1
|
Baig MA, Du Y, Zan Z, Fan Z. Influence of cell shape on sonoporation efficiency in microbubble-facilitated delivery using micropatterned cell arrays. Sci Rep 2024; 14:30845. [PMID: 39730459 DOI: 10.1038/s41598-024-81410-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Microbubble-facilitated sonoporation is a rapid, versatile, and non-viral intracellular delivery technique with potential for clinical and ex vivo cell engineering applications. We developed a micropatterning-based approach to investigate the impact of cell shape on sonoporation efficacy. Cationic microbubbles were employed to enhance sonoporation by binding to the cell membrane electrostatically. NIH/3T3 fibroblasts were micropatterned into circle, square, triangle, and rectangle. A two-plate system ensured high-throughput and efficient sonoporation by controlling cationic microbubble-cell attachment. High-speed video microscopy captured the acoustic dynamics of microbubbles under short ultrasound pulses. Our findings reveal that for NIH/3T3 fibroblasts, rectangular cells achieved the highest sonoporation and survival rate, while square-shaped cells demonstrated the greatest propidium iodide uptake. Triangle-shaped NIH/3T3 fibroblasts displayed an initial rise then a plateau in the sonoporation and survival rate as the ultrasound pulse duration increased from 10 cycles to 100 cycles, and then to 200 cycles. Conversely, rectangle-shaped cells showed a decrease followed by a stabilization. Circle-shaped and rectangle-shaped HeLa cells exhibited similar sonoporation outcomes, which were not as effective as NIH/3T3 fibroblasts. This study underscores the significance of cell shape in optimizing sonoporation efficiency and highlights the potential of combining micropatterning with controlled targeting sonoporation to advance intracellular delivery technologies.
Collapse
Affiliation(s)
- Mirza Albash Baig
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China
| | - Yanyao Du
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China
| | - Zhaoguang Zan
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China
| | - Zhenzhen Fan
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
2
|
Fernandes DA. Comprehensive Review on Bubbles: Synthesis, Modification, Characterization and Biomedical Applications. Bioconjug Chem 2024; 35:1639-1686. [PMID: 39377727 DOI: 10.1021/acs.bioconjchem.4c00137] [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: 10/09/2024]
Abstract
Accurate detection, treatment, and imaging of diseases are important for effective treatment outcomes in patients. In this regard, bubbles have gained much attention, due to their versatility. Bubbles usually 1 nm to 10 μm in size can be produced and loaded with a variety of lipids, polymers, proteins, and therapeutic and imaging agents. This review details the different production and loading methods for bubbles, for imaging and treatment of diseases/conditions such as cancer, tumor angiogenesis, thrombosis, and inflammation. Bubbles can also be used for perfusion measurements, important for diagnostic and therapeutic decision making in cardiac disease. The different factors important in the stability of bubbles and the different techniques for characterizing their physical and chemical properties are explained, for developing bubbles with advanced therapeutic and imaging features. Hence, the review provides important insights for researchers studying bubbles for biomedical applications.
Collapse
|
3
|
Przystupski D, Baczyńska D, Rossowska J, Kulbacka J, Ussowicz M. Calcium ion delivery by microbubble-assisted sonoporation stimulates cell death in human gastrointestinal cancer cells. Biomed Pharmacother 2024; 179:117339. [PMID: 39216448 DOI: 10.1016/j.biopha.2024.117339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Ultrasound-mediated cell membrane permeabilization - sonoporation, enhances drug delivery directly to tumor sites while reducing systemic side effects. The potential of ultrasound to augment intracellular calcium uptake - a critical regulator of cell death and proliferation - offers innovative alternative to conventional chemotherapy. However, calcium therapeutic applications remain underexplored in sonoporation studies. This research provides a comprehensive analysis of calcium sonoporation (CaSP), which combines ultrasound treatment with calcium ions and SonoVue microbubbles, on gastrointestinal cancer cells LoVo and HPAF-II. Initially, optimal sonoporation parameters were determined: an acoustic wave of 1 MHz frequency with a 50 % duty cycle at intensity of 2 W/cm2. Subsequently, various cellular bioeffects, such as viability, oxidative stress, metabolism, mitochondrial function, proliferation, and cell death, were assessed following CaSP treatment. CaSP significantly impaired cancer cell function by inducing oxidative and metabolic stress, evidenced by increased mitochondrial depolarization, decreased ATP levels, and elevated glucose uptake in a Ca2+ dose-dependent manner, leading to activation of the intrinsic apoptotic pathway. Cellular response to CaSP depended on the TP53 gene's mutational status: colon cancer cells were more susceptible to CaSP-induced apoptosis and G1 phase cell cycle arrest, whereas pancreatic cancer cells showed a higher necrotic response and G2 cell cycle arrest. These promising results encourage future research to optimize sonoporation parameters for clinical use, investigate synergistic effects with existing treatments, and assess long-term safety and efficacy in vivo. Our study highlights CaSP's clinical potential for improved safety and efficacy in cancer therapy, offering significant implications for the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- Dawid Przystupski
- Department of Paediatric Bone Marrow Transplantation, Oncology and Haematology, Wroclaw Medical University, Borowska 213, Wroclaw 50-556, Poland.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, Wroclaw 50-556, Poland
| | - Joanna Rossowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, Wroclaw 53-114, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, Wroclaw 50-556, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių 5, Vilnius 08410, Lithuania
| | - Marek Ussowicz
- Department of Paediatric Bone Marrow Transplantation, Oncology and Haematology, Wroclaw Medical University, Borowska 213, Wroclaw 50-556, Poland
| |
Collapse
|
4
|
Sharma D, Petchiny TN, Czarnota GJ. A Promising Therapeutic Strategy of Combining Acoustically Stimulated Nanobubbles and Existing Cancer Treatments. Cancers (Basel) 2024; 16:3181. [PMID: 39335153 PMCID: PMC11431001 DOI: 10.3390/cancers16183181] [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: 07/19/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
In recent years, ultrasound-stimulated microbubbles (USMBs) have gained great attention because of their wide theranostic applications. However, due to their micro-size, reaching the targeted site remains a challenge. At present, ultrasound-stimulated nanobubbles (USNBs) have attracted particular interest, and their small size allows them to extravasate easily in the blood vessels penetrating deeper into the tumor vasculature. Incorporating USNBs with existing cancer therapies such as chemotherapy, immunotherapy, and/or radiation therapy in several preclinical models has been demonstrated to have a profound effect on solid tumors. In this review, we provide an understanding of the composition and formation of nanobubbles (NBs), followed by the recent progress of the therapeutic combinatory effect of USNBs and other cancer therapies in cancer treatment.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Tera N. Petchiny
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics, and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
5
|
Mpekris F, Panagi M, Charalambous A, Voutouri C, Michael C, Papoui A, Stylianopoulos T. A synergistic approach for modulating the tumor microenvironment to enhance nano-immunotherapy in sarcomas. Neoplasia 2024; 51:100990. [PMID: 38520790 PMCID: PMC10978543 DOI: 10.1016/j.neo.2024.100990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
The lack of properly perfused blood vessels within tumors can significantly hinder the distribution of drugs, leading to reduced treatment effectiveness and having a negative impact on the quality of life of patients with cancer. This problem is particularly pronounced in desmoplastic cancers, where interactions between cancer cells, stromal cells, and the fibrotic matrix lead to tumor stiffness and the compression of most blood vessels within the tumor. To address this issue, two mechanotherapy approaches-mechanotherapeutics and ultrasound sonopermeation-have been employed separately to treat vascular abnormalities in tumors and have reached clinical trials. Here, we performed in vivo studies in sarcomas, to explore the conditions under which these two mechanotherapy strategies could be optimally combined to enhance perfusion and the efficacy of nano-immunotherapy. Our findings demonstrate that combination of the anti-histamine drug ketotifen, as a mechanotherapeutic, and sonopermeation effectively alleviates mechanical forces by decreasing 50 % collagen and hyaluronan levels and thus, reshaping the tumor microenvironment. Furthermore, the combined therapy normalizes the tumor vasculature by increasing two-fold the pericytes coverage. This combination not only improves six times tumor perfusion but also enhances drug delivery. As a result, blood vessel functionality is enhanced, leading to increased infiltration by 40 % of immune cells (CD4+ and CD8+ T-cells) and improving the antitumor efficacy of Doxil nanomedicine and anti-PD-1 immunotherapy. In conclusion, our research underscores the unique and synergistic potential of combining mechanotherapeutics and sonopermeation. Both approaches are undergoing clinical trials to enhance cancer therapy and have the potential to significantly improve nano-immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus.
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus
| | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus
| | - Antonia Papoui
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Cyprus.
| |
Collapse
|
6
|
Shakya G, Cattaneo M, Guerriero G, Prasanna A, Fiorini S, Supponen O. Ultrasound-responsive microbubbles and nanodroplets: A pathway to targeted drug delivery. Adv Drug Deliv Rev 2024; 206:115178. [PMID: 38199257 DOI: 10.1016/j.addr.2023.115178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Ultrasound-responsive agents have shown great potential as targeted drug delivery agents, effectively augmenting cell permeability and facilitating drug absorption. This review focuses on two specific agents, microbubbles and nanodroplets, and provides a sequential overview of their drug delivery process. Particular emphasis is given to the mechanical response of the agents under ultrasound, and the subsequent physical and biological effects on the cells. Finally, the state-of-the-art in their pre-clinical and clinical implementation are discussed. Throughout the review, major challenges that need to be overcome in order to accelerate their clinical translation are highlighted.
Collapse
Affiliation(s)
- Gazendra Shakya
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Marco Cattaneo
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Giulia Guerriero
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Anunay Prasanna
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Samuele Fiorini
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Outi Supponen
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|
7
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
8
|
Cattaneo M, Supponen O. Shell viscosity estimation of lipid-coated microbubbles. SOFT MATTER 2023; 19:5925-5941. [PMID: 37490014 DOI: 10.1039/d3sm00871a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Understanding the shell rheology of ultrasound contrast agent microbubbles is vital for anticipating their bioeffects in clinical practice. Past studies using sophisticated acoustic and optical techniques have made enormous progress in this direction, enabling the development of shell models that adequately reproduce the nonlinear behaviour of the coated microbubble under acoustic excitation. However, there have also been puzzling discrepancies and missing physical explanations for the dependency of shell viscosity on the equilibrium bubble radius, which demands further experimental investigations. In this study, we aim to unravel the cause of such behaviour by performing a refined characterisation of the shell viscosity. We use ultra-high-speed microscopy imaging, optical trapping and wide-field fluorescence to accurately record the individual microbubble response upon ultrasound driving across a range of bubble sizes. An advanced model of bubble dynamics is validated and employed to infer the shell viscosity of single bubbles from their radial time evolution. The resulting values reveal a prominent variability of the shell viscosity of about an order of magnitude and no dependency on the bubble size, which is contrary to previous studies. We find that the method called bubble spectroscopy, which has been used extensively in the past to determine the shell viscosity, is highly sensitive to methodology inaccuracies, and we demonstrate through analytical arguments that the previously reported unphysical trends are an artifact of these biases. We also show the importance of correct bubble sizing, as errors in this aspect can also lead to unphysical trends in shell viscosity, when estimated through a nonlinear fitting from the time response of the bubble.
Collapse
Affiliation(s)
- Marco Cattaneo
- Institute of Fluid Dynamics, Department of Mechanical and Process Engineering, ETH Zürich, Sonneggstrasse 3, 8092 Zürich, Switzerland.
| | - Outi Supponen
- Institute of Fluid Dynamics, Department of Mechanical and Process Engineering, ETH Zürich, Sonneggstrasse 3, 8092 Zürich, Switzerland.
| |
Collapse
|
9
|
Sharma D, Xuan Leong K, Palhares D, Czarnota GJ. Radiation combined with ultrasound and microbubbles: A potential novel strategy for cancer treatment. Z Med Phys 2023; 33:407-426. [PMID: 37586962 PMCID: PMC10517408 DOI: 10.1016/j.zemedi.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 08/18/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Several emerging technologies are helping to battle cancer. Cancer therapies have been effective at killing cancer cells, but a large portion of patients still die to this disease every year. As such, more aggressive treatments of primary cancers are employed and have been shown to be capable of saving a greater number of lives. Recent research advances the field of cancer therapy by employing the use of physical methods to alter tumor biology. It uses microbubbles to enhance radiation effect by damaging tumor vasculature followed by tumor cell death. The technique can specifically target tumor volumes by conforming ultrasound fields capable of microbubbles stimulation and localizing it to avoid vascular damage in surrounding tissues. Thus, this new application of ultrasound-stimulated microbubbles (USMB) can be utilized as a novel approach to cancer therapy by inducing vascular disruption resulting in tumor cell death. Using USMB alongside radiation has showed to augment the anti-vascular effect of radiation, resulting in enhanced tumor response. Recent work with nanobubbles has shown vascular permeation into intracellular space, extending the use of this new treatment method to potentially further improve the therapeutic effect of the ultrasound-based therapy. The significant enhancement of localized tumor cell kill means that radiation-based treatments can be made more potent with lower doses of radiation. This technique can manifest a greater impact on radiation oncology practice by increasing treatment effectiveness significantly while reducing normal tissue toxicity. This review article summarizes the past and recent advances in USMB enhancement of radiation treatments. The review mainly focuses on preclinical findings but also highlights some clinical findings that use USMB as a therapeutic modality in cancer therapy.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Kai Xuan Leong
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Daniel Palhares
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Kancheva M, Aronson L, Pattilachan T, Sautto F, Daines B, Thommes D, Shar A, Razavi M. Bubble-Based Drug Delivery Systems: Next-Generation Diagnosis to Therapy. J Funct Biomater 2023; 14:373. [PMID: 37504868 PMCID: PMC10382061 DOI: 10.3390/jfb14070373] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
Current radiologic and medication administration is systematic and has widespread side effects; however, the administration of microbubbles and nanobubbles (MNBs) has the possibility to provide therapeutic and diagnostic information without the same ramifications. Microbubbles (MBs), for instance, have been used for ultrasound (US) imaging due to their ability to remain in vessels when exposed to ultrasonic waves. On the other hand, nanobubbles (NBs) can be used for further therapeutic benefits, including chronic treatments for osteoporosis and cancer, gene delivery, and treatment for acute conditions, such as brain infections and urinary tract infections (UTIs). Clinical trials are also being conducted for different administrations and utilizations of MNBs. Overall, there are large horizons for the benefits of MNBs in radiology, general medicine, surgery, and many more medical applications. As such, this review aims to evaluate the most recent publications from 2016 to 2022 to report the current uses and innovations for MNBs.
Collapse
Affiliation(s)
- Mihaela Kancheva
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Lauren Aronson
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Tara Pattilachan
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Francesco Sautto
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Benjamin Daines
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Donald Thommes
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Angela Shar
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
11
|
Papadopoulou V, Stride EP, Borden MA, Eisenbrey JR, Dayton PA. Radiotherapy Sensitization With Ultrasound-Stimulated Intravenously Injected Oxygen Microbubbles Can Have Contrary Effects Depending on the Study Model. ULTRASOUND IN MEDICINE & BIOLOGY 2023:S0301-5629(23)00198-9. [PMID: 37442717 DOI: 10.1016/j.ultrasmedbio.2023.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/13/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023]
Affiliation(s)
- Virginie Papadopoulou
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Eleanor P Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Mark A Borden
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA; Biomaterial Engineering Program, University of Colorado, Boulder, CO, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Haram M, Hansen R, Bouget D, Myhre OF, Davies CDL, Hofsli E. Treatment of Liver Metastases With Focused Ultrasound and Microbubbles in Patients With Colorectal Cancer Receiving Chemotherapy. ULTRASOUND IN MEDICINE & BIOLOGY 2023:S0301-5629(23)00171-0. [PMID: 37336691 DOI: 10.1016/j.ultrasmedbio.2023.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 06/21/2023]
Abstract
OBJECTIVE Pre-clinical trials have obtained promising results that focused ultrasound (FUS) combined with microbubbles (MBs) increases tumor uptake and the therapeutic effect of drugs. The aims of the study described here were to investigate whether FUS and MBs could improve the effect of chemotherapy in patients with liver metastases from colorectal cancer and to investigate the safety and feasibility of using FUS + MBs. METHODS We included 17 patients with liver metastases from colorectal cancer, selected two lesions in each patient's liver and randomized the lesions for, respectively, treatment with FUS + MBs or control. After chemotherapy (FOLFIRI or FOLFOXIRI), the lesions were treated with FUS (frequency = 1.67 MHz, mechanical index = 0.5, pulse repetition frequency = 0.33 Hz, 33 oscillations, duty cycle = 0.2%-0.4% and MBs (SonoVue) for 35 min). Nine boluses of MBs were injected intravenously at 3.5 min intervals. Patients were scheduled for four cycles of treatment. Changes in the size of metastases were determined from computed tomography images. RESULTS Treatment with FUS + MBs is safe at the settings used. There was considerable variation in treatment response between lesions and mixed response between lesions receiving only chemotherapy. There is a tendency toward larger-volume reduction in lesions treated with FUS + MBs compared with control lesions, but a mixed response to chemotherapy and lesion heterogeneity make it difficult to interpret the results. CONCLUSION The combination of FUS and MBs is a safe, feasible and available strategy for improving the effect of chemotherapy in cancer patients. Therapeutic effect was not demonstrated in this trial. Multicenter trials with standardized protocols should be performed.
Collapse
Affiliation(s)
- Margrete Haram
- Department of Radiology and Nuclear Medicine, St. Olav's Hospital-Trondheim University Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital-Trondheim University Hospital, 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
| | - David Bouget
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ola Finneng Myhre
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Eva Hofsli
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital-Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
13
|
de Maar JS, Zandvliet MMJM, Veraa S, Tobón Restrepo M, Moonen CTW, Deckers R. Ultrasound and Microbubbles Mediated Bleomycin Delivery in Feline Oral Squamous Cell Carcinoma—An In Vivo Veterinary Study. Pharmaceutics 2023; 15:pharmaceutics15041166. [PMID: 37111651 PMCID: PMC10142092 DOI: 10.3390/pharmaceutics15041166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
To investigate the feasibility and tolerability of ultrasound and microbubbles (USMB)-enhanced chemotherapy delivery for head and neck cancer, we performed a veterinary trial in feline companion animals with oral squamous cell carcinomas. Six cats were treated with a combination of bleomycin and USMB therapy three times, using the Pulse Wave Doppler mode on a clinical ultrasound system and EMA/FDA approved microbubbles. They were evaluated for adverse events, quality of life, tumour response and survival. Furthermore, tumour perfusion was monitored before and after USMB therapy using contrast-enhanced ultrasound (CEUS). USMB treatments were feasible and well tolerated. Among 5 cats treated with optimized US settings, 3 had stable disease at first, but showed disease progression 5 or 11 weeks after first treatment. One cat had progressive disease one week after the first treatment session, maintaining a stable disease thereafter. Eventually, all cats except one showed progressive disease, but each survived longer than the median overall survival time of 44 days reported in literature. CEUS performed immediately before and after USMB therapy suggested an increase in tumour perfusion based on an increase in median area under the curve (AUC) in 6 out of 12 evaluated treatment sessions. In this small hypothesis-generating study, USMB plus chemotherapy was feasible and well-tolerated in a feline companion animal model and showed potential for enhancing tumour perfusion in order to increase drug delivery. This could be a forward step toward clinical translation of USMB therapy to human patients with a clinical need for locally enhanced treatment.
Collapse
Affiliation(s)
- Josanne S. de Maar
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| | - Maurice M. J. M. Zandvliet
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Stefanie Veraa
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Mauricio Tobón Restrepo
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Chrit T. W. Moonen
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| | - Roel Deckers
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
14
|
Kaykanat SI, Uguz AK. The role of acoustofluidics and microbubble dynamics for therapeutic applications and drug delivery. BIOMICROFLUIDICS 2023; 17:021502. [PMID: 37153864 PMCID: PMC10162024 DOI: 10.1063/5.0130769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/18/2023] [Indexed: 05/10/2023]
Abstract
Targeted drug delivery is proposed to reduce the toxic effects of conventional therapeutic methods. For that purpose, nanoparticles are loaded with drugs called nanocarriers and directed toward a specific site. However, biological barriers challenge the nanocarriers to convey the drug to the target site effectively. Different targeting strategies and nanoparticle designs are used to overcome these barriers. Ultrasound is a new, safe, and non-invasive drug targeting method, especially when combined with microbubbles. Microbubbles oscillate under the effect of the ultrasound, which increases the permeability of endothelium, hence, the drug uptake to the target site. Consequently, this new technique reduces the dose of the drug and avoids its side effects. This review aims to describe the biological barriers and the targeting types with the critical features of acoustically driven microbubbles focusing on biomedical applications. The theoretical part covers the historical developments in microbubble models for different conditions: microbubbles in an incompressible and compressible medium and bubbles encapsulated by a shell. The current state and the possible future directions are discussed.
Collapse
Affiliation(s)
- S. I. Kaykanat
- Department of Chemical Engineering, Boğaziçi University, 34342 Bebek, Istanbul, Türkiye
| | | |
Collapse
|
15
|
Moradi Kashkooli F, Jakhmola A, Hornsby TK, Tavakkoli JJ, Kolios MC. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions. J Control Release 2023; 355:552-578. [PMID: 36773959 DOI: 10.1016/j.jconrel.2023.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The application of biocompatible nanocarriers in medicine has provided several benefits over conventional treatment methods. However, achieving high treatment efficacy and deep penetration of nanocarriers in tumor tissue is still challenging. To address this, stimuli-responsive nano-sized drug delivery systems (DDSs) are an active area of investigation in delivering anticancer drugs. While ultrasound is mainly used for diagnostic purposes, it can also be applied to affect cellular function and the delivery/release of anticancer drugs. Therapeutic ultrasound (TUS) has shown potential as both a stand-alone anticancer treatment and a method to induce targeted drug release from nanocarrier systems. TUS approaches have been used to overcome various physiological obstacles, including endothelial barriers, the tumor microenvironment (TME), and immunological hurdles. Combining nanomedicine and ultrasound as a smart DDS can increase in situ drug delivery and improve access to impermeable tissues. Furthermore, smart DDSs can perform targeted drug release in response to distinctive TMEs, external triggers, or dual/multi-stimulus. This results in enhanced treatment efficacy and reduced damage to surrounding healthy tissue or organs at risk. Integrating DDSs and ultrasound is still in its early stages. More research and clinical trials are required to fully understand ultrasound's underlying physical mechanisms and interactions with various types of nanocarriers and different types of cells and tissues. In the present review, ultrasound-mediated nano-sized DDS, specifically focused on cancer treatment, is presented and discussed. Ultrasound interaction with nanoparticles (NPs), drug release mechanisms, and various types of ultrasound-sensitive NPs are examined. Additionally, in vitro, in vivo, and clinical applications of TUS are reviewed in light of the critical challenges that need to be considered to advance TUS toward an efficient, secure, straightforward, and accessible cancer treatment. This study also presents effective TUS parameters and safety considerations for this treatment modality and gives recommendations about system design and operation. Finally, future perspectives are considered, and different TUS approaches are examined and discussed in detail. This review investigates drug release and delivery through ultrasound-mediated nano-sized cancer treatment, both pre-clinically and clinically.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
16
|
Zhao X, Pellow C, Goertz DE. Intravital imaging and cavitation monitoring of antivascular ultrasound in tumor microvasculature. Theranostics 2023; 13:250-266. [PMID: 36593952 PMCID: PMC9800738 DOI: 10.7150/thno.79186] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
Rationale: Focused ultrasound-stimulated microbubbles have been shown to be capable of inducing blood flow shutdown and necrosis in a range of tissue types in an approach termed antivascular ultrasound or nonthermal ablation. In oncology, this approach has demonstrated tumor growth inhibition, and profound synergistic antitumor effects when combined with traditional platforms of chemo-, radiation- and immune-therapies. However, the exposure schemes employed have been broad and underlying mechanisms remain unclear with fundamental questions about exposures, vessel types and sizes involved, and the nature of bubble behaviors and their acoustic emissions resulting in vascular damage - impeding the establishment of standard protocols. Methods: Here, ultrasound transmitters and receivers are integrated into a murine dorsal window chamber tumor model for intravital microscopy studies capable of real-time visual and acoustic monitoring during antivascular ultrasound. Vessel type (normal and tumor-affected), caliber, and viability are assessed under higher pressure conditions (1, 2, and 3 MPa), and cavitation signatures are linked to the biological effects. Results: Vascular events occurred preferentially in tumor-affected vessels with greater incidence in smaller vessels and with more severity as a function of increasing pressure. Vascular blood flow shutdown was found to be due to a combination of focal disruption events and network-related flow changes. Acoustic emissions displayed elevated broadband noise and distinct sub- and ultra-harmonics and their associated third-order peaks with increasing pressure. Conclusions: The observed vascular events taken collectively with identified cavitation signatures provide an improved mechanistic understanding of antivascular ultrasound at the microscale, with implications for establishing a specific treatment protocol and control platform.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Medical Biophysics, University of Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| | | | - David E. Goertz
- Department of Medical Biophysics, University of Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| |
Collapse
|
17
|
McCorkell G, Nakayama M, Feltis B, Piva T, Geso M. Ultrasound-Stimulated Microbubbles Enhance Radiation-Induced Cell Killing. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:2449-2460. [PMID: 36100510 DOI: 10.1016/j.ultrasmedbio.2022.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 03/29/2022] [Accepted: 07/01/2022] [Indexed: 06/15/2023]
Abstract
Recent in vivo studies using ultrasound-stimulated microbubbles as a localized radiosensitizer have had impressive results. While in vitro studies have also obtained similar results using human umbilical vein endothelial cells (HUVEC), studies using other cell lines have had varying results. This study was aimed at investigating any increases in radiation-induced cell killing in vitro using two carcinoma lines not previously investigated before (metastatic follicular thyroid carcinoma cells [FTC-238] and non-small cell lung carcinoma cells [NCI-H727]), in addition to HUVEC. Cells were treated using a combination of 1.6% (v/v) microbubbles, ∼90 s of 2-MHz ultrasound (mechanical index = 0.8) and 0-6 Gy of kilovolt or MV X-rays. Cell viability assays obtained 72 h post-treatment were normalized to untreated controls, and analysis of variance was used to determine statistical significance. All cells treated with combined ultrasound-stimulated microbubbles and radiation exhibited decreased normalized survival, with statistically significant effects observed for the NCI-H727 cells. No statistically significant differences in effects were observed using kV compared with MV radiation. Further studies using increased microbubble concentrations may be required to achieve statistically significant results for the FTC-238 and HUVEC lines.
Collapse
Affiliation(s)
- Giulia McCorkell
- Department of Medical Radiations, School of Health and Biomedical Sciences, RMIT University, Victoria, Australia
| | - Masao Nakayama
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Bryce Feltis
- Department of Human Bioscience, School of Health and Biomedical Sciences, RMIT University, Victoria, Australia
| | - Terrence Piva
- Department of Human Bioscience, School of Health and Biomedical Sciences, RMIT University, Victoria, Australia
| | - Moshi Geso
- Department of Medical Radiations, School of Health and Biomedical Sciences, RMIT University, Victoria, Australia.
| |
Collapse
|
18
|
Chapla R, Huynh KT, Schutt CE. Microbubble–Nanoparticle Complexes for Ultrasound-Enhanced Cargo Delivery. Pharmaceutics 2022; 14:pharmaceutics14112396. [PMID: 36365214 PMCID: PMC9698658 DOI: 10.3390/pharmaceutics14112396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble–nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid–polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin–avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble–nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.
Collapse
Affiliation(s)
- Rachel Chapla
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katherine T. Huynh
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
19
|
Yu Y, Qiao W, Feng S, Yi C, Liu Z. Inhibition of Walker-256 Tumor Growth by Combining Microbubble-Enhanced Ultrasound and Endostar. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:2591-2600. [PMID: 35106800 DOI: 10.1002/jum.15949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVES This research is to investigate the anti-tumor effects by combining anti-vascular effect of microbubble enhanced ultrasound (MEUS) mechanical destruction and anti-angiogenic effect of Endostar. METHODS Rats bearing Walker-256 tumor were randomly divided into 4 groups treated by Endostar + MEUS combination, Endostar, MEUS or Sham ultrasound (US), respectively. MEUS was induced by Sonazoid microbubble and a focused therapeutic US device. Contrast-enhanced ultrasound (CEUS) was used to assess tumor perfusion before and after treatment. Microvessel density (MVD) was evaluated with immunohistochemical staining of CD31, CD34, and VEGFA. TUNEL assay was used to determine the apoptosis rate of tumor cells. RESULTS Endostar + MEUS combined group induced the most reduced blood perfusion and most retarded tumor growth compared with other 3 groups. Decreased MVD was shown in Endostar + MEUS, Endostar and MEUS group, but the lowest MVD value was presented in the combined treatment group. Significant increase was observed in the combined therapy group and MEUS group. CONCLUSIONS This study showed an improved anti-vascular and anti-angiogenic effect achieved by combining Endostar and MEUS, and may provide a new method potential for anti-tumor therapy.
Collapse
Affiliation(s)
- Yanlan Yu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wei Qiao
- Department of Ultrasound, General Hospital of Central Theatre Commander Theater, Wuhan, China
| | - Shuang Feng
- Department of Ultrasound, General Hospital of Southern Theatre Command, Guangzhou, China
| | - Cuo Yi
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
20
|
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: 25] [Impact Index Per Article: 8.3] [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
|
21
|
Sharma D, Czarnota GJ. Involvement of Ceramide Signalling in Radiation-Induced Tumour Vascular Effects and Vascular-Targeted Therapy. Int J Mol Sci 2022; 23:ijms23126671. [PMID: 35743121 PMCID: PMC9223569 DOI: 10.3390/ijms23126671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Sphingolipids are well-recognized critical components in several biological processes. Ceramides constitute a class of sphingolipid metabolites that are involved in important signal transduction pathways that play key roles in determining the fate of cells to survive or die. Ceramide accumulated in cells causes apoptosis; however, ceramide metabolized to sphingosine promotes cell survival and angiogenesis. Studies suggest that vascular-targeted therapies increase endothelial cell ceramide resulting in apoptosis that leads to tumour cure. Specifically, ultrasound-stimulated microbubbles (USMB) used as vascular disrupting agents can perturb endothelial cells, eliciting acid sphingomyelinase (ASMase) activation accompanied by ceramide release. This phenomenon results in endothelial cell death and vascular collapse and is synergistic with other antitumour treatments such as radiation. In contrast, blocking the generation of ceramide using multiple approaches, including the conversion of ceramide to sphingosine-1-phosphate (S1P), abrogates this process. The ceramide-based cell survival "rheostat" between these opposing signalling metabolites is essential in the mechanotransductive vascular targeting following USMB treatment. In this review, we aim to summarize the past and latest findings on ceramide-based vascular-targeted strategies, including novel mechanotransductive methodologies.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
- Correspondence: ; Tel.: +1-416-480-6100 (ext. 89533)
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
22
|
Application of Ultrasound Combined with Microbubbles for Cancer Therapy. Int J Mol Sci 2022; 23:ijms23084393. [PMID: 35457210 PMCID: PMC9026557 DOI: 10.3390/ijms23084393] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023] Open
Abstract
At present, cancer is one of the leading causes of death worldwide. Treatment failure remains one of the prime hurdles in cancer treatment due to the metastatic nature of cancer. Techniques have been developed to hinder the growth of tumours or at least to stop the metastasis process. In recent years, ultrasound therapy combined with microbubbles has gained immense success in cancer treatment. Ultrasound-stimulated microbubbles (USMB) combined with other cancer treatments including radiation therapy, chemotherapy or immunotherapy has demonstrated potential improved outcomes in various in vitro and in vivo studies. Studies have shown that low dose radiation administered with USMB can have similar effects as high dose radiation therapy. In addition, the use of USMB in conjunction with radiotherapy or chemotherapy can minimize the toxicity of high dose radiation or chemotherapeutic drugs, respectively. In this review, we discuss the biophysical properties of USMB treatment and its applicability in cancer therapy. In particular, we highlight important preclinical and early clinical findings that demonstrate the antitumour effect combining USMB and other cancer treatment modalities (radiotherapy and chemotherapy). Our review mainly focuses on the tumour vascular effects mediated by USMB and these cancer therapies. We also discuss several current limitations, in addition to ongoing and future efforts for applying USMB in cancer treatment.
Collapse
|
23
|
Escoffre JM, Sekkat N, Oujagir E, Bodard S, Mousset C, Presset A, Chautard R, Ayoub J, Lecomte T, Bouakaz A. Delivery of anti-cancer drugs using microbubble-assisted ultrasound in digestive oncology: From preclinical to clinical studies. Expert Opin Drug Deliv 2022; 19:421-433. [PMID: 35363586 DOI: 10.1080/17425247.2022.2061459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The combination of microbubbles (MBs) and ultrasound (US) is an emerging method for the noninvasive and targeted enhancement of intratumor chemotherapeutic uptake. This method showed an increased local drug extravasation in tumor tissue while reducing the systemic adverse effects in various tumor models. AREA COVERED We focused on preclinical and clinical studies investigating the therapeutic efficacy and safety of this technology for the treatment of colorectal, pancreatic and liver cancers. We discussed the limitations of the current investigations and future perspectives. EXPERT OPINION The therapeutic efficacy and the safety of delivery of standard chemotherapy regimen using MB-assisted US have been mainly demonstrated in subcutaneous models of digestive cancers. Although some clinical trials on pancreatic ductal carcinoma and hepatic metastases from various digestive cancers have shown promising results, successful evaluation of this method in terms of US settings, chemotherapeutic schemes and MBs-related parameters will need to be addressed in more relevant preclinical models of digestive cancers, in small and large animals before fully and successfully translating this technology for clinic use. Ultimately, a clear evidence of the correlation between the enhanced intratumoral concentrations of therapeutics and the increased therapeutic response of tumors have to be provided in clinical trials.
Collapse
Affiliation(s)
| | - Najib Sekkat
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France
| | - Edward Oujagir
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France
| | - Sylvie Bodard
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France
| | - Coralie Mousset
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France
| | - Antoine Presset
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France
| | - Romain Chautard
- Inserm UMR 1069, Nutrition, Croissance et Cancer (N2C), Université de Tours, Tours, France.,Department of Hepato-Gastroenterology & Digestive Oncology, CHRU de Tours, Tours, France
| | - Jean Ayoub
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France.,Departement of Echography & Doppler, CHRU de Tours, Tours, France
| | - Thierry Lecomte
- Inserm UMR 1069, Nutrition, Croissance et Cancer (N2C), Université de Tours, Tours, France.,Department of Hepato-Gastroenterology & Digestive Oncology, CHRU de Tours, Tours, France
| | - Ayache Bouakaz
- Université de Tours, UMR 1253, iBrain, Inserm, Tours, France
| |
Collapse
|
24
|
Sasaki N, Ikenaka Y, Aoshima K, Aoyagi T, Kudo N, Nakamura K, Takiguchi M. Safety Assessment of Ultrasound-Assisted Intravesical Chemotherapy in Normal Dogs: A Pilot Study. Front Pharmacol 2022; 13:837754. [PMID: 35370726 PMCID: PMC8974685 DOI: 10.3389/fphar.2022.837754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Intravesical chemotherapy after transurethral resection is a treatment option in patients with non-muscle invasive bladder cancer. The efficacy of intravesical chemotherapy is determined by the cellular uptake of intravesical drugs. Therefore, drug delivery technologies in the urinary bladder are promising tools for enhancing the efficacy of intravesical chemotherapy. Ultrasound-triggered microbubble cavitation may enhance the permeability of the urothelium, and thus may have potential as a drug delivery technology in the urinary bladder. Meanwhile, the enhanced permeability may increase systemic absorption of intravesical drugs, which may increase the adverse effects of the drug. The aim of this preliminary safety study was to assess the systemic absorption of an intravesical drug that was delivered by ultrasound-triggered microbubble cavitation in the urinary bladder of normal dogs. Pirarubicin, a derivative of doxorubicin, and an ultrasound contrast agent (Sonazoid) microbubbles were administered in the urinary bladder. Ultrasound (transmitting frequency 5 MHz; pulse duration 0.44 μsec; pulse repetition frequency 7.7 kHz; peak negative pressure −1.2 MPa) was exposed to the bladder using a diagnostic ultrasound probe (PLT-704SBT). The combination of ultrasound and microbubbles did not increase the plasma concentration of intravesical pirarubicin. In addition, hematoxylin and eosin staining showed that the combination of ultrasound and microbubble did not cause observable damages to the urothelium. Tissue pirarubicin concentration in the sonicated region was higher than that of the non-sonicated region in two of three dogs. The results of this pilot study demonstrate the safety of the combination of intravesical pirarubicin and ultrasound-triggered microbubble cavitation, that is, ultrasound-assisted intravesical chemotherapy.
Collapse
Affiliation(s)
- Noboru Sasaki
- Laboratory of Veterinary Internal Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- *Correspondence: Noboru Sasaki,
| | - Yoshinori Ikenaka
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Teiichiro Aoyagi
- Department of Urology, Tokyo Medical University Ibaraki Medical Center, Ami, Japan
| | - Nobuki Kudo
- Division of Bioengineering and Bioinformatics, Faculty of Information and Technology, Hokkaido University, Sapporo, Japan
| | - Kensuke Nakamura
- Laboratory of Veterinary Internal Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mitsuyoshi Takiguchi
- Laboratory of Veterinary Internal Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
25
|
Ingram N, McVeigh LE, Abou-Saleh RH, Batchelor DVB, Loadman PM, McLaughlan JR, Markham AF, Evans SD, Coletta PL. A Single Short 'Tone Burst' Results in Optimal Drug Delivery to Tumours Using Ultrasound-Triggered Therapeutic Microbubbles. Pharmaceutics 2022; 14:pharmaceutics14030622. [PMID: 35335995 PMCID: PMC8953493 DOI: 10.3390/pharmaceutics14030622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 12/10/2022] Open
Abstract
Advanced drug delivery systems, such as ultrasound-mediated drug delivery, show great promise for increasing the therapeutic index. Improvements in delivery by altering the ultrasound parameters have been studied heavily in vitro but relatively little in vivo. Here, the same therapeutic microbubble and tumour type are used to determine whether altering ultrasound parameters can improve drug delivery. Liposomes were loaded with SN38 and attached via avidin: biotin linkages to microbubbles. The whole structure was targeted to the tumour vasculature by the addition of anti-vascular endothelial growth factor receptor 2 antibodies. Tumour drug delivery and metabolism were quantified in SW480 xenografts after application of an ultrasound trigger to the tumour region. Increasing the trigger duration from 5 s to 2 min or increasing the number of 5 s triggers did not improve drug delivery, nor did changing to a chirp trigger designed to stimulate a greater proportion of the microbubble population, although this did show that the short tone trigger resulted in greater release of free SN38. Examination of ultrasound triggers in vivo to improve drug delivery is justified as there are multiple mechanisms at play that may not allow direct translation from in vitro findings. In this setting, a short tone burst gives the best ultrasound parameters for tumoural drug delivery.
Collapse
Affiliation(s)
- Nicola Ingram
- Leeds Institute of Medical Research, Faculty of Medicine and Health, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK; (L.E.M.); (J.R.M.); (A.F.M.)
- Correspondence: (N.I.); (P.L.C.)
| | - Laura E. McVeigh
- Leeds Institute of Medical Research, Faculty of Medicine and Health, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK; (L.E.M.); (J.R.M.); (A.F.M.)
| | - Radwa H. Abou-Saleh
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK; (R.H.A.-S.); (D.V.B.B.); (S.D.E.)
- Nanoscience and Technology Group, Faculty of Science, Galala University, Galala 43711, Egypt
- Department of Physics, Mansoura University, Mansoura 35516, Egypt
| | - Damien V. B. Batchelor
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK; (R.H.A.-S.); (D.V.B.B.); (S.D.E.)
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK;
| | - James R. McLaughlan
- Leeds Institute of Medical Research, Faculty of Medicine and Health, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK; (L.E.M.); (J.R.M.); (A.F.M.)
- School of Electronic and Electrical Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Alexander F. Markham
- Leeds Institute of Medical Research, Faculty of Medicine and Health, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK; (L.E.M.); (J.R.M.); (A.F.M.)
| | - Stephen D. Evans
- School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK; (R.H.A.-S.); (D.V.B.B.); (S.D.E.)
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Faculty of Medicine and Health, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK; (L.E.M.); (J.R.M.); (A.F.M.)
- Correspondence: (N.I.); (P.L.C.)
| |
Collapse
|
26
|
Chettab K, Matera EL, Lafond M, Coralie D, Favin-Lévêque C, Goy C, Strakhova R, Mestas JL, Lafon C, Dumontet C. Proof of Concept: Protein Delivery into Human Erythrocytes Using Stable Cavitation. Mol Pharm 2022; 19:929-935. [PMID: 35147436 DOI: 10.1021/acs.molpharmaceut.1c00907] [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: 11/28/2022]
Abstract
Human erythrocytes represent candidates of choice as carriers for a wide range of drugs due to their unique biophysical and physiological properties. In this study, we used a sonoporation device generating and monitoring acoustic stable cavitation without any addition of contrast or nucleation agents. The device was evaluated for bovine serum albumin (BSA) delivery into human erythrocytes. After determining the adequate hematocrit percentage compatible with the generation of stable cavitation, we determined the optimal sonoporation conditions allowing BSA delivery while preserving erythrocyte integrity. Our results demonstrate that stable cavitation allows efficient delivery of proteins into human erythrocytes with limited lysis of these cells. In conclusion, our study allowed for the development of a stable and regulated cavitation program and the establishment of sonoporation conditions suitable for intracellular protein delivery while maintaining erythrocyte integrity. Additional investigations are needed to move from the proof of concept to a larger-scale application.
Collapse
Affiliation(s)
- Kamel Chettab
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France.,Hospices Civils de Lyon, Centre Hospitaller Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Bénite 69310, France
| | - Eva-Laure Matera
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France
| | - Maxime Lafond
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Université Lyon, Lyon F-69003, France
| | - Durieux Coralie
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France
| | - Camille Favin-Lévêque
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France
| | - Clémence Goy
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France
| | - Regina Strakhova
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France
| | - Jean-Louis Mestas
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Université Lyon, Lyon F-69003, France
| | - Cyril Lafon
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Université Lyon, Lyon F-69003, France
| | - Charles Dumontet
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, Université de Lyon, Lyon 69008, France.,Hospices Civils de Lyon, Centre Hospitaller Lyon Sud, 165 Chemin du Grand Revoyet, Pierre-Bénite 69310, France
| |
Collapse
|
27
|
de Maar JS, Rousou C, van Elburg B, Vos HJ, Lajoinie GPR, Bos C, Moonen CTW, Deckers R. Ultrasound-Mediated Drug Delivery With a Clinical Ultrasound System: In Vitro Evaluation. Front Pharmacol 2021; 12:768436. [PMID: 34737709 PMCID: PMC8560689 DOI: 10.3389/fphar.2021.768436] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy efficacy is often reduced by insufficient drug uptake in tumor cells. The combination of ultrasound and microbubbles (USMB) has been shown to improve drug delivery and to enhance the efficacy of several drugs in vitro and in vivo, through effects collectively known as sonopermeation. However, clinical translation of USMB therapy is hampered by the large variety of (non-clinical) US set-ups and US parameters that are used in these studies, which are not easily translated to clinical practice. In order to facilitate clinical translation, the aim of this study was to prove that USMB therapy using a clinical ultrasound system (Philips iU22) in combination with clinically approved microbubbles (SonoVue) leads to efficient in vitro sonopermeation. To this end, we measured the efficacy of USMB therapy for different US probes (S5-1, C5-1 and C9-4) and US parameters in FaDu cells. The US probe with the lowest central frequency (i.e. 1.6 MHz for S5-1) showed the highest USMB-induced intracellular uptake of the fluorescent dye SYTOX™ Green (SG). These SG uptake levels were comparable to or even higher than those obtained with a custom-built US system with optimized US parameters. Moreover, USMB therapy with both the clinical and the custom-built US system increased the cytotoxicity of the hydrophilic drug bleomycin. Our results demonstrate that a clinical US system can be used to perform USMB therapy as efficiently as a single-element transducer set-up with optimized US parameters. Therefore, future trials could be based on these clinical US systems, including validated US parameters, in order to accelerate successful translation of USMB therapy.
Collapse
Affiliation(s)
- Josanne S de Maar
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Charis Rousou
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, Netherlands
| | - Benjamin van Elburg
- Physics of Fluids Group, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands
| | - Hendrik J Vos
- Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, Netherlands
| | - Guillaume P R Lajoinie
- Physics of Fluids Group, MIRA Institute of Biomedical Technology and Technical Medicine, University of Twente, Enschede, Netherlands
| | - Clemens Bos
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Chrit T W Moonen
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Roel Deckers
- Imaging and Oncology Division, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
28
|
Wahab S, Alshahrani MY, Ahmad MF, Abbas H. Current trends and future perspectives of nanomedicine for the management of colon cancer. Eur J Pharmacol 2021; 910:174464. [PMID: 34474029 DOI: 10.1016/j.ejphar.2021.174464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Colon cancer (CC) kills countless people every year throughout the globe. It persists as one of the highly lethal diseases to be treated because the overall survival rate for CC is meagre. Early diagnosis and efficient treatments are two of the biggest hurdles in the fight against cancer. In the present work, we will review thriving strategies for CC targeted drug delivery and critically explain the most recent progressions on emerging novel nanotechnology-based drug delivery systems. Nanotechnology-based animal and human clinical trial studies targeting CC are discussed. Advancements in nanotechnology-based drug delivery systems intended to enhance cellular uptake, improved pharmacokinetics and effectiveness of anticancer drugs have facilitated the powerful targeting of specific agents for CC therapy. This review provides insight into current progress and future opportunities for nanomedicines as potential curative targets for CC treatment. This information could be used as a platform for the future expansion of multi-functional nano constructs for CC's advanced detection and functional drug delivery.
Collapse
Affiliation(s)
- Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hashim Abbas
- Queens Medical Center, Nottingham University Hospitals, NHS, Nottingham, UK
| |
Collapse
|
29
|
Ultrasound contrast agents: microbubbles made simple for the pediatric radiologist. Pediatr Radiol 2021; 51:2117-2127. [PMID: 34117892 PMCID: PMC9288183 DOI: 10.1007/s00247-021-05080-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 02/25/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
The ability to provide prompt, real-time, easily accessible and radiation-free diagnostic assessments makes ultrasound (US) one of the most versatile imaging modalities. The introduction and development of stable microbubble-based ultrasound contrast agents (UCAs) in the early 1990s improved visualization of complex vascular structures, overcoming some of the limitations of B-mode and Doppler imaging. UCAs have been used extensively in the adult population to visualize vasculature and to evaluate perfusion and blood flow dynamics in organs and lesions. Since the first observations that air bubbles within a liquid can generate a strong echogenic effect, to the early makeshift approaches with agitated saline, and later to the development of industrially produced and federally approved UCAs, these agents have evolved to become both clinically and commercially viable. Perhaps the most exciting potential of UCAs is being uncovered by current research that explores the use of these agents for molecular imaging and therapeutic applications. As contrast-enhanced ultrasound (CEUS) becomes more widely available, it is important for pediatric radiologists to understand the physics of the interaction between the US signal and the microbubbles in order to properly utilize them for the highest level of diagnostic imaging and interventions. In this article we introduce the composition of UCAs and the physics of their behavior in US, and we offer a brief history of their development over the last decades.
Collapse
|
30
|
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: 64] [Impact Index Per Article: 16.0] [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
|
31
|
Ultrasound-Enabled Therapeutic Delivery and Regenerative Medicine: Physical and Biological Perspectives. ACS Biomater Sci Eng 2021; 7:4371-4387. [PMID: 34460238 DOI: 10.1021/acsbiomaterials.1c00276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of ultrasound in medicine and biological sciences is expanding rapidly beyond its use in conventional diagnostic imaging. Numerous studies have reported the effects of ultrasound on cellular and tissue physiology. Advances in instrumentation and electronics have enabled successful in vivo applications of therapeutic ultrasound. Despite path breaking advances in understanding the biophysical and biological mechanisms at both microscopic and macroscopic scales, there remain substantial gaps. With the progression of research in this area, it is important to take stock of the current understanding of the field and to highlight important areas for future work. We present herein key developments in the biological applications of ultrasound especially in the context of nanoparticle delivery, drug delivery, and regenerative medicine. We conclude with a brief perspective on the current promise, limitations, and future directions for interfacing ultrasound technology with biological systems, which could provide guidance for future investigations in this interdisciplinary area.
Collapse
|
32
|
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
|
33
|
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: 0.8] [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
|
34
|
Zhang Y, Tang N, Huang L, Qiao W, Zhu Q, Liu Z. Effect of diagnostic ultrasound and microbubble-enhanced chemotherapy on metastasis of rabbit VX2 tumor. Med Phys 2021; 48:3927-3935. [PMID: 33774845 DOI: 10.1002/mp.14867] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Ultrasound-targeted microbubble destruction (UTMD) has been widely applied to enhance chemotherapy of tumors, yet few studies have focused on the metastatic potential induced by UTMD. This study aimed to explore the metastasis of VX2 tumors after treatment with UTMD and chemotherapy. METHODS Forty-four New Zealand rabbits bearing subcutaneous VX2 tumors were enrolled for the treatment of UTMD with chemotherapy. For UTMD, the tumors were insonated using two pulsing protocols of diagnostic ultrasound (DUS, VINNO and ECARE) with a mechanical index (MI) of 0.29-0.33, tone burst of 8.0 cycles, and frequencies of 3-4 MHz. A total dose of 2 ml SonoVue® was injected intermittently during 10-min UTMD exposure. The combination therapy was treated using doxorubicin (DOX, 2 mg/kg) and DUS, while the tumors treated using DOX only served as the control. Tumor size was measured using the tumor volume formula. Survival time was observed until animal death or the end of the study (120 days). Specific organs (lung, liver, kidney, and brain) were removed for metastatic evaluation. RESULTS There were no statistical differences in overall metastasis classification and individual organ metastases among all groups (P > 0.05). The tumor growth rate only showed inhibition on the 5th day (P < 0.01). The survival time did not demonstrate any significant difference between UTMD and chemotherapy only (P > 0.05). CONCLUSIONS UTMD using long-pulse DUS with commercial microbubbles did not pose a risk of metastasis enhancement in DOX chemotherapy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Najiao Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Leidan Huang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wei Qiao
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.,General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
35
|
Sanwal R, Joshi K, Ditmans M, Tsai SSH, Lee WL. Ultrasound and Microbubbles for Targeted Drug Delivery to the Lung Endothelium in ARDS: Cellular Mechanisms and Therapeutic Opportunities. Biomedicines 2021; 9:biomedicines9070803. [PMID: 34356867 PMCID: PMC8301318 DOI: 10.3390/biomedicines9070803] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by increased permeability of the alveolar–capillary membrane, a thin barrier composed of adjacent monolayers of alveolar epithelial and lung microvascular endothelial cells. This results in pulmonary edema and severe hypoxemia and is a common cause of death after both viral (e.g., SARS-CoV-2) and bacterial pneumonia. The involvement of the lung in ARDS is notoriously heterogeneous, with consolidated and edematous lung abutting aerated, less injured regions. This makes treatment difficult, as most therapeutic approaches preferentially affect the normal lung regions or are distributed indiscriminately to other organs. In this review, we describe the use of thoracic ultrasound and microbubbles (USMB) to deliver therapeutic cargo (drugs, genes) preferentially to severely injured areas of the lung and in particular to the lung endothelium. While USMB has been explored in other organs, it has been under-appreciated in the treatment of lung injury since ultrasound energy is scattered by air. However, this limitation can be harnessed to direct therapy specifically to severely injured lungs. We explore the cellular mechanisms governing USMB and describe various permutations of cargo administration. Lastly, we discuss both the challenges and potential opportunities presented by USMB in the lung as a tool for both therapy and research.
Collapse
Affiliation(s)
- Rajiv Sanwal
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kushal Joshi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Mihails Ditmans
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Scott S. H. Tsai
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
| | - Warren L. Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1T8, Canada; (R.S.); (K.J.); (M.D.); (S.S.H.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Toronto, ON M5B 1T8, Canada
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, ON M5B 2K3, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence: ; Tel.: +416-864-6060 (ext. 77655)
| |
Collapse
|
36
|
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: 5.3] [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
|
37
|
Abstract
Pancreatic cancer (PC) is one of the deadliest malignancies. The high mortality rate of PC largely results from delayed diagnosis and early metastasis. Therefore, identifying novel treatment targets for patients with PC is urgently required to improve survival rates. A major barrier to successful treatment of PC is the presence of a hypoxic tumor microenvironment, which is associated with poor prognosis, treatment resistance, increased invasion and metastasis. Recent studies have identified a number of novel molecules and pathways in PC cells that promote cancer cells progression under hypoxic conditions, which may provide new therapy strategies to inhibit the development and metastasis of PC. This review summarizes the latest research of hypoxia in PC and provides an overview of how the current therapies have the capacity to overcome hypoxia and improve PC patient treatment. These findings will eventually provide guidance for future PC management and clinical trials and hopefully improve the survival of patients with PC.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
38
|
Fagerland SMT, Berg S, Hill DK, Snipstad S, Sulheim E, Hyldbakk A, Kim J, Davies CDL. Ultrasound-Mediated Delivery of Chemotherapy into the Transgenic Adenocarcinoma of the Mouse Prostate Model. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:3032-3045. [PMID: 32800470 DOI: 10.1016/j.ultrasmedbio.2020.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 06/11/2023]
Abstract
Ultrasound (US) in combination with microbubbles (MB) has had promising results in improving delivery of chemotherapeutic agents. However, most studies are done in immunodeficient mice with xenografted tumors. We used two phenotypes of the spontaneous transgenic adenocarcinoma of the mouse prostate (TRAMP) model to evaluate if US + MB could enhance the therapeutic efficacy of cabazitaxel (Cab). Cab was either injected intravenously as free drug or encapsulated into nanoparticles. In both cases, Cab transiently reduced tumor and prostate volume in the TRAMP model. No additional therapeutic efficacy was observed combining Cab with US + MB, except for one tumor. Additionally, histology grading and immunostaining of Ki67 did not reveal differences between treatment groups. Mass spectrometry revealed that nanoparticle encapsulation of Cab increased the circulation time and enhanced the accumulation in liver and spleen compared with free Cab. The therapeutic results in this spontaneous, clinically relevant tumor model differ from the improved therapeutic response observed in xenografts combining US + MB and chemotherapy.
Collapse
Affiliation(s)
- Stein-Martin T Fagerland
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Deborah K Hill
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Astrid Hyldbakk
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Jana Kim
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | |
Collapse
|
39
|
Rong N, Zhang M, Wang Y, Wu H, Qi H, Fu X, Li D, Yang C, Wang Y, Fan Z. Effects of extracellular matrix rigidity on sonoporation facilitated by targeted microbubbles: Bubble attachment, bubble dynamics, and cell membrane permeabilization. ULTRASONICS SONOCHEMISTRY 2020; 67:105125. [PMID: 32298974 DOI: 10.1016/j.ultsonch.2020.105125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/19/2020] [Accepted: 04/07/2020] [Indexed: 06/11/2023]
Abstract
In this study, we investigated the effects of extracellular matrix rigidity, an important physical property of microenvironments regulating cell morphology and functions, on sonoporation facilitated by targeted microbubbles, highlighting the role of microbubbles. We conducted mechanistic studies at the cellular level on physiologically relevant soft and rigid substrates. By developing a unique imaging strategy, we first resolved details of the 3D attachment configurations between targeted microbubbles and cell membrane. High-speed video microscopy then unveiled bubble dynamics driven by a single ultrasound pulse. Finally, we evaluated the cell membrane permeabilization using a small molecule model drug. Our results demonstrate that: (1) stronger targeted microbubble attachment was formed for cells cultured on the rigid substrate, while six different attachment configurations were revealed in total; (2) more violent bubble oscillation was observed for cells cultured on the rigid substrate, while one third of bubbles attached to cells on the soft substrate exhibited deformation shortly after ultrasound was turned off; (3) higher acoustic pressure was needed to permeabilize the cell membrane for cells on the soft substrate, while under the same ultrasound condition, acoustically-activated microbubbles generated larger pores as compared to cells cultured on the soft substrate. The current findings provide new insights to understand the underlying mechanisms of sonoporation in a physiologically relevant context and may be useful for the clinical translation of sonoporation.
Collapse
Affiliation(s)
- Ning Rong
- Department of Biomedical Engineering, School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Meiru Zhang
- Department of Biomedical Engineering, School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Yulin Wang
- Department of Biomedical Engineering, School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Hao Wu
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Hui Qi
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Xing Fu
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Dachao Li
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Chunmei Yang
- Department of Biomedical Engineering, School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Yan Wang
- Department of Biomedical Engineering, School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Zhenzhen Fan
- Department of Biomedical Engineering, School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China; State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
40
|
Ingram N, McVeigh LE, Abou-Saleh RH, Maynard J, Peyman SA, McLaughlan JR, Fairclough M, Marston G, Valleley EMA, Jimenez-Macias JL, Charalambous A, Townley W, Haddrick M, Wierzbicki A, Wright A, Volpato M, Simpson PB, Treanor DE, Thomson NH, Loadman PM, Bushby RJ, Johnson BR, Jones PF, Evans JA, Freear S, Markham AF, Evans SD, Coletta PL. Ultrasound-triggered therapeutic microbubbles enhance the efficacy of cytotoxic drugs by increasing circulation and tumor drug accumulation and limiting bioavailability and toxicity in normal tissues. Theranostics 2020; 10:10973-10992. [PMID: 33042265 PMCID: PMC7532679 DOI: 10.7150/thno.49670] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Most cancer patients receive chemotherapy at some stage of their treatment which makes improving the efficacy of cytotoxic drugs an ongoing and important goal. Despite large numbers of potent anti-cancer agents being developed, a major obstacle to clinical translation remains the inability to deliver therapeutic doses to a tumor without causing intolerable side effects. To address this problem, there has been intense interest in nanoformulations and targeted delivery to improve cancer outcomes. The aim of this work was to demonstrate how vascular endothelial growth factor receptor 2 (VEGFR2)-targeted, ultrasound-triggered delivery with therapeutic microbubbles (thMBs) could improve the therapeutic range of cytotoxic drugs. Methods: Using a microfluidic microbubble production platform, we generated thMBs comprising VEGFR2-targeted microbubbles with attached liposomal payloads for localised ultrasound-triggered delivery of irinotecan and SN38 in mouse models of colorectal cancer. Intravenous injection into tumor-bearing mice was used to examine targeting efficiency and tumor pharmacodynamics. High-frequency ultrasound and bioluminescent imaging were used to visualise microbubbles in real-time. Tandem mass spectrometry (LC-MS/MS) was used to quantitate intratumoral drug delivery and tissue biodistribution. Finally, 89Zr PET radiotracing was used to compare biodistribution and tumor accumulation of ultrasound-triggered SN38 thMBs with VEGFR2-targeted SN38 liposomes alone. Results: ThMBs specifically bound VEGFR2 in vitro and significantly improved tumor responses to low dose irinotecan and SN38 in human colorectal cancer xenografts. An ultrasound trigger was essential to achieve the selective effects of thMBs as without it, thMBs failed to extend intratumoral drug delivery or demonstrate enhanced tumor responses. Sensitive LC-MS/MS quantification of drugs and their metabolites demonstrated that thMBs extended drug exposure in tumors but limited exposure in healthy tissues, not exposed to ultrasound, by persistent encapsulation of drug prior to elimination. 89Zr PET radiotracing showed that the percentage injected dose in tumors achieved with thMBs was twice that of VEGFR2-targeted SN38 liposomes alone. Conclusions: thMBs provide a generic platform for the targeted, ultrasound-triggered delivery of cytotoxic drugs by enhancing tumor responses to low dose drug delivery via combined effects on circulation, tumor drug accumulation and exposure and altered metabolism in normal tissues.
Collapse
Affiliation(s)
- Nicola Ingram
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Laura E. McVeigh
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Radwa H. Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- Department of Physics, Faculty of Science, Mansoura University, Egypt
| | - Juliana Maynard
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - James R. McLaughlan
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Michael Fairclough
- Wolfson Molecular Imaging Centre, University of Manchester, Palatine Road, Manchester, M20 3LI, United Kingdom
| | - Gemma Marston
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Elizabeth M. A. Valleley
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Jorge L. Jimenez-Macias
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Antonia Charalambous
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - William Townley
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Antonia Wierzbicki
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Alexander Wright
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Milène Volpato
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Peter B. Simpson
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Darren E. Treanor
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Neil H. Thomson
- School of Dentistry, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Richard J. Bushby
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Benjamin R.G. Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - Pamela F. Jones
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - J. Anthony Evans
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Steven Freear
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Alexander F. Markham
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| |
Collapse
|
41
|
Al-Jawadi S, Thakur SS. Ultrasound-responsive lipid microbubbles for drug delivery: A review of preparation techniques to optimise formulation size, stability and drug loading. Int J Pharm 2020; 585:119559. [PMID: 32574685 DOI: 10.1016/j.ijpharm.2020.119559] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023]
Abstract
Lipid-shelled microbubbles have received extensive interest to enhance ultrasound-responsive drug delivery outcomes due to their high biocompatibility. While therapeutic effectiveness of microbubbles is well established, there remain limitations in sample homogeneity, stability profile and drug loading properties which restrict these formulations from seeing widespread use in the clinical setting. In this review, we evaluate and discuss the most encouraging leads in lipid microbubble design and optimisation. We examine current applications in drug delivery for the systems and subsequently detail shell compositions and preparation strategies that improve monodispersity while retaining ultrasound responsiveness. We review how excipients and storage techniques help maximise stability and introduce different characterisation and drug loading techniques and evaluate their impact on formulation performance. The review concludes with current quality control measures in place to ensure lipid microbubbles can be reproducibly used in drug delivery.
Collapse
Affiliation(s)
- Sana Al-Jawadi
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Sachin S Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
42
|
Kooiman K, Roovers S, Langeveld SAG, Kleven RT, Dewitte H, O'Reilly MA, Escoffre JM, Bouakaz A, Verweij MD, Hynynen K, Lentacker I, Stride E, Holland CK. Ultrasound-Responsive Cavitation Nuclei for Therapy and Drug Delivery. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1296-1325. [PMID: 32165014 PMCID: PMC7189181 DOI: 10.1016/j.ultrasmedbio.2020.01.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 05/03/2023]
Abstract
Therapeutic ultrasound strategies that harness the mechanical activity of cavitation nuclei for beneficial tissue bio-effects are actively under development. The mechanical oscillations of circulating microbubbles, the most widely investigated cavitation nuclei, which may also encapsulate or shield a therapeutic agent in the bloodstream, trigger and promote localized uptake. Oscillating microbubbles can create stresses either on nearby tissue or in surrounding fluid to enhance drug penetration and efficacy in the brain, spinal cord, vasculature, immune system, biofilm or tumors. This review summarizes recent investigations that have elucidated interactions of ultrasound and cavitation nuclei with cells, the treatment of tumors, immunotherapy, the blood-brain and blood-spinal cord barriers, sonothrombolysis, cardiovascular drug delivery and sonobactericide. In particular, an overview of salient ultrasound features, drug delivery vehicles, therapeutic transport routes and pre-clinical and clinical studies is provided. Successful implementation of ultrasound and cavitation nuclei-mediated drug delivery has the potential to change the way drugs are administered systemically, resulting in more effective therapeutics and less-invasive treatments.
Collapse
Affiliation(s)
- Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Silke Roovers
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Simone A G Langeveld
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert T Kleven
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Medical School of the Vrije Universiteit Brussel, Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Martin D Verweij
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Christy K Holland
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA; Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
43
|
Mohamed TMD, Ji-Bin LMD, John REP. Recent Advances in Microbubble-Augmented Cancer Therapy. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2020. [DOI: 10.37015/audt.2020.200055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|