical Sciences, Department of Mechanical Engineering, University of Southampton, UK
| | - Anastasia Polydorou
- Faculty of Engineering and Physical Sciences, Department of Mechanical Engineering, University of Southampton, UK; Centre for Human Development, Stem Cells and Regeneration, Bioengineering Sciences, Faculty of Medicine, University of Southampton, UK
| | - Jonathan P May
- Faculty of Engineering and Physical Sciences, Department of Mechanical Engineering, University of Southampton, UK; Centre for Human Development, Stem Cells and Regeneration, Bioengineering Sciences, Faculty of Medicine, University of Southampton, UK
| | - Eleanor Stride
- Department of Engineering Science, University of Oxford, UK
| | - Nicholas D Evans
- Faculty of Engineering and Physical Sciences, Department of Mechanical Engineering, University of Southampton, UK; Centre for Human Development, Stem Cells and Regeneration, Bioengineering Sciences, Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, UK
| | - Dario Carugo
- Department of Pharmaceutics, School of Pharmacy, University College London (UCL), UK.
| |
Collapse
|
25
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
26
|
Vidallon MLP, Giles LW, Crawford S, Bishop AI, Tabor RF, de Campo L, Teo BM. Exploring the transition of polydopamine-shelled perfluorohexane emulsion droplets into microbubbles using small- and ultra-small-angle neutron scattering. Phys Chem Chem Phys 2021; 23:9843-9850. [PMID: 33908524 DOI: 10.1039/d1cp01146d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Perfluorocarbon emulsion droplets are interesting colloidal systems with applications, ranging from diagnostics and theranostics to drug delivery, due to their controllable phase transition into microbubbles via heat application or acoustic droplet vapourisation. This work highlights the application of small- and ultra-small-angle neutron scattering (SANS and USANS, respectively), in combination with contrast variation techniques, in observing the in situ phase transition of polydopamine-stabilised perfluorohexane (PDA/PFH) emulsion droplets into microbubbles during heating. Results show peak USANS intensities at temperatures around 90 °C, which indicates that the phase transition of PDA/PFH emulsion droplets occurs at significantly higher temperatures than the bulk boiling point of pure liquid PFH (56 °C). Analysis and model fitting of the SANS and USANS data allowed us to estimate droplet sizes and interfacial properties at different temperatures (20 °C, 90 °C, and 20 °C after cooling), giving valuable insights about the transformation of these polydisperse emulsion droplet systems.
Collapse
|
27
|
Gray MD, Elbes D, Paverd C, Lyka E, Coviello CM, Cleveland RO, Coussios CC. Dual-Array Passive Acoustic Mapping for Cavitation Imaging With Enhanced 2-D Resolution. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:647-663. [PMID: 32845836 DOI: 10.1109/tuffc.2020.3019573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Passive acoustic mapping (PAM) techniques have been developed for the purposes of detecting, localizing, and quantifying cavitation activity during therapeutic ultrasound procedures. Implementation with conventional diagnostic ultrasound arrays has allowed planar mapping of bubble acoustic emissions to be overlaid with B-mode anatomical images, with a variety of beamforming approaches providing enhanced resolution at the cost of extended computation times. However, no passive signal processing techniques implemented to date have overcome the fundamental physical limitation of the conventional diagnostic array aperture that results in point spread functions with axial/lateral beamwidth ratios of nearly an order of magnitude. To mitigate this problem, the use of a pair of orthogonally oriented diagnostic arrays was recently proposed, with potential benefits arising from the substantially expanded range of observation angles. This article presents experiments and simulations intended to demonstrate the performance and limitations of the dual-array system concept. The key finding of this study is that source pair resolution of better than 1 mm is now possible in both dimensions of the imaging plane using a pair of 7.5-MHz center frequency conventional arrays at a distance of 7.6cm. With an eye toward accelerating computations for real-time applications, channel count reductions of up to a factor of eight induce negligible performance losses. Modest sensitivities to sound speed and relative array position uncertainties were identified, but if these can be kept on the order of 1% and 1 mm, respectively, then the proposed methods offer the potential for a step improvement in cavitation monitoring capability.
Collapse
|
28
|
Grundy M, Bau L, Hill C, Paverd C, Mannaris C, Kwan J, Crake C, Coviello C, Coussios C, Carlisle R. Improved therapeutic antibody delivery to xenograft tumors using cavitation nucleated by gas-entrapping nanoparticles. Nanomedicine (Lond) 2021; 16:37-50. [PMID: 33426913 DOI: 10.2217/nnm-2020-0263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aims: Testing ultrasound-mediated cavitation for enhanced delivery of the therapeutic antibody cetuximab to tumors in a mouse model. Methods: Tumors with strong EGF receptor expression were grown bilaterally. Cetuximab was coadministered intravenously with cavitation nuclei, consisting of either the ultrasound contrast agent Sonovue or gas-stabilizing nanoscale SonoTran Particles. One of the two tumors was exposed to focused ultrasound. Passive acoustic mapping localized and monitored cavitation activity. Both tumors were then excised and cetuximab concentration was quantified. Results: Cavitation increased tumoral cetuximab concentration. When nucleated by Sonovue, a 2.1-fold increase (95% CI 1.3- to 3.4-fold) was measured, whereas SonoTran Particles gave a 3.6-fold increase (95% CI 2.3- to 5.8-fold). Conclusions: Ultrasound-mediated cavitation, especially when nucleated by nanoscale gas-entrapping particles, can noninvasively increase site-specific delivery of therapeutic antibodies to solid tumors.
Collapse
Affiliation(s)
- Megan Grundy
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Luca Bau
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Claudia Hill
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Catherine Paverd
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Christophoros Mannaris
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - James Kwan
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK
| | - Calum Crake
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | | | - Constantin Coussios
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| | - Robert Carlisle
- Department of Engineering Science, Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory (BUBBL), Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
29
|
Pellow C, O'Reilly MA, Hynynen K, Zheng G, Goertz DE. Simultaneous Intravital Optical and Acoustic Monitoring of Ultrasound-Triggered Nanobubble Generation and Extravasation. NANO LETTERS 2020; 20:4512-4519. [PMID: 32374617 DOI: 10.1021/acs.nanolett.0c01310] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ultrasound-activated nanobubbles are being widely investigated as contrast agents and therapeutic vehicles. Nanobubbles hold potential for accessing the tumor extravascular compartment, though this relies on clinically debated passive accumulation for which evidence to date is indirect. We recently reported ultrasound-triggered conversion of high payload porphyrin-encapsulated microbubbles to nanobubbles, with actively enhanced permeability for local delivery. This platform holds implications for optical/ultrasound-based imaging and therapeutics. While promising, it remains to be established how nanobubbles are generated and whether they extravasate intact. Here, insights into the conversion process are reported, complemented by novel simultaneous intravital and acoustic monitoring in tumor-affected functional circulation. The first direct acoustic evidence of extravascular intact nanobubbles are presented. These insights collectively advance this delivery platform and multimodal micro- and nanobubbles, extending their utility for imaging and therapeutics within and beyond the vasculature.
Collapse
Affiliation(s)
- Carly Pellow
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 2C1, Canada
| | - Meaghan A O'Reilly
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| | - Kullervo Hynynen
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| | - Gang Zheng
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 2C1, Canada
| | - David E Goertz
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| |
Collapse
|
30
|
Stride E, Segers T, Lajoinie G, Cherkaoui S, Bettinger T, Versluis M, Borden M. Microbubble Agents: New Directions. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1326-1343. [PMID: 32169397 DOI: 10.1016/j.ultrasmedbio.2020.01.027] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 05/24/2023]
Abstract
Microbubble ultrasound contrast agents have now been in use for several decades and their safety and efficacy in a wide range of diagnostic applications have been well established. Recent progress in imaging technology is facilitating exciting developments in techniques such as molecular, 3-D and super resolution imaging and new agents are now being developed to meet their specific requirements. In parallel, there have been significant advances in the therapeutic applications of microbubbles, with recent clinical trials demonstrating drug delivery across the blood-brain barrier and into solid tumours. New agents are similarly being tailored toward these applications, including nanoscale microbubble precursors offering superior circulation times and tissue penetration. The development of novel agents does, however, present several challenges, particularly regarding the regulatory framework. This article reviews the developments in agents for diagnostic, therapeutic and "theranostic" applications; novel manufacturing techniques; and the opportunities and challenges for their commercial and clinical translation.
Collapse
Affiliation(s)
- Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| | - Tim Segers
- Physics of Fluids Group, Technical Medical (TechMed) Centre, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | - Guillaume Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Centre, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | - Samir Cherkaoui
- Bracco Suisse SA - Business Unit Imaging, Global R&D, Plan-les-Ouates, Switzerland
| | - Thierry Bettinger
- Bracco Suisse SA - Business Unit Imaging, Global R&D, Plan-les-Ouates, Switzerland
| | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Centre, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | - Mark Borden
- Mechanical Engineering Department, University of Colorado, Boulder, CO, USA
| |
Collapse
|
31
|
Murphy EM, Centner CS, Bates PJ, Malik MT, Kopechek JA. Delivery of thymoquinone to cancer cells with as1411-conjugated nanodroplets. PLoS One 2020; 15:e0233466. [PMID: 32437399 PMCID: PMC7241745 DOI: 10.1371/journal.pone.0233466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
Systemic delivery of conventional chemotherapies can cause negative systemic toxicity, including reduced immunity and damage to organs such as the heart and kidneys-limiting the maximum dose that can be administered. Targeted therapies appear to address this problem by having a specific target while mitigating off-target effects. Biocompatible perfluorocarbon-based nanodroplet emulsions encapsulated by a phospholipid shell are in development for delivery of molecular compounds and hold promise as vehicles for targeted delivery of chemotherapeutics to tumors. When ultrasound is applied, perfluorocarbon will undergo a phase change-ultimately inducing transient perforation of the cell membrane when in close proximity, which is more commonly known as "sonoporation." Sonoporation allows enhanced intracellular delivery of molecular compounds and will reseal to encapsulate the molecular compound intracellularly. In this study, we investigated delivery of thymoquinone (TQ), a natural hydrophobic phytochemical compound with bioactivity in cancer cells. In addition, we conjugated a G-quadruplex aptamer, 'AS1411', to TQ-loaded nanodroplets and explored their effects on multiple human cancer cell lines. AS1411 binds nucleolin, which is over-expressed on the surface of cancer cells, and in addition to its tumor-targeting properties AS1411 has also been shown to induce anti-cancer effects. Thymoquinone was loaded onto AS1411-conjugated nanodroplet emulsion to assess activity against cancer cells. Confocal microscopy indicated uptake of AS1411-conjugated nanodroplets by cancer cells. Furthermore, AS1411-conjugated nanoemulsions loaded with TQ significantly enhanced cytotoxicity in cancer cells compared to free compound. These results demonstrate that AS1411 can be conjugated onto nanodroplet emulsions for targeted delivery to human cancer cells. This novel formulation offers significant potential for targeted delivery of hydrophobic chemotherapeutics to tumors for cancer treatment.
Collapse
Affiliation(s)
- Emily M. Murphy
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, United States of America
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Connor S. Centner
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, United States of America
| | - Paula J. Bates
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
- Molecular Targets Program of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Mohammad T. Malik
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
- Molecular Targets Program of the James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, United States of America
| | - Jonathan A. Kopechek
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|
32
|
Ho YJ, Li JP, Fan CH, Liu HL, Yeh CK. Ultrasound in tumor immunotherapy: Current status and future developments. J Control Release 2020; 323:12-23. [PMID: 32302759 DOI: 10.1016/j.jconrel.2020.04.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/24/2022]
Abstract
Immunotherapy has considerable potential in eliminating cancers by activating the host's own immune system, while the thermal and mechanical effects of ultrasound have various applications in tumor therapy. Hyperthermia, ablation, histotripsy, and microbubble stable/inertial cavitation can alter the tumor microenvironment to enhance immunoactivation to inhibit tumor growth. Microbubble cavitation can increase vessel permeability and thereby improve the delivery of immune cells, cytokines, antigens, and antibodies to tumors. Violent microbubble cavitation can disrupt tumor cells and efficiently expose them to numerous antigens so as to promote the maturity of antigen-presenting cells and subsequent adaptive immune-cell activation. This review provides an overview and compares the mechanisms of ultrasound-induced immune modulation for peripheral and brain tumor therapy, even degenerative brain diseases therapy. The possibility of reversing tumors to an immunoactive microenvironment by utilizing the cavitation of microbubbles loaded with therapeutic gases is also proposed as another potential pathway for immunotherapy. Finally, we disuss the challenges and opportunities of ultrasound in immunotherapy for future development.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ju-Pi Li
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan; School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang-Gung University, Taoyuan 333, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
33
|
Owen J, Thomas E, Menon J, Gray M, Skaripa-Koukelli I, Gill MR, Wallington S, Miller RL, Vallis KA, Carlisle R. Indium-111 labelling of liposomal HEGF for radionuclide delivery via ultrasound-induced cavitation. J Control Release 2020; 319:222-233. [PMID: 31891732 DOI: 10.1016/j.jconrel.2019.12.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023]
Abstract
The purpose of this exploratory study was to investigate the combination of a radiopharmaceutical, nanoparticles and ultrasound (US) enhanced delivery to develop a clinically viable therapeutic strategy for tumours overexpressing the epidermal growth factor receptor (EGFR). Molecularly targeted radionuclides have great potential for cancer therapy but are sometimes associated with insufficient delivery resulting in sub-cytotoxic amounts of radioactivity being delivered to the tumour. Liposome formulations are currently used in the clinic to reduce the side effects and improve the pharmacokinetic profile of chemotherapeutic drugs. However, in contrast to non-radioactive agents, loading and release of radiotherapeutics from liposomes can be challenging in the clinical setting. US-activated cavitation agents such as microbubbles (MBs) have been used to release therapeutics from liposomes to enhance the distribution/delivery in a target area. In an effort to harness the benefits of these techniques, the development of a liposome loaded radiopharmaceutical construct for enhanced delivery via acoustic cavitation was studied. The liposomal formulation was loaded with peptide, human epidermal growth factor (HEGF), coupled to a chelator for subsequent radiolabelling with 111Indium ([111In]In3+), in a manner designed to be compatible with preparation in a radiopharmacy. Liposomes were efficiently radiolabelled (57%) within 1 h, with release of ~12% of the radiopeptide following a 20 s exposure to US-mediated cavitation in vitro. In clonogenic studies this level of release resulted in cytotoxicity specifically in cells over-expressing the epidermal growth factor receptor (EGFR), with over 99% reduction in colony survival compared to controls. The formulation extended the circulation time and changed the biodistribution compared to the non-liposomal radiopeptide in vivo, although interestingly the biodistribution did not resemble that of liposome constructs currently used in the clinic. Cavitation of MBs co-injected with liposomes into tumours expressing high levels of EGFR resulted in a 2-fold enhancement in tumour uptake within 20 min. However, owing to the poor vascularisation of the tumour model used the same level of uptake was achieved without US after 24 h. By combining acoustic-cavitation-sensitive liposomes with radiopharmaceuticals this research represents a new concept in achieving targeted delivery of radiopharmaceuticals.
Collapse
Affiliation(s)
- Joshua Owen
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Eloise Thomas
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Jyothi Menon
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK; College of Pharmacy, The University of Rhode Island, Kingston, RI 02881, USA
| | - Michael Gray
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Irini Skaripa-Koukelli
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Martin R Gill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Sheena Wallington
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Rebecca L Miller
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Robert Carlisle
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
34
|
Wang L, Li X, Dong Y, Wang P, Xu M, Zheng C, Jiao Y, Zou C. Effects of Cytotoxic T Lymphocyte-Associated Antigen 4 Immunoglobulin Combined with Microbubble-Mediated Irradiation on Hemodynamics of the Renal Artery in Rats with Diabetic Nephropathy. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:703-711. [PMID: 31864804 DOI: 10.1016/j.ultrasmedbio.2019.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 06/10/2023]
Abstract
Cytotoxic T lymphocyte-associated antigen 4 immunoglobulin (CTLA-4-Ig) can inhibit the effect of B7-1 and improve renal hemodynamics in rats with diabetic nephropathy (DN). Nevertheless, a strategy that could increase the permeation of CTLA-4-Ig through endothelial cells and basement membrane remains to be discovered. We investigated the effect of CTLA-4-Ig combined with microbubble-mediated irradiation on the hemodynamics of renal arteries in DN rats. Rats were treated with CTLA-4-Ig and/or microbubble exposure. After 8 wk of intervention, color Doppler ultrasonography was used to detect peak systolic velocity (PSV), end-diastolic velocity (EDV), mean velocity (MV), systolic acceleration (SAC), pulsatility index (PI) and resistance index (RI) of the renal artery trunk. The CTLA-4-Ig + microbubble exposure group exhibited significantly higher PSV, EDV and MV than the CTLA-4-Ig group, which had significantly higher values than the non-intervention group. The CTLA-4-Ig + microbubble exposure group exhibited significantly lower SAC, PI and RI than the CTLA-4-Ig group, which had significantly lower values than the non-intervention group. Our results indicate that both CTLA-4-Ig and CTLA-4-Ig + microbubble exposure can reduce the blood flow resistance and improve the blood flow velocity of renal arteries in rats. Moreover, the effect of CTLA-4-Ig + microbubble exposure is better than that of CTLA-4-Ig alone. Our study provides a new, effective and non-invasive strategy for the treatment of DN.
Collapse
Affiliation(s)
- Liang Wang
- Department of Ultrasonic Diagnosis, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiuyun Li
- Department of Ultrasonic Diagnosis, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanyan Dong
- Department of Ultrasonic Diagnosis, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengfei Wang
- Department of Ultrasonic Diagnosis, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Maosheng Xu
- Department of Ultrasonic Diagnosis, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chao Zheng
- Diabetes Center and Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Jiao
- Obstetrics and Gynecology Ultrasonic Department, Wenzhou City People's Hospital, Wenzhou, China
| | - Chunpeng Zou
- Department of Ultrasonic Diagnosis, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
35
|
Paris JL, Vallet-Regí M. Ultrasound-Activated Nanomaterials for Therapeutics. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020; 93:220-229. [PMID: 39650549 PMCID: PMC7617089 DOI: 10.1246/bcsj.20190346] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Ultrasound has attracted much attention in recent years as an external stimulus capable of activating different types of nanomaterials for therapeutic application. One of the characteristics that makes ultrasound an especially appealing triggering stimulus for nanomedicine is its capacity to be non-invasively applied in a focused manner at deep regions of the body. Combining ultrasound with nanoparticles, different biological effects can be achieved. In this work, an overview of the four main types of inducible responses will be provided: inducing drug release, producing ultrasound-derived biological effects, modifying nanoparticle biodistribution and developing theranostic agents. Several examples of each one of these applications are presented here to illustrate the key concepts underlying recent developments in the discipline.
Collapse
Affiliation(s)
- Juan L. Paris
- Department of Life Sciences, Nano4Health Unit, Nanomedicine Group. International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040-Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
36
|
de la Torre P, Pérez-Lorenzo MJ, Alcázar-Garrido Á, Flores AI. Cell-Based Nanoparticles Delivery Systems for Targeted Cancer Therapy: Lessons from Anti-Angiogenesis Treatments. Molecules 2020; 25:E715. [PMID: 32046010 PMCID: PMC7038177 DOI: 10.3390/molecules25030715] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 02/05/2023] Open
Abstract
The main strategy of cancer treatment has focused on attacking the tumor cells. Some cancers initially responsive to chemotherapy become treatment-resistant. Another strategy is to block the formation of tumor vessels. However, tumors also become resistant to anti-angiogenic treatments, mostly due to other cells and factors present in the tumor microenvironment, and hypoxia in the central part of the tumor. The need for new cancer therapies is significant. The use of nanoparticle-based therapy will improve therapeutic efficacy and targeting, while reducing toxicity. However, due to inefficient accumulation in tumor sites, clearance by reticuloendothelial organs and toxicity, internalization or conjugation of drug-loaded nanoparticles (NPs) into mesenchymal stem cells (MSCs) can increase efficacy by actively delivering them into the tumor microenvironment. Nanoengineering MSCs with drug-loaded NPs can increase the drug payload delivered to tumor sites due to the migratory and homing abilities of MSCs. However, MSCs have some disadvantages, and exosomes and membranes from different cell types can be used to transport drug-loaded NPs actively to tumors. This review gives an overview of different cancer approaches, with a focus on hypoxia and the emergence of NPs as drug-delivery systems and MSCs as cellular vehicles for targeted delivery due to their tumor-homing potential.
Collapse
Affiliation(s)
| | | | | | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas 12), Avda. de Cordoba s/n, 28041 Madrid, Spain; (P.d.l.T.); (M.J.P.-L.)
| |
Collapse
|
37
|
Xu W, Zhang X, Hu X, Zhiyi C, Huang P. Translational Prospects of ultrasound-mediated tumor immunotherapy: Preclinical advances and safety considerations. Cancer Lett 2019; 460:86-95. [DOI: 10.1016/j.canlet.2019.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022]
|
38
|
Thomas RG, Jonnalagadda US, Kwan JJ. Biomedical Applications for Gas-Stabilizing Solid Cavitation Agents. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10106-10115. [PMID: 31045378 DOI: 10.1021/acs.langmuir.9b00795] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
For over a decade, advancements in ultrasound-enhanced drug delivery strategies have demonstrated remarkable success in providing targeted drug delivery for a broad range of diseases. In order to achieve enhanced drug delivery, these strategies harness the mechanical effects from bubble oscillations (i.e., cavitation) of a variety of exogenous cavitation agents. Recently, solid cavitation agents have emerged due to their capacity for drug-loading and sustained cavitation duration. Unlike other cavitation agents, solid cavitation agents stabilize gaseous bubbles on hydrophobic surface cavities. Thus, the design of these particles is crucial. In this Review, we provide an overview of the different designs for solid cavitation agents such as nanocups, nanocones, and porous structures, as well as the current status of their development. Considering the numerous advantages of solid cavitation agents, we anticipate further innovations for this new type of cavitation agent across a broad range of biomedical applications.
Collapse
Affiliation(s)
- Reju G Thomas
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore , 637459
| | - Umesh S Jonnalagadda
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore , 637459
| | - James J Kwan
- School of Chemical and Biomedical Engineering , Nanyang Technological University , Singapore , 637459
| |
Collapse
|
39
|
Thomas E, Menon JU, Owen J, Skaripa-Koukelli I, Wallington S, Gray M, Mannaris C, Kersemans V, Allen D, Kinchesh P, Smart S, Carlisle R, Vallis KA. Ultrasound-mediated cavitation enhances the delivery of an EGFR-targeting liposomal formulation designed for chemo-radionuclide therapy. Theranostics 2019; 9:5595-5609. [PMID: 31534505 PMCID: PMC6735398 DOI: 10.7150/thno.34669] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/08/2019] [Indexed: 12/19/2022] Open
Abstract
Nanomedicines allow active targeting of cancer for diagnostic and therapeutic applications through incorporation of multiple functional components. Frequently, however, clinical translation is hindered by poor intratumoural delivery and distribution. The application of physical stimuli to promote tumour uptake is a viable route to overcome this limitation. In this study, ultrasound-mediated cavitation of microbubbles was investigated as a mean of enhancing the delivery of a liposome designed for chemo-radionuclide therapy targeted to EGFR overexpressing cancer. Method: Liposomes (111In-EGF-LP-Dox) were prepared by encapsulation of doxorubicin (Dox) and surface functionalisation with Indium-111 tagged epidermal growth factor. Human breast cancer cell lines with high and low EGFR expression (MDA-MB-468 and MCF7 respectively) were used to study selectivity of liposomal uptake, subcellular localisation of drug payload, cytotoxicity and DNA damage. Liposome extravasation following ultrasound-induced cavitation of microbubbles (SonoVue®) was studied using a tissue-mimicking phantom. In vivo stability, pharmacokinetic profile and biodistribution were evaluated following intravenous administration of 111In-labelled, EGF-functionalised liposomes to mice bearing subcutaneous MDA-MB-468 xenografts. Finally, the influence of ultrasound-mediated cavitation on the delivery of liposomes into tumours was studied. Results: Liposomes were loaded efficiently with Dox, surface decorated with 111In-EGF and showed selective uptake in MDA-MB-468 cells compared to MCF7. Following binding to EGFR, Dox was released into the intracellular space and 111In-EGF shuttled to the cell nucleus. DNA damage and cell kill were higher in MDA-MB-468 than MCF7 cells. Moreover, Dox and 111In were shown to have an additive cytotoxic effect in MDA-MB-468 cells. US-mediated cavitation increased the extravasation of liposomes in an in vitro gel phantom model. In vivo, the application of ultrasound with microbubbles increased tumour uptake by 66% (p<0.05) despite poor vascularisation of MDA-MB-468 xenografts (as shown by DCE-MRI). Conclusion:111In-EGF-LP-Dox designed for concurrent chemo-radionuclide therapy showed specificity for and cytotoxicity towards EGFR-overexpressing cancer cells. Delivery to tumours was enhanced by the use of ultrasound-mediated cavitation indicating that this approach has the potential to deliver cytotoxic levels of therapeutic radionuclide to solid tumours.
Collapse
Affiliation(s)
- Eloise Thomas
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Jyothi U. Menon
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Joshua Owen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Irini Skaripa-Koukelli
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sheena Wallington
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Christophoros Mannaris
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Veerle Kersemans
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Danny Allen
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Paul Kinchesh
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Sean Smart
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Katherine A. Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
40
|
Su X, Thomas RG, Bharatula LD, Kwan JJ. Remote targeted implantation of sound-sensitive biodegradable multi-cavity microparticles with focused ultrasound. Sci Rep 2019; 9:9612. [PMID: 31270380 PMCID: PMC6610131 DOI: 10.1038/s41598-019-46022-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/20/2019] [Indexed: 01/04/2023] Open
Abstract
Ultrasound-enhanced drug delivery has shown great promise in providing targeted burst release of drug at the site of the disease. Yet current solid ultrasound-responsive particles are non-degradable with limited potential for drug-loading. Here, we report on an ultrasound-responsive multi-cavity poly(lactic-co-glycolic acid) microparticle (mcPLGA MP) loaded with rhodamine B (RhB) with or without 4',6-diamidino-2-phenylindole (DAPI) to represent small molecule therapeutics. After exposure to high intensity focused ultrasound (HIFU), these delivery vehicles were remotely implanted into gel and porcine tissue models, where the particles rapidly released their payload within the first day and sustained release for at least seven days. RhB-mcPLGA MPs were implanted with HIFU into and beyond the sub-endothelial space of porcine arteries without observable damage to the artery. HIFU also guided the location of implantation; RhB-mcPLGA MPs were only observed at the focus of the HIFU away from the direction of ultrasound. Once implanted, DAPI co-loaded RhB-mcPLGA MPs released DAPI into the arterial wall, staining the nucleus of the cells. Our work shows the potential for HIFU-guided implantation of drug-loaded particles as a strategy to improve the local and sustained delivery of a therapeutic for up to two weeks.
Collapse
Affiliation(s)
- Xiaoqian Su
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Reju George Thomas
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Lakshmi Deepika Bharatula
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - James J Kwan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore.
| |
Collapse
|