1
|
Snipstad S, Einen C, Kastellet AB, Fernandez JL, Mühlenpfordt M, Kurbatskaya A, Årseth C, Berg S, Bjørkøy A, Davies CDL. Ultrasound and Microbubble-Induced Reduction of Functional Vasculature Depends on the Microbubble, Tumor Type and Time After Treatment. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00337-5. [PMID: 39389855 DOI: 10.1016/j.ultrasmedbio.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/15/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024]
Abstract
OBJECTIVE Ultrasound in combination with microbubbles can enhance accumulation and improve the distribution of various therapeutic agents in tumor tissue, leading to improved efficacy. Understanding the impact of treatment on the tumor microenvironment, concurrently with how microenvironment attributes affect treatment outcome, will be important for selecting appropriate patient cohorts in future clinical trials. The main aim of this work was to investigate the influence of ultrasound and microbubbles on the functional vasculature of cancer tissue. METHODS Four different tumor models in mice (bone, pancreatic, breast and colon cancer) were characterized with respect to vascular parameters using contrast-enhanced ultrasound imaging. The effect of treatment with microbubbles and ultrasound was then investigated using immunohistochemistry and confocal microscopy, quantifying the total amount of vasculature and fraction of functional vessels. Two different microbubbles were used, the clinical contrast agent SonoVue and the large bubbles generated by Acoustic Cluster Therapy (ACT), tailored for therapeutic purposes. RESULTS The colon cancer model displayed slower flow but a higher vascular volume than the other models. The pancreatic model showed the fastest flow but also the lowest vascular volume. Ultrasound and SonoVue transiently reduced the amount of functional vasculature in breast and colon tumors immediately after treatment. No reduction was observed for ACT, likely due to shorter ultrasound pulses and lower pressures applied. CONCLUSION Variation between tumor models due to tissue characteristics emphasizes the importance of evaluating treatment suitability in the specific tissue of interest, as altered perfusion could have a large impact on drug delivery and therapeutic outcome.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Caroline Einen
- Porelab and Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andrea Berge Kastellet
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jessica Lage Fernandez
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna Kurbatskaya
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Charlotte Årseth
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | | |
Collapse
|
2
|
Mühlenpfordt M, Olsen EB, Kotopoulis S, Torp SH, Snipstad S, Davies CDL, Olsman M. Real-Time Intravital Imaging of Acoustic Cluster Therapy-Induced Vascular Effects in the Murine Brain. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1212-1226. [PMID: 36858913 DOI: 10.1016/j.ultrasmedbio.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE The blood-brain barrier (BBB) is an obstacle for cerebral drug delivery. Controlled permeabilization of the barrier by external stimuli can facilitate the delivery of drugs to the brain. Acoustic Cluster Therapy (ACT®) is a promising strategy for transiently and locally increasing the permeability of the BBB to macromolecules and nanoparticles. However, the mechanism underlying the induced permeability change and subsequent enhanced accumulation of co-injected molecules requires further elucidation. METHODS In this study, the behavior of ACT® bubbles in microcapillaries in the murine brain was observed using real-time intravital multiphoton microscopy. For this purpose, cranial windows aligned with a ring transducer centered around an objective were mounted to the skull of mice. Dextrans labeled with 2 MDa fluorescein isothiocyanate (FITC) were injected to delineate the blood vessels and to visualize extravasation. DISCUSSION Activated ACT® bubbles were observed to alter the blood flow, inducing transient and local increases in the fluorescence intensity of 2 MDa FITC-dextran and subsequent extravasation in the form of vascular outpouchings. The observations indicate that ACT® induced a transient vascular leakage without causing substantial damage to the vessels in the brain. CONCLUSION The study gave novel insights into the mechanism underlying ACT®-induced enhanced BBB permeability which will be important considering treatment optimization for a safe and efficient clinical translation of ACT®.
Collapse
Affiliation(s)
- Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Exact Therapeutics AS, Oslo, Norway.
| | - Emma Bøe Olsen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Spiros Kotopoulis
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre H Torp
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Pathology, St. Olav's Hospital, Trondheim University Hospital Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | | | - Marieke Olsman
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
3
|
Effect of acoustic cluster therapy (ACT®) combined with chemotherapy in a patient-derived xenograft mouse model of pancreatic cancer. J Control Release 2022; 352:1134-1143. [PMID: 36372388 DOI: 10.1016/j.jconrel.2022.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022]
Abstract
Pancreatic ductal adenocarcinomas respond poorly to chemotherapy, in part due to the dense tumor stroma that hinders drug delivery. Ultrasound (US) in combination with microbubbles has previously shown promise as a means to improve drug delivery, and the therapeutic efficacy of ultrasound-mediated drug delivery is currently being evaluated in multiple clinical trials. However, most of these utilize echogenic contrast agents engineered for imaging, which might not be optimal compared to specialized formulations tailored for drug delivery. In this study, we evaluated the in vivo efficacy of phase-shifting microbubble-microdroplet clusters that, upon insonation, form bubbles in the size range of 20-30 μm. We developed a patient-derived xenograft model of pancreatic cancer implanted in mice that largely retained the stromal content of the originating tumor and compared tumor growth in mice given chemotherapeutics (nab-paclitaxel plus gemcitabine or liposomal irinotecan) with mice given the same chemotherapeutics in addition to ultrasound and acoustic cluster therapy. We found that acoustic cluster therapy significantly improved the effect of both chemotherapeutic regimens and resulted in 7.2 times higher odds of complete remission of the tumor compared to the chemotherapeutics alone.
Collapse
|
4
|
van Wamel A, Mühlenpfordt M, Hansen R, Healey A, Villanueva FS, Kotopoulis S, Davies CDL, Chen X. Ultrafast Microscopy Imaging of Acoustic Cluster Therapy Bubbles: Activation and Oscillation. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1840-1857. [PMID: 35773079 DOI: 10.1016/j.ultrasmedbio.2022.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/15/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Acoustic Cluster Therapy (ACT®) is a platform for improving drug delivery and has had promising pre-clinical results. A clinical trial is ongoing. ACT® is based on microclusters of microbubbles-microdroplets that, when sonicated, form a large ACT® bubble. The aim of this study was to obtain new knowledge on the dynamic formation and oscillations of ACT® bubbles by ultrafast optical imaging in a microchannel. The high-speed recordings revealed the microbubble-microdroplet fusion, and the gas in the microbubble acted as a vaporization seed for the microdroplet. Subsequently, the bubble grew by gas diffusion from the surrounding medium and became a large ACT® bubble with a diameter of 5-50 μm. A second ultrasound exposure at lower frequency caused the ACT® bubble to oscillate. The recorded oscillations were compared with simulations using the modified Rayleigh-Plesset equation. A term accounting for the physical boundary imposed by the microchannel wall was included. The recorded oscillation amplitudes were approximately 1-2 µm, hence similar to oscillations of smaller contrast agent microbubbles. These findings, together with our previously reported promising pre-clinical therapeutic results, suggest that these oscillations covering a large part of the vessel wall because of the large bubble volume can substantially improve therapeutic outcome.
Collapse
Affiliation(s)
- Annemieke van Wamel
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Spiros Kotopoulis
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Fan CH, Ho YJ, Lin CW, Wu N, Chiang PH, Yeh CK. State-of-the-art of ultrasound-triggered drug delivery from ultrasound-responsive drug carriers. Expert Opin Drug Deliv 2022; 19:997-1009. [PMID: 35930441 DOI: 10.1080/17425247.2022.2110585] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The development of new tools to locally and non-invasively transferring therapeutic substances at the desired site in deep living tissue has been a long sought-after goal within the drug delivery field. Among the established methods, ultrasound (US) with US-responsive carriers holds great promise and demonstrates on-demand delivery of a variety of functional substances with spatial precision of several millimeters in deep-seated tissues in animal models and humans. These properties have motivated several explorations of US with US responsive carriers as a modality for neuromodulation and the treatment of various diseases, such as stroke and cancer. AREAS COVERED This article briefly discussed three specific mechanisms that enhance in vivo drug delivery via US with US-responsive carriers: 1) permeabilizing cellular membrane, 2) increasing the permeability of vessels, and 3) promoting cellular endocytotic uptake. Besides, a series of US-responsive drug carriers are discussed, with an emphasis on the relation between structural feature and therapeutic outcome. EXPERT OPINION This article summarized current development for each of US-responsive drug carrier, focusing on the routes of enhancing delivery and applications. The mechanisms of interaction between US-responsive carriers and US energy, such as cavitation, hyperthermia, and reactive oxygen species, as well as how these interactions can improve drug delivery into target cell/tissue. It can be expected that there are serval efforts to further identification of US-responsive particles, design of novel US waveform sequence, and survey of optimal combination between US parameters and US-responsive carriers for better controlling the spatiotemporal drug release profile, stability, and safety in vivo. The authors believe these will provide novel tools for precisely designing treatment strategies and significantly benefit the clinical management of several diseases.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Hua Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
6
|
Ni N, Wang W, Sun Y, Sun X, Leong DT. Inducible endothelial leakiness in nanotherapeutic applications. Biomaterials 2022; 287:121640. [PMID: 35772348 DOI: 10.1016/j.biomaterials.2022.121640] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/02/2022]
Abstract
All intravenous delivered nanomedicine needs to escape from the blood vessel to exert their therapeutic efficacy at their designated site of action. Failure to do so increases the possibility of detrimental side effects and negates their therapeutic intent. Many powerful anticancer nanomedicine strategies rely solely on the tumor derived enhanced permeability and retention (EPR) effect for the only mode of escaping from the tumor vasculature. However, not all tumors have the EPR effect nor can the EPR effect be induced or controlled for its location and timeliness. In recent years, there have been exciting developments along the lines of inducing endothelial leakiness at the tumor to decrease the dependence of EPR. Physical disruption of the endothelial-endothelial cell junctions with coordinated biological intrinsic pathways have been proposed that includes various modalities like ultrasound, radiotherapy, heat and even nanoparticles, appear to show good progress towards the goal of inducing endothelial leakiness. This review explains the intricate and complex biological background behind the endothelial cells with linkages on how updated reported nanomedicine strategies managed to induce endothelial leakiness. This review will also end off with fresh insights on where the future of inducible endothelial leakiness holds.
Collapse
Affiliation(s)
- Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Weiyi Wang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yu Sun
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore; Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, PR China
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.
| |
Collapse
|
7
|
Kotopoulis S, Lam C, Haugse R, Snipstad S, Murvold E, Jouleh T, Berg S, Hansen R, Popa M, Mc Cormack E, Gilja OH, Poortinga A. Formulation and characterisation of drug-loaded antibubbles for image-guided and ultrasound-triggered drug delivery. ULTRASONICS SONOCHEMISTRY 2022; 85:105986. [PMID: 35358937 PMCID: PMC8967728 DOI: 10.1016/j.ultsonch.2022.105986] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 06/13/2023]
Abstract
The aim of this study was to develop high load-capacity antibubbles that can be visualized using diagnostic ultrasound and the encapsulated drug can be released and delivered using clinically translatable ultrasound. The antibubbles were developed by optimising a silica nanoparticle stabilised double emulsion template. We produced an emulsion with a mean size diameter of 4.23 ± 1.63 µm where 38.9 ± 3.1% of the droplets contained a one or more cores. Following conversion to antibubbles, the mean size decreased to 2.96 ± 1.94 µm where 99% of antibubbles were <10 µm. The antibubbles had a peak attenuation of 4.8 dB/cm at 3.0 MHz at a concentration of 200 × 103 particles/mL and showed distinct attenuation spikes at frequencies between 5.5 and 13.5 MHz. No increase in subharmonic response was observed for the antibubbles in contrast to SonoVue®. High-speed imaging revealed that antibubbles can release their cores at MIs of 0.6. In vivo imaging indicated that the antibubbles have a long half-life of 68.49 s vs. 40.02 s for SonoVue®. The antibubbles could be visualised using diagnostic ultrasound and could be disrupted at MIs of ≥0.6. The in vitro drug delivery results showed that antibubbles can significantly improve drug delivery (p < 0.0001) and deliver the drug within the antibubbles. In conclusion antibubbles are a viable concept for ultrasound guided drug delivery.
Collapse
Affiliation(s)
- Spiros Kotopoulis
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Neoety AS, Kløfta, Norway.
| | - Christina Lam
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ragnhild Haugse
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Quality and Development, Hospital Pharmacies Enterprise in Western Norway, Bergen, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Elisa Murvold
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; KinN Therapeutics, Bergen, Norway
| | - Tæraneh Jouleh
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Sigrid Berg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Rune Hansen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Mihaela Popa
- Department of Clinical Science, University of Bergen, Bergen, Norway; CCBIO, Department of Clinical Science, University of Bergen, Norway
| | - Emmet Mc Cormack
- Department of Clinical Science, University of Bergen, Bergen, Norway; KinN Therapeutics, Bergen, Norway
| | - Odd Helge Gilja
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Albert Poortinga
- Polymer Technology, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
8
|
Omata D, Munakata L, Maruyama K, Suzuki R. Enhanced Vascular Permeability by Microbubbles and Ultrasound in Drug Delivery. Biol Pharm Bull 2021; 44:1391-1398. [PMID: 34602547 DOI: 10.1248/bpb.b21-00453] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ultrasound and microbubbles, an ultrasound contrast agent, have recently increased attention to developing novel drug delivery systems. Ultrasound exposure can induce mechanical effects derived from microbubbles behaviors such as an expansion, contraction, and collapse depending on ultrasound conditions. These mechanical effects induce several biological effects, including enhancement of vascular permeability. For drug delivery, one promising approach is enhancing vascular permeability using ultrasound and microbubbles, resulting in improved drug transport to targeted tissues. This approach is applied to several tissues and drugs to cure diseases. This review describes the enhancement of vascular permeability by ultrasound and microbubbles and its therapeutic application, including our recent study. We also discuss the current situation of the field and its potential future perspectives.
Collapse
Affiliation(s)
- Daiki Omata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University
| | - Kazuo Maruyama
- Laboratory of Theranostics, Faculty of Pharma-Science, Teikyo University.,Advanced Comprehensive Research Organization (ACRO), Teikyo University
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University.,Advanced Comprehensive Research Organization (ACRO), Teikyo University
| |
Collapse
|
9
|
Snipstad S, Vikedal K, Maardalen M, Kurbatskaya A, Sulheim E, Davies CDL. Ultrasound and microbubbles to beat barriers in tumors: Improving delivery of nanomedicine. Adv Drug Deliv Rev 2021; 177:113847. [PMID: 34182018 DOI: 10.1016/j.addr.2021.113847] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022]
Abstract
Successful delivery of drugs and nanomedicine to tumors requires a functional vascular network, extravasation across the capillary wall, penetration through the extracellular matrix, and cellular uptake. Nanomedicine has many merits, but penetration deep into the tumor interstitium remains a challenge. Failure of cancer treatment can be caused by insufficient delivery of the therapeutic agents. After intravenous administration, nanomedicines are often found in off-target organs and the tumor extracellular matrix close to the capillary wall. With circulating microbubbles, ultrasound exposure focused toward the tumor shows great promise in improving the delivery of therapeutic agents. In this review, we address the impact of focused ultrasound and microbubbles to overcome barriers for drug delivery such as perfusion, extravasation, and transport through the extracellular matrix. Furthermore, we discuss the induction of an immune response with ultrasound and delivery of immunotherapeutics. The review discusses mainly preclinical results and ends with a summary of ongoing clinical trials.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Krister Vikedal
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Matilde Maardalen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna Kurbatskaya
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | | |
Collapse
|
10
|
Sulheim E, Hanson I, Snipstad S, Vikedal K, Mørch Y, Boucher Y, Davies CDL. Sonopermeation with Nanoparticle‐Stabilized Microbubbles Reduces Solid Stress and Improves Nanomedicine Delivery to Tumors. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Einar Sulheim
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Ingunn Hanson
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Sofie Snipstad
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Krister Vikedal
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
| | - Yves Boucher
- Edwin L. Steele Laboratory for Tumor Biology Massachusetts General Hospital Boston MA 02114 USA
| | - Catharina de Lange Davies
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| |
Collapse
|
11
|
Olsman M, Mühlenpfordt M, Olsen EB, Torp SH, Kotopoulis S, Rijcken CJF, Hu Q, Thewissen M, Snipstad S, de Lange Davies C. Acoustic Cluster Therapy (ACT®) enhances accumulation of polymeric micelles in the murine brain. J Control Release 2021; 337:285-295. [PMID: 34274386 DOI: 10.1016/j.jconrel.2021.07.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/22/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
The restrictive nature of the blood-brain barrier (BBB) prevents efficient treatment of many brain diseases. Focused ultrasound in combination with microbubbles has shown to safely and transiently increase BBB permeability. Here, the potential of Acoustic Cluster Therapy (ACT®), a microbubble platform specifically engineered for theranostic purposes, to increase the permeability of the BBB and improve accumulation of IRDye® 800CW-PEG and core-crosslinked polymeric micelles (CCPM) in the murine brain, was studied. Contrast enhanced magnetic resonance imaging (MRI) showed increased BBB permeability in all animals after ACT®. Near infrared fluorescence (NIRF) images of excised brains 1 h post ACT® revealed an increased accumulation of the IRDye® 800CW-PEG (5.2-fold) and CCPM (3.7-fold) in ACT®-treated brains compared to control brains, which was retained up to 24 h post ACT®. Confocal laser scanning microscopy (CLSM) showed improved extravasation and penetration of CCPM into the brain parenchyma after ACT®. Histological examination of brain sections showed no treatment related tissue damage. This study demonstrated that ACT® increases the permeability of the BBB and enhances accumulation of macromolecules and clinically relevant nanoparticles to the brain, taking a principal step in enabling improved treatment of various brain diseases.
Collapse
Affiliation(s)
- Marieke Olsman
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Emma Bøe Olsen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre H Torp
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Spiros Kotopoulis
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Exact Therapeutics, Oslo, Norway
| | | | - Qizhi Hu
- Cristal Therapeutics, Maastricht, the Netherlands
| | | | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olavs Hospital, Trondheim, Norway
| | | |
Collapse
|
12
|
Xia H, Yang D, He W, Zhu X, Yan Y, Liu Z, Liu T, Yang J, Tan S, Jiang J, Hou X, Gao H, Ni L, Lu J. Ultrasound-mediated microbubbles cavitation enhanced chemotherapy of advanced prostate cancer by increasing the permeability of blood-prostate barrier. Transl Oncol 2021; 14:101177. [PMID: 34271256 PMCID: PMC8287239 DOI: 10.1016/j.tranon.2021.101177] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/04/2021] [Accepted: 07/08/2021] [Indexed: 12/25/2022] Open
Abstract
Combination therapy increased cell apoptosis and the inhibition of cell viability. Combination therapy enhanced chemotherapy efficacy by increasing cell permeability. Success in developing an orthotopic model of prostate tumor implantation in mice. Combination therapy inhibited tumor growth and prolonged the survival of mice.
Although chemotherapy is an important treatment for advanced prostate cancer, its efficacy is relatively limited. Ultrasound-induced cavitation plays an important role in drug delivery and gene transfection. However, whether cavitation can improve the efficacy of chemotherapy for prostate cancer remains unclear. In this study, we treated RM-1 mouse prostate carcinoma cells with a combination of ultrasound-mediated microbubble cavitation and paclitaxel. Our results showed that combination therapy led to a more pronounced inhibition of cell viability and increased cell apoptosis. The enhanced efficacy of chemotherapy was attributed to the increased cell permeability induced by cavitation. Importantly, compared with chemotherapy alone (nab-paclitaxel), chemotherapy combined with ultrasound-mediated microbubble cavitation significantly inhibited tumor growth and prolonged the survival of tumor-bearing mice in an orthotopic mouse model of RM-1 prostate carcinoma, indicating the synergistic effects of combined therapy on tumor reduction. Furthermore, we analyzed tumor-infiltrating lymphocytes and found that during chemotherapy, the proportions of CTLA4+ cells and PD-1+/CTLA4+ cells in CD8+ T cells slightly increased after cavitation treatment.
Collapse
Affiliation(s)
- Haizhui Xia
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Decao Yang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Wei He
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Ye Yan
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Tong Liu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Jianling Yang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Shi Tan
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Jie Jiang
- Department of Ultrasound, Peking University Third Hospital, Beijing 100191, China
| | - Xiaofei Hou
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Medical Research Building, Beijing 100084, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
13
|
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: 7.0] [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
|
14
|
Snipstad S, Mørch Ý, Sulheim E, Åslund A, Pedersen A, Davies CDL, Hansen R, Berg S. Sonopermeation Enhances Uptake and Therapeutic Effect of Free and Encapsulated Cabazitaxel. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1319-1333. [PMID: 33549379 DOI: 10.1016/j.ultrasmedbio.2020.12.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/18/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Delivery of drugs and nanomedicines to tumors is often heterogeneous and insufficient and, thus, of limited efficacy. Microbubbles in combination with ultrasound have been found to improve delivery to tumors, enhancing accumulation and penetration. We used a subcutaneous prostate cancer xenograft model in mice to investigate the effect of free and nanoparticle-encapsulated cabazitaxel in combination with ultrasound and microbubbles with a lipid shell or a shell of nanoparticles. Sonopermeation reduced tumor growth and prolonged survival (26%-100%), whether the free drug was co-injected with lipid-shelled microbubbles or the nanoformulation was co-injected with lipid-shelled or nanoparticle-shelled microbubbles. Coherently with the improved therapeutic response, we found enhanced uptake of nanoparticles directly after ultrasound treatment that lasted several weeks (2.3 × -15.8 × increase). Neither cavitation dose nor total accumulation of nanoparticles could explain the variation within treatment groups, emphasizing the need for a better understanding of the tumor biology and mechanisms involved in ultrasound-mediated treatment.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Andreas Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - André Pedersen
- Department of Health Research, SINTEF Digital, Trondheim, Norway
| | | | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical imaging, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
15
|
Afadzi M, Myhre OF, Yemane PT, Bjorkoy A, Torp SH, van Wamel A, Lelu S, Angelsen BAJ, de Lange Davies C. Effect of Acoustic Radiation Force on the Distribution of Nanoparticles in Solid Tumors. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:432-445. [PMID: 32986550 DOI: 10.1109/tuffc.2020.3027072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Acoustic radiation force (ARF) might improve the distribution of nanoparticles (NPs) in tumors. To study this, tumors growing subcutaneously in mice were exposed to focused ultrasound (FUS) either 15 min or 4 h after the injection of NPs, to investigate the effect of ARF on the transport of NPs across the vessel wall and through the extracellular matrix. Quantitative analysis of confocal microscopy images from frozen tumor sections was performed to estimate the displacement of NPs from blood vessels. Using the same experimental exposure parameters, ARF was simulated and compared with the experimental data. Enhanced interstitial transport of NPs in tumor tissues was observed when FUS (10 MHz, acoustic power 234 W/cm2, 3.3% duty cycle) was given either 15 min or 4 h after NP administration. According to acoustic simulations, the FUS generated an ARF per unit volume of 2.0×106 N/m3. The displacement of NPs was larger when FUS was applied 4 h after NP injection compared with after 15 min. This study shows that ARF might contribute to a modest improved distribution of NPs into the tumor interstitium.
Collapse
|
16
|
Bjånes TK, Riedel B, Schjøtt J. Ultrasound and microbubble assisted drug delivery - A clinical pharmacological perspective. Pharmacol Res 2021; 165:105475. [PMID: 33524538 DOI: 10.1016/j.phrs.2021.105475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Tormod K Bjånes
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Bettina Riedel
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Jan Schjøtt
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
17
|
Sun T, Dasgupta A, Zhao Z, Nurunnabi M, Mitragotri S. Physical triggering strategies for drug delivery. Adv Drug Deliv Rev 2020; 158:36-62. [PMID: 32589905 DOI: 10.1016/j.addr.2020.06.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Physically triggered systems hold promise for improving drug delivery by enhancing the controllability of drug accumulation and release, lowering non-specific toxicity, and facilitating clinical translation. Several external physical stimuli including ultrasound, light, electric fields and magnetic fields have been used to control drug delivery and they share some common features such as spatial targeting, spatiotemporal control, and minimal invasiveness. At the same time, they possess several distinctive features in terms of interactions with biological entities and/or the extent of stimulus response. Here, we review the key advances of such systems with a focus on discussing their physical mechanisms, the design rationales, and translational challenges.
Collapse
Affiliation(s)
- Tao Sun
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anshuman Dasgupta
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, TX 79902, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Bush N, Healey A, Shah A, Box G, Kirkin V, Eccles S, Sontum PC, Kotopoulis S, Kvåle S, van Wamel A, Davies CDL, Bamber J. Theranostic Attributes of Acoustic Cluster Therapy and Its Use for Enhancing the Effectiveness of Liposomal Doxorubicin Treatment of Human Triple Negative Breast Cancer in Mice. Front Pharmacol 2020; 11:75. [PMID: 32153400 PMCID: PMC7044119 DOI: 10.3389/fphar.2020.00075] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Acoustic cluster therapy (ACT) comprises co-administration of a formulation containing microbubble/microdroplet clusters (PS101), together with a regular medicinal drug (e.g., a chemotherapeutic) and local ultrasound (US) insonation of the targeted pathological tissue (e.g., the tumor). PS101 is confined to the vascular compartment and, when the clusters are exposed to regular diagnostic imaging US fields, the microdroplets undergo a phase-shift to produce bubbles with a median diameter of 22 µm when unconstrained by the capillary wall. In vivo these bubbles transiently lodge in the tumor's microvasculature. Low frequency ultrasound (300 kHz) at a low mechanical index (MI = 0.15) is then applied to drive oscillations of the deposited ACT bubbles to induce a range of biomechanical effects that locally enhance extravasation, distribution, and uptake of the co-administered drug, significantly increasing its therapeutic efficacy. METHODS In this study we investigated the therapeutic efficacy of ACT with liposomal doxorubicin for the treatment of triple negative breast cancer using orthotopic human tumor xenografts (MDA-MB-231-H.luc) in athymic mice (ICR-NCr-Foxn1nu). Doxil® (6 mg/kg, i.v.) was administered at days 0 and 21, each time immediately followed by three sequential ACT (20 ml/kg PS101) treatment procedures (n = 7-10). B-mode and nonlinear ultrasound images acquired during the activation phase were correlated to the therapeutic efficacy. RESULTS Results show that combination with ACT induces a strong increase in the therapeutic efficacy of Doxil®, with 63% of animals in complete, stable remission at end of study, vs. 10% for Doxil® alone (p < 0.02). A significant positive correlation (p < 0.004) was found between B-mode contrast enhancement during ACT activation and therapy response. These observations indicate that ACT may also be used as a theranostic agent and that ultrasound contrast enhancement during or before ACT treatment may be employed as a biomarker of therapeutic response during clinical use.
Collapse
Affiliation(s)
- Nigel Bush
- Joint Department of Physics, Institute of Cancer Research, London, United Kingdom
| | | | - Anant Shah
- Joint Department of Physics, Institute of Cancer Research, London, United Kingdom
| | - Gary Box
- Department of Physics, CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Vladimir Kirkin
- Department of Physics, CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Sue Eccles
- Department of Physics, CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | | | | | | | | | | | - Jeffrey Bamber
- Joint Department of Physics, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
19
|
Bush N, Healey A, Shah A, Box G, Kirkin V, Kotopoulis S, Kvåle S, Sontum PC, Bamber J. Therapeutic Dose Response of Acoustic Cluster Therapy in Combination With Irinotecan for the Treatment of Human Colon Cancer in Mice. Front Pharmacol 2019; 10:1299. [PMID: 31803049 PMCID: PMC6877694 DOI: 10.3389/fphar.2019.01299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction: Acoustic Cluster Therapy (ACT) comprises coadministration of a formulation containing microbubble-microdroplet clusters (PS101) together with a regular medicinal drug and local ultrasound (US) insonation of the targeted pathological tissue. PS101 is confined to the vascular compartment and when the clusters are exposed to regular diagnostic imaging US fields, the microdroplets undergo a phase shift to produce bubbles with a median diameter of 22 µm. Low frequency, low mechanical index US is then applied to drive oscillations of the deposited ACT bubbles to induce biomechanical effects that locally enhance extravasation, distribution, and uptake of the coadministered drug, significantly increasing its therapeutic efficacy. Methods: The therapeutic efficacy of ACT with irinotecan (60 mg/kg i.p.) was investigated using three treatment sessions given on day 0, 7, and 14 on subcutaneous human colorectal adenocarcinoma xenografts in mice. Treatment was performed with three back-to-back PS101+US administrations per session with PS101 doses ranging from 0.40-2.00 ml PS101/kg body weight (n = 8-15). To induce the phase shift, 45 s of US at 8 MHz at an MI of 0.30 was applied using a diagnostic US system; low frequency exposure consisted of 1 or 5 min at 500 kHz with an MI of 0.20. Results: ACT with irinotecan induced a strong, dose dependent increase in the therapeutic effect (R2 = 0.95). When compared to irinotecan alone, at the highest dose investigated, combination treatment induced a reduction in average normalized tumour volume from 14.6 (irinotecan), to 5.4 (ACT with irinotecan, p = 0.002) on day 27. Median survival increased from 34 days (irinotecan) to 54 (ACT with irinotecan, p = 0.002). Additionally, ACT with irinotecan induced an increase in the fraction of complete responders; from 7% to 26%. There was no significant difference in the therapeutic efficacy whether the low frequency US lasted 1 or 5 min. Furthermore, there was no significant difference between the enhancement observed in the efficacy of ACT with irinotecan when PS101+US was administered before or after irinotecan. An increase in early dropouts was observed at higher PS101 doses. Both mean tumour volume (on day 27) and median survival indicate that the PS101 dose response was linear in the range investigated.
Collapse
Affiliation(s)
- Nigel Bush
- Joint Department of Physics, Institute of Cancer Research, London, United Kingdom
| | - Andrew Healey
- CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Anant Shah
- Joint Department of Physics, Institute of Cancer Research, London, United Kingdom
| | - Gary Box
- CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Vladimir Kirkin
- CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | | | | | | | - Jeffrey Bamber
- Joint Department of Physics, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
20
|
Xia Y, Na X, Wu J, Ma G. The Horizon of the Emulsion Particulate Strategy: Engineering Hollow Particles for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1801159. [PMID: 30260511 DOI: 10.1002/adma.201801159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/06/2018] [Indexed: 05/13/2023]
Abstract
With their hierarchical structures and the substantial surface areas, hollow particles have gained immense research interest in biomedical applications. For scalable fabrications, emulsion-based approaches have emerged as facile and versatile strategies. Here, the recent achievements in this field are unfolded via an "emulsion particulate strategy," which addresses the inherent relationship between the process control and the bioactive structures. As such, the interior architectures are manipulated by harnessing the intermediate state during the emulsion revolution (intrinsic strategy), whereas the external structures are dictated by tailoring the building blocks and solidification procedures of the Pickering emulsion (extrinsic strategy). Through integration of the intrinsic and extrinsic emulsion particulate strategy, multifunctional hollow particles demonstrate marked momentum for label-free multiplex detections, stimuli-responsive therapies, and stem cell therapies.
Collapse
Affiliation(s)
- Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xiangming Na
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- PLA Key Laboratory of Biopharmaceutical Production & Formulation Engineering Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Jiangsu National Synergetic Innovation Center for Advanced Materials, Nanjing, 211816, P. R. China
| |
Collapse
|
21
|
Andersen KK, Healey A, Bush NL, Frijlink ME, Hoff L. A Harmonic Dual-Frequency Transducer for Acoustic Cluster Therapy. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2381-2390. [PMID: 31230911 DOI: 10.1016/j.ultrasmedbio.2019.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/22/2019] [Accepted: 04/07/2019] [Indexed: 06/09/2023]
Abstract
Acoustic Cluster Therapy (ACT) is a two-component formulation of commercially available microbubbles (Sonazoid; GE Healthcare, Oslo, Norway) and microdroplets (perfluorated oil) currently under development for cancer treatment. The microbubbles and microdroplets have opposite surface charges to form microbubble/microdroplet clusters, which are administered to patients together with a drug. When the clusters and drug reach the target tumour, two ultrasound (US) exposure regimes are used: First, high-frequency (>2.0 MHz) US evaporates the oil and forms ACT bubbles that lodge at the microvascular level. Second, low-frequency (0.5 MHz) US induces stable mechanical oscillations of the ACT bubbles, causing localized micro-streaming, radiation and shear forces that increase the uptake of the drugs to the target tumour. This report describes the design and testing of a dual-frequency transducer and a laboratory setup for pre-clinical in vivo studies of ACT on murine tumour models. The dual-frequency transducer utilizes the 5th harmonic (2.7 MHz) and fundamental (0.5 MHz) of a single piezoceramic disk for the high-frequency and low-frequency regimes, respectively. Two different aperture radii are used to align the high-frequency and low-frequency beam maxima, and the high-frequency -3 dB beam width diameter is 6 mm, corresponding to the largest tumour sizes we expect to treat. The low-frequency -3 dB beam width extends 6 mm. Although unconventional, the 5th harmonic exhibit a 44% efficiency and can therefore be used for transmission of acoustic energy. Moreover, both in vitro and in vivo measurements demonstrate that the 5th harmonic can be used to evaporate the microbubble/microdroplet clusters. For the in vivo measurements, we used the kidneys of non-tumour-bearing mice as tumour surrogates. Based on this, the transducer is deemed suited for pre-clinical in vivo studies of ACT and replaces a cumbersome test setup consisting of two transducers.
Collapse
Affiliation(s)
| | | | - Nigel L Bush
- Joint Department of Physics, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | | - Lars Hoff
- University of South-Eastern Norway, Horten, Norway
| |
Collapse
|
22
|
Roovers S, Segers T, Lajoinie G, Deprez J, Versluis M, De Smedt SC, Lentacker I. The Role of Ultrasound-Driven Microbubble Dynamics in Drug Delivery: From Microbubble Fundamentals to Clinical Translation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10173-10191. [PMID: 30653325 DOI: 10.1021/acs.langmuir.8b03779] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In the last couple of decades, ultrasound-driven microbubbles have proven excellent candidates for local drug delivery applications. Besides being useful drug carriers, microbubbles have demonstrated the ability to enhance cell and tissue permeability and, as a consequence, drug uptake herein. Notwithstanding the large amount of evidence for their therapeutic efficacy, open issues remain. Because of the vast number of ultrasound- and microbubble-related parameters that can be altered and the variability in different models, the translation from basic research to (pre)clinical studies has been hindered. This review aims at connecting the knowledge gained from fundamental microbubble studies to the therapeutic efficacy seen in in vitro and in vivo studies, with an emphasis on a better understanding of the response of a microbubble upon exposure to ultrasound and its interaction with cells and tissues. More specifically, we address the acoustic settings and microbubble-related parameters (i.e., bubble size and physicochemistry of the bubble shell) that play a key role in microbubble-cell interactions and in the associated therapeutic outcome. Additionally, new techniques that may provide additional control over the treatment, such as monodisperse microbubble formulations, tunable ultrasound scanners, and cavitation detection techniques, are discussed. An in-depth understanding of the aspects presented in this work could eventually lead the way to more efficient and tailored microbubble-assisted ultrasound therapy in the future.
Collapse
Affiliation(s)
- Silke Roovers
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| | - Tim Segers
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center , University of Twente , P.O. Box 217, 7500 AE Enschede , The Netherlands
| | - Guillaume Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center , University of Twente , P.O. Box 217, 7500 AE Enschede , The Netherlands
| | - Joke Deprez
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| | - Michel Versluis
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center , University of Twente , P.O. Box 217, 7500 AE Enschede , The Netherlands
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| | - Ine Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Faculty of Pharmaceutical Sciences , Ghent University , Ottergemsesteenweg 460 , Ghent , Belgium
| |
Collapse
|
23
|
Abumanhal-Masarweh H, Koren L, Zinger A, Yaari Z, Krinsky N, Kaneti G, Dahan N, Lupu-Haber Y, Suss-Toby E, Weiss-Messer E, Schlesinger-Laufer M, Shainsky-Roitman J, Schroeder A. Sodium bicarbonate nanoparticles modulate the tumor pH and enhance the cellular uptake of doxorubicin. J Control Release 2019; 296:1-13. [PMID: 30615983 DOI: 10.1016/j.jconrel.2019.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 12/22/2022]
Abstract
Acidic pH in the tumor microenvironment is associated with cancer metabolism and creates a physiological barrier that prevents from drugs to penetrate cells. Specifically, ionizable weak-base drugs, such as doxorubicin, freely permeate membranes in their uncharged form, however, in the acidic tumor microenvironment these drugs become charged and their cellular permeability is retarded. In this study, 100-nm liposomes loaded with sodium bicarbonate were used as adjuvants to elevate the tumor pH. Combined treatment of triple-negative breast cancer cells (4T1) with doxorubicin and sodium-bicarbonate enhanced drug uptake and increased its anti-cancer activity. In vivo, mice bearing orthotropic 4T1 breast cancer tumors were administered either liposomal or free bicarbonate intravenously. 3.7 ± 0.3% of the injected liposomal dose was detected in the tumor after twenty-four hours, compared to 0.17% ± 0.04% in the group injected free non-liposomal bicarbonate, a 21-fold increase. Analyzing nanoparticle biodistribution within the tumor tissue revealed that 93% of the PEGylated liposomes accumulated in the extracellular matrix, while 7% were detected intracellularly. Mice administered bicarbonate-loaded liposomes reached an intra-tumor pH value of 7.38 ± 0.04. Treating tumors with liposomal bicarbonate combined with a sub-therapeutic dose of doxorubicin achieved an improved therapeutic outcome, compared to mice treated with doxorubicin or bicarbonate alone. Interestingly, analysis of the tumor microenvironment demonstrated an increase in immune cell' population (T-cell, B-cell and macrophages) in tumors treated with liposomal bicarbonate. This study demonstrates that targeting metabolic adjuvants with nanoparticles to the tumor microenvironment can enhance anticancer drug activity and improve treatment.
Collapse
Affiliation(s)
- Hanan Abumanhal-Masarweh
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel; Russell Berrie Nanotechnology Institute, The Norman Seiden Multidisciplinary Graduate Program, Technion - Israel Institute of Technology, Haifa 3200, Israel
| | - Lilach Koren
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Assaf Zinger
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Zvi Yaari
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Nitzan Krinsky
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel; The Interdisciplinary Program for Biotechnology, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Galoz Kaneti
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Nitsan Dahan
- Life Sciences and Engineering Infrastructure Center, Lorry I. Lokey Interdisciplinary Center, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Yael Lupu-Haber
- Life Sciences and Engineering Infrastructure Center, Lorry I. Lokey Interdisciplinary Center, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Edith Suss-Toby
- Bioimging Center, Biomedical Core Facility, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Esther Weiss-Messer
- Bioimging Center, Biomedical Core Facility, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Michal Schlesinger-Laufer
- The Pre-Clinical Research Authority Unit, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Janna Shainsky-Roitman
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Avi Schroeder
- Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
24
|
Snipstad S, Sulheim E, de Lange Davies C, Moonen C, Storm G, Kiessling F, Schmid R, Lammers T. Sonopermeation to improve drug delivery to tumors: from fundamental understanding to clinical translation. Expert Opin Drug Deliv 2018; 15:1249-1261. [PMID: 30415585 DOI: 10.1080/17425247.2018.1547279] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Ultrasound in combination with microbubbles can make cells and tissues more accessible for drugs, thereby achieving improved therapeutic outcomes. In this review, we introduce the term 'sonopermeation', covering mechanisms such as pore formation (traditional sonoporation), as well as the opening of intercellular junctions, stimulated endocytosis/transcytosis, improved blood vessel perfusion and changes in the (tumor) microenvironment. Sonopermeation has gained a lot of interest in recent years, especially for delivering drugs through the otherwise impermeable blood-brain barrier, but also to tumors. AREAS COVERED In this review, we summarize various in vitro assays and in vivo setups that have been employed to unravel the fundamental mechanisms involved in ultrasound-enhanced drug delivery, as well as clinical trials that are ongoing in patients with brain, pancreatic, liver and breast cancer. We summarize the basic principles of sonopermeation, describe recent findings obtained in (pre-) clinical trials, and discuss future directions. EXPERT OPINION We suggest that an improved mechanistic understanding, and microbubbles and ultrasound equipment specialized for drug delivery (and not for imaging) are key aspects to create more effective treatment regimens by sonopermeation. Real-time feedback and tools to predict therapeutic outcome and which tumors/patients will benefit from sonopermeation-based interventions will be important to promote clinical translation.
Collapse
Affiliation(s)
- Sofie Snipstad
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Einar Sulheim
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Catharina de Lange Davies
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway
| | - Chrit Moonen
- d Imaging Division , University Medical Center , Utrecht , The Netherlands
| | - Gert Storm
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands
| | - Fabian Kiessling
- g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| | - Ruth Schmid
- b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway
| | - Twan Lammers
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands.,g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
25
|
A versatile and robust microfluidic device for capillary-sized simple or multiple emulsions production. Biomed Microdevices 2018; 20:94. [DOI: 10.1007/s10544-018-0340-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
26
|
|
27
|
Huang FY, Lei J, Sun Y, Yan F, Chen B, Zhang L, Lu Z, Cao R, Lin YY, Wang CC, Tan GH. Induction of enhanced immunogenic cell death through ultrasound-controlled release of doxorubicin by liposome-microbubble complexes. Oncoimmunology 2018; 7:e1446720. [PMID: 29900064 DOI: 10.1080/2162402x.2018.1446720] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 02/07/2023] Open
Abstract
Immunogenic cell death (ICD) is a specific kind of cell death that stimulates the immune system to combat cancer cells. Ultrasound (US)-controlled targeted release of drugs by liposome-microbubble complexes is a promising approach due to its non-invasive nature and visibility through ultrasound imaging. However, it is not known whether this approach can enhance ICD induced by drugs, such as doxorubicin. Herein, we prepared a doxorubicin-liposome-microbubble complex (MbDox), and the resultant MbDox was then characterized and tested for US-controlled release of Dox (MbDox+US treatment) to enhance the induction of ICD in LL/2 and CT26 cancer cells and in syngeneic murine models. We found that MbDox+US treatment caused more cellular uptake and nuclear accumulation of Dox in tumor cells, and more accumulation of Dox in tumor tissues. Enhanced induction of ICD occurred both in vitro and in vivo. MbDox+US treatment induced more apoptosis, stronger membrane exposure and the release of ER stress proteins and DAMPs in tumor cells, and increased DC maturation in vitro. In addition, MbDox+US treatment also resulted in stronger therapeutic effects in immunocompetent mice than in immunodeficient mice. Moreover, MbDox+US enhancement of ICD was also evidenced by a higher proportion of activated CD8+ T-lymphocytes but lower Treg in tumor tissues. Taken together, our results demonstrate that US-controlled release of ICD inducers into nuclei using liposome-microbubble complexes may be an effective approach to enhance the induction of ICD for tumor treatment.
Collapse
Affiliation(s)
- Feng-Ying Huang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Jing Lei
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China.,Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical College, Haikou , China
| | - Yan Sun
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Fei Yan
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Bin Chen
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Liming Zhang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Zhuoxuan Lu
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Rong Cao
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Ying-Ying Lin
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| | - Cai-Chun Wang
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China.,Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical College, Haikou , China
| | - Guang-Hong Tan
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou , China
| |
Collapse
|
28
|
Åslund AK, Snipstad S, Healey A, Kvåle S, Torp SH, Sontum PC, Davies CDL, van Wamel A. Efficient Enhancement of Blood-Brain Barrier Permeability Using Acoustic Cluster Therapy (ACT). Theranostics 2017; 7:23-30. [PMID: 28042313 PMCID: PMC5196882 DOI: 10.7150/thno.16577] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/23/2016] [Indexed: 12/27/2022] Open
Abstract
The blood-brain barrier (BBB) is a major obstacle in drug delivery for diseases of the brain, and today there is no standardized route to surpass it. One technique to locally and transiently disrupt the BBB, is focused ultrasound in combination with gas-filled microbubbles. However, the microbubbles used are typically developed for ultrasound imaging, not BBB disruption. Here we describe efficient opening of the BBB using the promising novel Acoustic Cluster Therapy (ACT), that recently has been used in combination with Abraxane® to successfully treat subcutaneous tumors of human prostate adenocarcinoma in mice. ACT is based on the conjugation of microbubbles to liquid oil microdroplets through electrostatic interactions. Upon activation in an ultrasound field, the microdroplet phase transfers to form a larger bubble that transiently lodges in the microvasculature. Further insonation induces volume oscillations of the activated bubble, which in turn induce biomechanical effects that increase the permeability of the BBB. ACT was able to safely and temporarily permeabilize the BBB, using an acoustic power 5-10 times lower than applied for conventional microbubbles, and successfully deliver small and large molecules into the brain.
Collapse
|
29
|
Lelu S, Afadzi M, Berg S, Aslund AKO, Torp SH, Sattler W, de L Davies C. Primary Porcine Brain Endothelial Cells as In Vitro Model to Study Effects of Ultrasound and Microbubbles on Blood-Brain Barrier Function. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2017; 64:281-290. [PMID: 27529871 DOI: 10.1109/tuffc.2016.2597004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Focused ultrasound (FUS) in the presence of microbubbles transiently and reversibly opens the blood-brain barrier (BBB) in rodents and humans, thereby providing a time window for increased drug delivery into brain tissue. To get insight into the underlying mechanisms that govern ultrasound (US)-mediated opening of the BBB, in vitro models are a useful alternative. In this paper, we have utilized an in vitro BBB model that consists of primary porcine brain endothelial cells (PBECs). PBEC monolayers are grown on permeable membranes, which allow assessment of key features of BBB function as well as US treatment. This experimental model is characterized by low permeability for both small molecules and proteins, has a high transendothelial electrical resistance, and expresses tight junctions and efflux pumps. Here, we compare the effects of inertial and stable cavitation in the presence of SonoVue microbubbles on PBEC monolayers' electrical resistance and permeability properties. Our results point out the fragility of PBEC monolayers, which enhances results variability. In particular, we show that handling of the inserts, such as medium change and transfer to the US setup, modifies the cellular response, and immunostaining of the monolayers introduces damage and cell detachment within the US-exposed monolayers. Our results indicate that stable cavitation might have a more pronounced impact on cell permeability as compared with inertial cavitation in vitro. This paper might contribute to further development of experimental setups that are suitable to characterize the impact of FUS and microbubbles on BBB properties in vitro.
Collapse
|
30
|
Park K. Ultrasound and microbubble enhanced treatment of inoperable pancreatic cancer. J Control Release 2016; 243:381. [PMID: 28024837 DOI: 10.1016/j.jconrel.2016.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Kinam Park
- Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN 47907, USA.
| |
Collapse
|
31
|
Sonoporation with Acoustic Cluster Therapy (ACT®) induces transient tumour volume reduction in a subcutaneous xenograft model of pancreatic ductal adenocarcinoma. J Control Release 2016; 245:70-80. [PMID: 27871988 DOI: 10.1016/j.jconrel.2016.11.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/08/2016] [Accepted: 11/17/2016] [Indexed: 12/31/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest cancers with survival averaging only 3months if untreated following diagnosis. A major limitation in effectively treating PDAC using conventional and targeted chemotherapeutic agents, is inadequate drug delivery to the target location, predominantly due to a poorly vascularised, desmoplastic tumour microenvironment. Ultrasound in combination with ultrasound contrast agents, i.e., microbubbles, that flow through the vasculature and capillaries can be used to disrupt such mechanical barriers, potentially allowing for a greater therapeutic efficacy. This phenomenon is commonly referred to as sonoporation. In an attempt to improve the efficacy of sonoporation, novel microbubble formulations are being developed to address the limitation of commercially produced clinical diagnostic ultrasound contrast agents. In our work here we evaluate the ability of a novel formulation; namely Acoustic Cluster Therapy (ACT®) to improve the therapeutic efficacy of the chemotherapeutic agent paclitaxel, longitudinally in a xenograft model of PDAC. Results indicated that ACT® bubbles alone demonstrated no observable toxic effects, whilst ACT® in combination with paclitaxel can transiently reduce tumour volumes significantly, three days posttreatment (p=0.0347-0.0458). Quantitative 3D ultrasound validated the calliper measurements. Power Doppler ultrasound imaging indicated that ACT® in combination with paclitaxel was able to transiently sustain peak vasculature percentages as observed in the initial stages of tumour development. Nevertheless, there was no significant difference in tumour vasculature percentage at the end of treatment. The high vascular percentage correlated to the transient decrease and overall inhibition of the tumour volumes. In conclusion, ACT® improves the therapeutic efficacy of paclitaxel in a PDAC xenograft model allowing for transient tumour volume reduction and sustained tumour vasculature percentage.
Collapse
|
32
|
Myhre O, Bjørgan M, Grant D, Hustvedt SO, Sontum PC, Dirven H. Safety assessment in rats and dogs of Acoustic Cluster Therapy, a novel concept for ultrasound mediated, targeted drug delivery. Pharmacol Res Perspect 2016; 4:e00274. [PMID: 28097007 PMCID: PMC5226284 DOI: 10.1002/prp2.274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 10/10/2016] [Indexed: 01/29/2023] Open
Abstract
Acoustic Cluster Therapy (ACT) represents a novel concept for targeted drug delivery. Ultrasound is applied to activate intravenously administered free-flowing clusters of microbubbles and microdroplets within the target pathology, depositing 20-30 μm large bubbles in the microvasculature for 5-10 min. Further application of ultrasound induces biomechanical effects which increase vascular permeability and enhance localized extravasation of coadministered drugs. Herein we report investigations done to assess the preclinical safety of ACT, using doses up to 1 mL/kg (3 μL perfluoromethyl-cyclopentane/kg). In dogs, half the animals were exposed to ultrasound activation in the heart for 1 min, no ultrasound was applied in the other half. Posttreatment observation time was 24 h. Clinical signs, ophthalmoscopy, clinical pathology, macro-, and microscopy were used as endpoints. No differences between groups with and without ultrasound activation were observed. Short-lasting leukopenia and thrombocytopenia, possibly secondary to a slight and short-lasting increase in plasma histamine and complement split products, were the only effects noted. In rats ACT was activated in the liver for 5 min. Histopathology and clinical chemistry parameters remained unchanged. Lastly, rats were treated with ACT activated in the heart and thereafter placed on a rotarod for evaluation of motor coordination. No differences were observed between animals treated with ACT and controls. In conclusion, ACT appeared safe at dose-levels up to 1 mL/kg and with activation either in the heart or the liver. These results, together with positive efficacy data upon coinjection with cytotoxic compounds encourage further preclinical safety studies with the objective of entering subsequent clinical trials.
Collapse
Affiliation(s)
- Oddvar Myhre
- Norwegian Institute of Public Health Oslo Norway
| | | | | | | | | | | |
Collapse
|
33
|
van Wamel A, Sontum PC, Healey A, Kvåle S, Bush N, Bamber J, de Lange Davies C. Acoustic Cluster Therapy (ACT) enhances the therapeutic efficacy of paclitaxel and Abraxane® for treatment of human prostate adenocarcinoma in mice. J Control Release 2016; 236:15-21. [PMID: 27297780 DOI: 10.1016/j.jconrel.2016.06.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 01/05/2023]
Abstract
Acoustic cluster therapy (ACT) is a novel approach for ultrasound mediated, targeted drug delivery. In the current study, we have investigated ACT in combination with paclitaxel and Abraxane® for treatment of a subcutaneous human prostate adenocarcinoma (PC3) in mice. In combination with paclitaxel (12mg/kg given i.p.), ACT induced a strong increase in therapeutic efficacy; 120days after study start, 42% of the animals were in stable, complete remission vs. 0% for the paclitaxel only group and the median survival was increased by 86%. In combination with Abraxane® (12mg paclitaxel/kg given i.v.), ACT induced a strong increase in the therapeutic efficacy; 60days after study start 100% of the animals were in stable, remission vs. 0% for the Abraxane® only group, 120days after study start 67% of the animals were in stable, complete remission vs. 0% for the Abraxane® only group. For the ACT+Abraxane group 100% of the animals were alive after 120days vs. 0% for the Abraxane® only group. Proof of concept for Acoustic Cluster Therapy has been demonstrated; ACT markedly increases the therapeutic efficacy of both paclitaxel and Abraxane® for treatment of human prostate adenocarcinoma in mice.
Collapse
Affiliation(s)
- Annemieke van Wamel
- Dept. of Physics, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.
| | | | | | | | - Nigel Bush
- Joint Dept. of Physics, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | - Jeffrey Bamber
- Joint Dept. of Physics, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | | |
Collapse
|
34
|
Acoustic Cluster Therapy (ACT)--A novel concept for ultrasound mediated, targeted drug delivery. Int J Pharm 2015; 495:1019-27. [PMID: 26408933 DOI: 10.1016/j.ijpharm.2015.09.047] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/16/2015] [Accepted: 09/19/2015] [Indexed: 01/29/2023]
Abstract
A novel approach for ultrasound (US) mediated drug delivery - Acoustic Cluster Therapy (ACT) - is proposed, and basic characteristics of the ACT formulation are elucidated. The concept comprises administration of free flowing clusters of negatively charged microbubbles and positively charged microdroplets. The clusters are activated within the target pathology by diagnostic US, undergo an ensuing liquid-to-gas phase shift and transiently deposit 20-30 μm large bubbles in the microvasculature, occluding blood flow for ∼5-10 min. Further application of US will induce biomechanical effects that increases the vascular permeability, leading to a locally enhanced extravasation of components from the vascular compartment (e.g. released or co-administered drugs). Methodologies are detailed for determination of vital in-vitro characteristics of the ACT compound; cluster concentration and size distribution. It is shown how these attributes can be engineered through various formulation parameters, and their significance as predictors of biological behaviour, such as deposit characteristics, is demonstrated by US imaging in a dog model. Furthermore, in-vivo properties of the activated ACT bubbles are studied by intravital microscopy in a rat model, confirming the postulated behaviour of the concept.
Collapse
|