1
|
Xing L, Zhang J, Gao Y, Jiang M, Zhao L, Guan S. Experimental Study of Gas-Liquid Compound Droplet Impact on Superhydrophobic Surface. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:4270-4286. [PMID: 39899656 DOI: 10.1021/acs.langmuir.4c04983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
The impact dynamics of gas-liquid compound droplets (GLCD) on surfaces play a crucial role in optimizing processes such as atomization, coating, and microencapsulation. The dynamic behaviors of GLCD impacting superhydrophobic surfaces under varying impact heights, viscosities, and gas-liquid volume ratios (Φ) were investigated via high-speed photography. Three collision categories are defined according to GLCD morphology evolution under different parameters, i.e., oscillation-breakup, rebound collision, and impact-breakup. As the impact height increases, the occurrence of droplet breakup shifts progressively from the retraction stage to the spreading stage. Notably, the maximum rebound coefficient of the compound droplet initially increases and then decreases with increasing impact height, and finally increases again due to the bubble breakup, which differs from the behavior of the homogeneous droplets. Furthermore, the maximum spreading and rebound heights of the GLCD are suppressed by increasing the liquid-phase viscosity. For Φ = 0.48 GLCD, increasing the liquid-phase viscosity from 4.7 to 5.5 mPa·s raises the critical breakup impact height from 90 mm to 120 mm. Meanwhile, the average dimensionless breakup time also increases from 0.54 to 1.72. As the Φ increases, the liquid film of the GLCD gradually thins, resulting in a decrease in the maximum spreading coefficient and enhanced probability of droplet breakup. Additionally, increasing the Φ from 0.48 to 0.56 leads to an increase in the average dimensionless breakup time by 0.88. This study provides a fundamental understanding of the dynamic behavior of compound droplets and offer valuable insights for related engineering applications.
Collapse
Affiliation(s)
- Lei Xing
- School of Mechanical Science and Engineering, Northeast Petroleum University, Daqing 163318, Heilongjiang, China
- Postdoctoral Research Station of Daqing Oilfield, Daqing 163458, Heilongjiang, China
- Key Laboratory of Petroleum and Petrochemical Multiphase Media Treatment and Pollution Prevention, Daqing 163318, Heilongjiang, China
| | - Jinming Zhang
- School of Mechanical Science and Engineering, Northeast Petroleum University, Daqing 163318, Heilongjiang, China
| | - Yang Gao
- PetroChina Research Institute of Petroleum Exploration and Development, Beijing 100083, China
- State Key Laboratory of Continental Shale Oil, Beijing 100083, China
| | - Minghu Jiang
- School of Mechanical Science and Engineering, Northeast Petroleum University, Daqing 163318, Heilongjiang, China
- Key Laboratory of Petroleum and Petrochemical Multiphase Media Treatment and Pollution Prevention, Daqing 163318, Heilongjiang, China
| | - Lixin Zhao
- School of Mechanical Science and Engineering, Northeast Petroleum University, Daqing 163318, Heilongjiang, China
- Key Laboratory of Petroleum and Petrochemical Multiphase Media Treatment and Pollution Prevention, Daqing 163318, Heilongjiang, China
| | - Shuai Guan
- School of Mechanical Science and Engineering, Northeast Petroleum University, Daqing 163318, Heilongjiang, China
- Key Laboratory of Petroleum and Petrochemical Multiphase Media Treatment and Pollution Prevention, Daqing 163318, Heilongjiang, China
| |
Collapse
|
2
|
Chen G, Li B, Li T, Lin M, Zhong H, Xie X, Zhang Q, Chen Q, Meng X, Xiao Z, Shuai X. Core-Satellite Nanoassembly Overcomes Spatial Heterogeneity of Dendric Cell Distribution in Pancreatic Tumors for Effective Chemoimmunotherapy. ACS NANO 2025; 19:4739-4753. [PMID: 39834130 DOI: 10.1021/acsnano.4c15444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Pancreatic cancer therapies such as chemotherapy and immunotherapy are hindered by the dense extracellular matrix known as physical barriers, leading to heterogeneity impeding the effective penetration of chemotherapeutic agents and activation of antitumor immune responses. To address this challenge, we developed a hybrid nanoassembly with a distinct core-satellite-like heterostructure, PLAF@P/T-PD, which is responsive to both internal pH/redox and external ultrasound stimulations. This heterostructural nanoassembly features a polymersome core encapsulating an ultrasound contrast agent perfluoropentane and a chemotherapeutic agent Taxol (PLAF@P/T) electrostatically coated with satellite-like polyplexes carrying an immune agonist dsDNA (PD), which brings about synergistic functions inside the pancreatic tumor. The PLAF@P/T core functions as an enhancer for intratumor delivery through size enlargement and charge conversion in response to reactive oxygen species (ROS) and low pH, which triggers polyplex release and enables ultrasound-assisted tumor-penetrating Taxol delivery. Meanwhile, the released cationic polyplexes function as nucleic nanomedicine preferentially engulfed by peripheral dendritic cells (DCs) for immune modulation. Animal studies in mouse orthotopic pancreatic tumor model demonstrated exceptional therapeutic efficacy against both primary and metastatic tumors, which underlines the potential of this heterostructural nanoplatform for overcoming the therapeutic challenges associated with the heterogeneous physical barrier hindering intratumor drug delivery in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Gengjia Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
- Department of Radiology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Bo Li
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Tan Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Minzhao Lin
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Huihai Zhong
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiaoxue Xie
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiaoyun Zhang
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Qi Chen
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaochun Meng
- Department of Radiology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
3
|
Wilson MG, Riis TS, Kubanek J. Controlled ultrasonic interventions through the human skull. Front Hum Neurosci 2024; 18:1412921. [PMID: 38979100 PMCID: PMC11228146 DOI: 10.3389/fnhum.2024.1412921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Transcranial focused ultrasound enables precise and non-invasive manipulations of deep brain circuits in humans, promising to provide safe and effective treatments of various neurological and mental health conditions. Ultrasound focused to deep brain targets can be used to modulate neural activity directly or localize the release of psychoactive drugs. However, these applications have been impeded by a key barrier-the human skull, which attenuates ultrasound strongly and unpredictably. To address this issue, we have developed an ultrasound-based approach that directly measures and compensates for the ultrasound attenuation by the skull. No additional skull imaging, simulations, assumptions, or free parameters are necessary; the method measures the attenuation directly by emitting a pulse of ultrasound from an array on one side of the head and measuring with an array on the opposite side. Here, we apply this emerging method to two primary future uses-neuromodulation and local drug release. Specifically, we show that the correction enables effective stimulation of peripheral nerves and effective release of propofol from nanoparticle carriers through an ex vivo human skull. Neither application was effective without the correction. Moreover, the effects show the expected dose-response relationship and targeting specificity. This article highlights the need for precise control of ultrasound intensity within the skull and provides a direct and practical approach for addressing this lingering barrier.
Collapse
Affiliation(s)
- Matthew G Wilson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Thomas S Riis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Jan Kubanek
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
4
|
Hallam KA, Nikolai RJ, Jhunjhunwala A, Emelianov SY. Laser-activated perfluorocarbon nanodroplets for intracerebral delivery and imaging via blood-brain barrier opening and contrast-enhanced imaging. J Nanobiotechnology 2024; 22:356. [PMID: 38902773 PMCID: PMC11191388 DOI: 10.1186/s12951-024-02601-6] [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: 02/07/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Ultrasound and photoacoustic (US/PA) imaging is a promising tool for in vivo visualization and assessment of drug delivery. However, the acoustic properties of the skull limit the practical application of US/PA imaging in the brain. To address the challenges in targeted drug delivery to the brain and transcranial US/PA imaging, we introduce and evaluate an intracerebral delivery and imaging strategy based on the use of laser-activated perfluorocarbon nanodroplets (PFCnDs). METHODS Two specialized PFCnDs were developed to facilitate blood‒brain barrier (BBB) opening and contrast-enhanced US/PA imaging. In mice, PFCnDs were delivered to brain tissue via PFCnD-induced BBB opening to the right side of the brain. In vivo, transcranial US/PA imaging was performed to evaluate the utility of PFCnDs for contrast-enhanced imaging through the skull. Ex vivo, volumetric US/PA imaging was used to characterize the spatial distribution of PFCnDs that entered brain tissue. Immunohistochemical analysis was performed to confirm the spatial extent of BBB opening and the accuracy of the imaging results. RESULTS In vivo, transcranial US/PA imaging revealed localized photoacoustic (PA) contrast associated with delivered PFCnDs. In addition, contrast-enhanced ultrasound (CEUS) imaging confirmed the presence of nanodroplets within the same area. Ex vivo, volumetric US/PA imaging revealed PA contrast localized to the area of the brain where PFCnD-induced BBB opening had been performed. Immunohistochemical analysis revealed that the spatial distribution of immunoglobulin (IgG) extravasation into the brain closely matched the imaging results. CONCLUSIONS Using our intracerebral delivery and imaging strategy, PFCnDs were successfully delivered to a targeted area of the brain, and they enabled contrast-enhanced US/PA imaging through the skull. Ex vivo imaging, and immunohistochemistry confirmed the accuracy and precision of the approach.
Collapse
Affiliation(s)
- Kristina A Hallam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert J Nikolai
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Anamik Jhunjhunwala
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Stanislav Y Emelianov
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA.
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
5
|
Wilson MG, Parikh A, Dara A, Beaver AS, Kubanek J. Targeted drug release from stable and safe ultrasound-sensitive nanocarriers. Front Mol Biosci 2024; 11:1408767. [PMID: 38962281 PMCID: PMC11219560 DOI: 10.3389/fmolb.2024.1408767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/17/2024] [Indexed: 07/05/2024] Open
Abstract
Targeted delivery of medication has the promise of increasing the effectiveness and safety of current systemic drug treatments. Focused ultrasound is emerging as noninvasive and practical energy for targeted drug release. However, it has yet to be determined which nanocarriers and ultrasound parameters can provide both effective and safe release. Perfluorocarbon nanodroplets have the potential to achieve these goals, but current approaches have either been effective or safe, but not both. We found that nanocarriers with highly stable perfluorocarbon cores mediate effective drug release so long as they are activated by ultrasound of sufficiently low frequency. We demonstrate a favorable safety profile of this formulation in a non-human primate. To facilitate translation of this approach into humans, we provide an optimized method for manufacturing the nanocarriers. This study provides a recipe and release parameters for effective and safe drug release from nanoparticle carriers in the body part specified by focused ultrasonic waves.
Collapse
Affiliation(s)
- Matthew G. Wilson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | | | | | | | | |
Collapse
|
6
|
Wilson MG, Webb TD, Odéen H, Kubanek J. Remotely controlled drug release in deep brain regions of non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.09.561539. [PMID: 37873134 PMCID: PMC10592699 DOI: 10.1101/2023.10.09.561539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Many areas of science and medicine would benefit from selective release of drugs in specific regions of interest. Nanoparticle drug carriers activated by focused ultrasound-remotely applied, depth-penetrating energy-may provide such selective interventions. Here, we developed stable, ultrasound-responsive nanoparticles that can be used to release drugs effectively and safely in non-human primates. The nanoparticles were used to release propofol in deep brain visual regions. The release reversibly modulated the subjects' visual choice behavior and was specific to the targeted region and to the released drug. Gadolinium-enhanced MRI imaging suggested an intact blood-brain barrier. Blood draws showed normal clinical chemistry and hematology. In summary, this study provides a safe and effective approach to release drugs on demand in selected deep brain regions at levels sufficient to modulate behavior.
Collapse
|
7
|
Van Namen A, Jandhyala S, Spatarelu CP, Tichauer KM, Samkoe KS, Luke GP. Multiplex Ultrasound Imaging of Perfluorocarbon Nanodroplets Enabled by Decomposition of Postvaporization Dynamics. NANO LETTERS 2024; 24:209-214. [PMID: 38156794 DOI: 10.1021/acs.nanolett.3c03719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Despite the real-time, nonionizing, and cost-effective nature of ultrasound imaging, there is a dearth of methods to visualize two or more populations of contrast agents simultaneously─a technique known as multiplex imaging. Here, we present a new approach to multiplex ultrasound imaging using perfluorocarbon (PFC) nanodroplets. The nanodroplets, which undergo a liquid-to-gas phase transition in response to an acoustic trigger, act as activatable contrast agents. This work characterized the dynamic responses of two PFC nanodroplets with boiling points of 28 and 56 °C. These characteristic responses were then used to demonstrate that the relative concentrations of the two populations of PFC nanodroplets could be accurately measured in the same imaging volume within an average error of 1.1%. Overall, the findings indicate the potential of this approach for multiplex ultrasound imaging, allowing for the simultaneous visualization of multiple molecular targets simultaneously.
Collapse
Affiliation(s)
- Austin Van Namen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Sidhartha Jandhyala
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | | | - Kenneth M Tichauer
- Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616, United States
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
- Translational Engineering in Cancer Research Program, Dartmouth Cancer Center, Lebanon, New Hampshire 03766, United States
| | - Geoffrey P Luke
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
- Translational Engineering in Cancer Research Program, Dartmouth Cancer Center, Lebanon, New Hampshire 03766, United States
| |
Collapse
|
8
|
Wang W, Shi Y, Chai W, Kevin Tang KW, Pyatnitskiy I, Xie Y, Liu X, He W, Jeong J, Hsieh JC, Lozano AR, Artman B, Henkelman G, Chen B, Wang H. Ultrasound programmable hydrogen-bonded organic frameworks for sono-chemogenetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570721. [PMID: 38106007 PMCID: PMC10723392 DOI: 10.1101/2023.12.08.570721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The precise control of mechanochemical activation within deep tissues via non-invasive ultrasound holds profound implications for advancing our understanding of fundamental biomedical sciences and revolutionizing disease treatments. However, a theory-guided mechanoresponsive materials system with well-defined ultrasound activation has yet to be explored. Here we present the concept of using porous hydrogen-bonded organic frameworks (HOFs) as toolkits for focused ultrasound programmably triggered drug activation to control specific cellular events in the deep brain, through on-demand scission of the supramolecular interactions. A theoretical model is developed to visualize the mechanochemical scission and ultrasound mechanics, providing valuable guidelines for the rational design of mechanoresponsive materials at the molecular level to achieve programmable and spatiotemporal activation control. To demonstrate the practicality of this approach, we encapsulate designer drug clozapine N-oxide (CNO) into the optimal HOF nanoparticles for FUS gated release to activate engineered G-protein-coupled receptors in the mice and rat ventral tegmental area (VTA), and hence achieved targeted neural circuits modulation even at depth 9 mm with a latency of seconds. This work demonstrates the capability of ultrasound to precisely control molecular interaction and develops ultrasound programmable HOFs to minimally invasive and spatiotemporally control cellular events, thereby facilitating the establishment of precise molecular therapeutic possibilities. We anticipate that this research could serve as a source of inspiration for precise and non-invasive molecular manipulation techniques, potentially applicable in programming molecular robots to achieve sophisticated control over cellular events in deep tissues.
Collapse
|
9
|
Purohit MP, Roy KS, Xiang Y, Yu BJ, Azadian MM, Muwanga G, Hart AR, Taoube AK, Lopez DG, Airan RD. Acoustomechanically activatable liposomes for ultrasonic drug uncaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563690. [PMID: 37961368 PMCID: PMC10634775 DOI: 10.1101/2023.10.23.563690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Ultrasound-activatable drug-loaded nanocarriers enable noninvasive and spatiotemporally-precise on-demand drug delivery throughout the body. However, most systems for ultrasonic drug uncaging utilize cavitation or heating as the drug release mechanism and often incorporate relatively exotic excipients into the formulation that together limit the drug-loading potential, stability, and clinical translatability and applicability of these systems. Here we describe an alternate strategy for the design of such systems in which the acoustic impedance and osmolarity of the internal liquid phase of a drug-loaded particle is tuned to maximize ultrasound-induced drug release. No gas phase, cavitation, or medium heating is necessary for the drug release mechanism. Instead, a non-cavitation-based mechanical response to ultrasound mediates the drug release. Importantly, this strategy can be implemented with relatively common pharmaceutical excipients, as we demonstrate here by implementing this mechanism with the inclusion of a few percent sucrose into the internal buffer of a liposome. Further, the ultrasound protocols sufficient for in vivo drug uncaging with this system are achievable with current clinical therapeutic ultrasound systems and with intensities that are within FDA and society guidelines for safe transcranial ultrasound application. Finally, this current implementation of this mechanism should be versatile and effective for the loading and uncaging of any therapeutic that may be loaded into a liposome, as we demonstrate for four different drugs in vitro, and two in vivo. These acoustomechanically activatable liposomes formulated with common pharmaceutical excipients promise a system with high clinical translational potential for ultrasonic drug uncaging of myriad drugs of clinical interest.
Collapse
Affiliation(s)
| | - Kanchan Sinha Roy
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Yun Xiang
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Brenda J. Yu
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Biophysics Program, Stanford University, Stanford, CA, 94305 USA
| | - Matine M. Azadian
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Neurosciences Program, Stanford University, Stanford, CA, 94305 USA
| | - Gabriella Muwanga
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Neurosciences Program, Stanford University, Stanford, CA, 94305 USA
| | - Alex R. Hart
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Chemistry, Stanford University, Stanford, CA, 94305 USA
| | - Ali K. Taoube
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
| | - Diego Gomez Lopez
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, TN 37235 USA
| | - Raag D. Airan
- Department of Radiology, Stanford University, Stanford, CA, 94305 USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305 USA
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305 USA
| |
Collapse
|
10
|
Aliabouzar M, Kripfgans OD, Brian Fowlkes J, Fabiilli ML. Bubble nucleation and dynamics in acoustic droplet vaporization: a review of concepts, applications, and new directions. Z Med Phys 2023; 33:387-406. [PMID: 36775778 PMCID: PMC10517405 DOI: 10.1016/j.zemedi.2023.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 02/12/2023]
Abstract
The development of phase-shift droplets has broadened the scope of ultrasound-based biomedical applications. When subjected to sufficient acoustic pressures, the perfluorocarbon phase in phase-shift droplets undergoes a phase-transition to a gaseous state. This phenomenon, termed acoustic droplet vaporization (ADV), has been the subject of substantial research over the last two decades with great progress made in design of phase-shift droplets, fundamental physics of bubble nucleation and dynamics, and applications. Here, we review experimental approaches, carried out via high-speed microscopy, as well as theoretical models that have been proposed to study the fundamental physics of ADV including vapor nucleation and ADV-induced bubble dynamics. In addition, we highlight new developments of ADV in tissue regeneration, which is a relatively recently exploited application. We conclude this review with future opportunities of ADV for advanced applications such as in situ microrheology and pressure estimation.
Collapse
Affiliation(s)
- Mitra Aliabouzar
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Oliver D Kripfgans
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Applied Physics Program, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - J Brian Fowlkes
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Applied Physics Program, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Applied Physics Program, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
12
|
Spatarelu CP, Jandhyala S, Luke GP. Dual-drug loaded ultrasound-responsive nanodroplets for on-demand combination chemotherapy. ULTRASONICS 2023; 133:107056. [PMID: 37269682 DOI: 10.1016/j.ultras.2023.107056] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/13/2023] [Accepted: 05/23/2023] [Indexed: 06/05/2023]
Abstract
Phase-changing nanodroplets are nanometric sized constructs that can be vaporized via external stimuli, such as focused ultrasound, to generate gaseous bubbles that are visible in ultrasound. Their activation can also be leveraged to release their payload, creating a method for ultrasound-modulated localized drug delivery. Here, we develop a perfluoropentane core nanodroplet that can simultaneously load paclitaxel and doxorubicin, and release them in response to an acoustic trigger. A double emulsion method is used to incorporate the two drugs with different physio-chemical properties, which allows for a combinatorial chemotherapy regimen to be used. Their loading, release, and biological effects on a triple negative breast cancer mouse model are investigated. We show that activation enhances the drug-delivery effect and delays the tumor growth rate in vivo. Overall, the phase-changing nanodroplets are a useful platform to allow on-demand delivery of combinations of drugs.
Collapse
Affiliation(s)
- Catalina-Paula Spatarelu
- Thayer School of Engineering, Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, United States
| | - Sidhartha Jandhyala
- Thayer School of Engineering, Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, United States
| | - Geoffrey P Luke
- Thayer School of Engineering, Dartmouth College, 15 Thayer Drive, Hanover, NH 03755, United States; Translational Engineering in Cancer Research Program, Dartmouth Cancer Center, 1 Medical Center Drive, Lebanon, NH 03766, United States.
| |
Collapse
|
13
|
Pereira LF, Dallagnol CA, Moulepes TH, Hirota CY, Kutsmi P, dos Santos LV, Pirich CL, Picheth GF. Oxygen therapy alternatives in COVID-19: From classical to nanomedicine. Heliyon 2023; 9:e15500. [PMID: 37089325 PMCID: PMC10106793 DOI: 10.1016/j.heliyon.2023.e15500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023] Open
Abstract
Around 10-15% of COVID-19 patients affected by the Delta and the Omicron variants exhibit acute respiratory insufficiency and require intensive care unit admission to receive advanced respiratory support. However, the current ventilation methods display several limitations, including lung injury, dysphagia, respiratory muscle atrophy, and hemorrhage. Furthermore, most of the ventilatory techniques currently offered require highly trained professionals and oxygen cylinders, which may attain short supply owing to the high demand and misuse. Therefore, the search for new alternatives for oxygen therapeutics has become extremely important for maintaining gas exchange in patients affected by COVID-19. This review highlights and suggest new alternatives based on micro and nanostructures capable of supplying oxygen and/or enabling hematosis during moderate or acute COVID-19 cases.
Collapse
Affiliation(s)
- Luis F.T. Pereira
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Camila A. Dallagnol
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Tassiana H. Moulepes
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Clara Y. Hirota
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Pedro Kutsmi
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
| | - Lucas V. dos Santos
- Department of Biochemistry, Federal University of Paraná, Curitiba, PR, Brazil
| | - Cleverton L. Pirich
- Department of Bioprocess Engineering and Biotechnology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Guilherme F. Picheth
- School of Medicine, Pontifical Catholic University of Paraná, Curitiba, PR, Brazil
- Department of Biochemistry, Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
14
|
Edwards IA, De Carlo F, Sitta J, Varner W, Howard CM, Claudio PP. Enhancing Targeted Therapy in Breast Cancer by Ultrasound-Responsive Nanocarriers. Int J Mol Sci 2023; 24:ijms24065474. [PMID: 36982548 PMCID: PMC10053544 DOI: 10.3390/ijms24065474] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Currently, the response to cancer treatments is highly variable, and severe side effects and toxicity are experienced by patients receiving high doses of chemotherapy, such as those diagnosed with triple-negative breast cancer. The main goal of researchers and clinicians is to develop new effective treatments that will be able to specifically target and kill tumor cells by employing the minimum doses of drugs exerting a therapeutic effect. Despite the development of new formulations that overall can increase the drugs’ pharmacokinetics, and that are specifically designed to bind overexpressed molecules on cancer cells and achieve active targeting of the tumor, the desired clinical outcome has not been reached yet. In this review, we will discuss the current classification and standard of care for breast cancer, the application of nanomedicine, and ultrasound-responsive biocompatible carriers (micro/nanobubbles, liposomes, micelles, polymeric nanoparticles, and nanodroplets/nanoemulsions) employed in preclinical studies to target and enhance the delivery of drugs and genes to breast cancer.
Collapse
Affiliation(s)
- Isaiah A. Edwards
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Flavia De Carlo
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Juliana Sitta
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William Varner
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Candace M. Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Pier Paolo Claudio
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Correspondence:
| |
Collapse
|
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
|
Xiao L, Wu Y, Dai J, Zhang W, Cao Y. Laser-activated nanoparticles for ultrasound/photoacoustic imaging-guided prostate cancer treatment. Front Bioeng Biotechnol 2023; 11:1141984. [PMID: 37025361 PMCID: PMC10070956 DOI: 10.3389/fbioe.2023.1141984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/13/2023] [Indexed: 04/08/2023] Open
Abstract
Prostate cancer (PCa) is the most common malignant tumor in men. Prostate-specific membrane antigen (PSMA), which is overexpressed on the surface of Prostate cancer cells, may serve as a potential therapeutic target. Recently, image-guided and targeted therapy for prostate cancers has attracted much attention by using Prostate-specific membrane antigen targeting nanoparticle. In this study, we produced PSMA-targeted light-responsive nanosystems. These nanosystems of liquid perfluorocarbon cores and polymer shells were loaded with the photosensitizer IR780 and therapeutic drugs paclitaxel. The liquid perfluorocarbon (PFP) in nanoparticles can perform ultrasound-enhanced imaging by liquid-gas transition and promote the deliver and release of paclitaxel. IR780 can perform photothermal therapy (PTT) guided by photoacoustic (PA) imaging. Combination treatment with photothermal therapy and chemotherapy exhibited excellent inhibition of cell proliferation in vitro and a significant therapeutic effect in vivo. In conclusion, we successfully formulated PSMA-targeted nanosystems with precision targeting and ultrasound/PA dual-modality imaging for anti-tumor effects.
Collapse
Affiliation(s)
- Linkang Xiao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Urology Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Chongqing General Hospital, Chongqing, China
| | - Yunfang Wu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Urology Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Chongqing Wanzhou District Maternal and Child Health Hospital, Chongqing, China
| | - Junyong Dai
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Urology Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Chongqing University Cancer Hospital, Chongqing, China
| | - Weili Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Urology Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- *Correspondence: Weili Zhang, ; Yang Cao,
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Urology Surgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- *Correspondence: Weili Zhang, ; Yang Cao,
| |
Collapse
|
17
|
Huang S, Guo W, An J, Zhang J, Dong F, Wang D, Feng F, Zhang J. Enhanced Acoustic Droplet Vaporization through the Active Magnetic Accumulation of Drug-Loaded Magnetic Particle-Encapsulated Nanodroplets (MPE-NDs) in Cancer Therapy. NANO LETTERS 2022; 22:8143-8151. [PMID: 36194752 DOI: 10.1021/acs.nanolett.2c02580] [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] [Indexed: 06/16/2023]
Abstract
The application of drug-loaded nanodroplets is still limited by their insufficient accumulation owing to the enhanced permeability and retention (EPR) effect failure in cancer therapy. To overcome these limitations, we propose an alternative magnetic particle-encapsulated nanodroplet (MPE-ND) with outstanding biosafety and magnetic targeting by encapsulating fluorinated Fe3O4-SiO2 nanoparticles inside the liquid core of the nanodroplets. Meanwhile, doxorubicin (DOX) can be stably loaded into the shell through both electrostatic and hydrophobic interactions to obtain drug-loaded MPE-NDs. Both in vitro and in vivo experiments have consistently demonstrated that drug-loaded MPE-NDs can significantly increase the local drug concentration and enhance the damage of tumor tissues through acoustic droplet vaporization under a static magnetic field (eADV therapy). Histological examination reveals that eADV therapy efficiently suppresses tumor proliferation by inducing apoptosis, destroying supply vessels, and inhibiting neovascularization. Drug-loaded MPE-NDs can be expected to open a new gateway for ultrasound-triggered drug delivery and cancer treatment.
Collapse
Affiliation(s)
- Shuo Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Wenyu Guo
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jian An
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jiabin Zhang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, College of Future Technology, College of Future Technology, Peking University, Beijing, 100871, China
| | - Feihong Dong
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, College of Future Technology, College of Future Technology, Peking University, Beijing, 100871, China
| | - Di Wang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Feng Feng
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- College of Engineering, Peking University, Beijing, 100871, China
- National Biomedical Imaging Center, Peking University, Beijing, 100871, China
| |
Collapse
|
18
|
Ma P, Lai X, Luo Z, Chen Y, Loh XJ, Ye E, Li Z, Wu C, Wu YL. Recent advances in mechanical force-responsive drug delivery systems. NANOSCALE ADVANCES 2022; 4:3462-3478. [PMID: 36134346 PMCID: PMC9400598 DOI: 10.1039/d2na00420h] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/15/2022] [Indexed: 06/16/2023]
Abstract
Mechanical force responsive drug delivery systems (in terms of mechanical force induced chemical bond breakage or physical structure destabilization) have been recently explored to exhibit a controllable pharmaceutical release behaviour at a molecular level. In comparison with chemical or biological stimulus triggers, mechanical force is not only an external but also an internal stimulus which is closely related to the physiological status of patients. However, although this mechanical force stimulus might be one of the most promising and feasible sources to achieve on-demand pharmaceutical release, current research in this field is still limited. Hence, this tutorial review aims to comprehensively evaluate the recent advances in mechanical force-responsive drug delivery systems based on different types of mechanical force, in terms of direct stimulation by compressive, tensile, and shear force, or indirect/remote stimulation by ultrasound and a magnetic field. Furthermore, the exciting developments and current challenges in this field will also be discussed to provide a blueprint for potential clinical translational research of mechanical force-responsive drug delivery systems.
Collapse
Affiliation(s)
- Panqin Ma
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 China
| | - Xiyu Lai
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 China
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 China
| | - Ying Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 China
| | - Xian Jun Loh
- Institute of Materials Research and Engineering, ASTAR (Agency for Science, Technology and Research) 2 Fusionopolis Way Innovis, #08-03 138634 Singapore
| | - Enyi Ye
- Institute of Materials Research and Engineering, ASTAR (Agency for Science, Technology and Research) 2 Fusionopolis Way Innovis, #08-03 138634 Singapore
| | - Zibiao Li
- Institute of Materials Research and Engineering, ASTAR (Agency for Science, Technology and Research) 2 Fusionopolis Way Innovis, #08-03 138634 Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2) Agency for Science, Technology, and Research (ASTAR) Singapore 138634 Singapore
- Department of Materials Science and Engineering, National University of Singapore 9 Engineering Drive 1 Singapore 117576 Singapore
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University Xiamen 361102 China
| |
Collapse
|
19
|
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
|
20
|
Zhang S, Zhang S, Luo S, Tang P, Wan M, Wu D, Gao W. Ultrasound-assisted brain delivery of nanomedicines for brain tumor therapy: advance and prospect. J Nanobiotechnology 2022; 20:287. [PMID: 35710426 PMCID: PMC9205090 DOI: 10.1186/s12951-022-01464-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/18/2022] [Indexed: 12/14/2022] Open
Abstract
Nowadays, brain tumors are challenging problems, and the key of therapy is ensuring therapeutic drugs cross the blood-brain barrier (BBB) effectively. Although the efficiency of drug transport across the BBB can be increased by innovating and modifying nanomedicines, they exert insufficient therapeutic effects on brain tumors due to the complex environment of the brain. It is worth noting that ultrasound combined with the cavitation effect of microbubbles can assist BBB opening and enhance brain delivery of nanomedicines. This ultrasound-assisted brain delivery (UABD) technology with related nanomedicines (UABD nanomedicines) can safely open the BBB, facilitate the entry of drugs into the brain, and enhance the therapeutic effect on brain tumors. UABD nanomedicines, as the main component of UABD technology, have great potential in clinical application and have been an important area of interest in the field of brain tumor therapy. However, research on UABD nanomedicines is still in its early stages despite the fact that they have been associated with many disciplines, including material science, brain science, ultrasound, biology, and medicine. Some aspects of UABD theory and technology remain unclear, especially the mechanisms of BBB opening, relationship between materials of nanomedicines and UABD technology, cavitation and UABD nanomedicines design theories. This review introduces the research status of UABD nanomedicines, investigates their properties and applications of brain tumor therapy, discusses the advantages and drawbacks of UABD nanomedicines for the treatment of brain tumors, and offers their prospects. We hope to encourage researchers from various fields to participate in this area and collaborate on developing UABD nanomedicines into powerful tools for brain tumor therapy.
Collapse
Affiliation(s)
- Shuo Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shuai Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Siyuan Luo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Peng Tang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| | - Wei Gao
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
21
|
Lee H, Partanen M, Lee M, Jeong S, Lee HJ, Kim K, Ryu W, Dholakia K, Oh K. A laser-driven optical atomizer: photothermal generation and transport of zeptoliter-droplets along a carbon nanotube deposited hollow optical fiber. NANOSCALE 2022; 14:5138-5146. [PMID: 35302135 DOI: 10.1039/d1nr06211e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
From mechanical syringes to electric field-assisted injection devices, precise control of liquid droplet generation has been sought after, and the present state-of-the-art technologies have provided droplets ranging from nanoliter to subpicoliter volume sizes. In this study, we present a new laser-driven method to generate liquid droplets with a zeptoliter volume, breaking the fundamental limits of previous studies. We guided an infrared laser beam through a hollow optical fiber (HOF) with a ring core whose end facet was coated with single-walled carbon nanotubes. The laser light was absorbed by this nanotube film and efficiently generated a highly localized microring heat source. This evaporated the liquid inside the HOF, which rapidly recondensed into zeptoliter droplets in the surrounding air at room temperature. We spectroscopically confirmed the chemical structures of the liquid precursor maintained in the droplets by atomizing dye-dissolved glycerol. Moreover, we explain the fundamental physical principles as well as functionalities of the optical atomizer and perform a detailed characterization of the droplets. Our approach has strong prospects for nanoscale delivery of biochemical substances in minuscule zeptoliter volumes.
Collapse
Affiliation(s)
- Hyeonwoo Lee
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Mikko Partanen
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
- Photonics Group, Department of Electronics and Nanoengineering, Aalto University, P.O. Box 13500, 00076 Aalto, Finland
| | - Mingyu Lee
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Sunghoon Jeong
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Hyeung Joo Lee
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Kwanpyo Kim
- Department of Physics, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| | - Wonhyoung Ryu
- Biomedical and Energy System Laboratory, Department of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| | - Kishan Dholakia
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
- SUPA, School of Physics and Astronomy, University of St Andrews, KY16 9SS, UK.
| | - Kyunghwan Oh
- Photonic Device Physics Laboratory, Department of Physics, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea.
| |
Collapse
|
22
|
Zhang W, Shi Y, Abd Shukor S, Vijayakumaran A, Vlatakis S, Wright M, Thanou M. Phase-shift nanodroplets as an emerging sonoresponsive nanomaterial for imaging and drug delivery applications. NANOSCALE 2022; 14:2943-2965. [PMID: 35166273 DOI: 10.1039/d1nr07882h] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanodroplets - emerging phase-changing sonoresponsive materials - have attracted substantial attention in biomedical applications for both tumour imaging and therapeutic purposes due to their unique response to ultrasound. As ultrasound is applied at different frequencies and powers, nanodroplets have been shown to cavitate by the process of acoustic droplet vapourisation (ADV), causing the development of mechanical forces which promote sonoporation through cellular membranes. This allows drugs to be delivered efficiently into deeper tissues where tumours are located. Recent reviews on nanodroplets are mostly focused on the mechanism of cavitation and their applications in biomedical fields. However, the chemistry of the nanodroplet components has not been discussed or reviewed yet. In this review, the commonly used materials and preparation methods of nanodroplets are summarised. More importantly, this review provides examples of variable chemistry components in nanodroplets which link them to their efficiency as ultrasound-multimodal imaging agents to image and monitor drug delivery. Finally, the drawbacks of current research, future development, and future direction of nanodroplets are discussed.
Collapse
Affiliation(s)
- Weiqi Zhang
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | - Yuhong Shi
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | | | | | - Stavros Vlatakis
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | - Michael Wright
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| | - Maya Thanou
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| |
Collapse
|
23
|
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: 68] [Impact Index Per Article: 17.0] [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
|
24
|
Tehrani Fateh S, Moradi L, Kohan E, Hamblin MR, Shiralizadeh Dezfuli A. Comprehensive review on ultrasound-responsive theranostic nanomaterials: mechanisms, structures and medical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:808-862. [PMID: 34476167 PMCID: PMC8372309 DOI: 10.3762/bjnano.12.64] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/15/2021] [Indexed: 05/03/2023]
Abstract
The field of theranostics has been rapidly growing in recent years and nanotechnology has played a major role in this growth. Nanomaterials can be constructed to respond to a variety of different stimuli which can be internal (enzyme activity, redox potential, pH changes, temperature changes) or external (light, heat, magnetic fields, ultrasound). Theranostic nanomaterials can respond by producing an imaging signal and/or a therapeutic effect, which frequently involves cell death. Since ultrasound (US) is already well established as a clinical imaging modality, it is attractive to combine it with rationally designed nanoparticles for theranostics. The mechanisms of US interactions include cavitation microbubbles (MBs), acoustic droplet vaporization, acoustic radiation force, localized thermal effects, reactive oxygen species generation, sonoluminescence, and sonoporation. These effects can result in the release of encapsulated drugs or genes at the site of interest as well as cell death and considerable image enhancement. The present review discusses US-responsive theranostic nanomaterials under the following categories: MBs, micelles, liposomes (conventional and echogenic), niosomes, nanoemulsions, polymeric nanoparticles, chitosan nanocapsules, dendrimers, hydrogels, nanogels, gold nanoparticles, titania nanostructures, carbon nanostructures, mesoporous silica nanoparticles, fuel-free nano/micromotors.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lida Moradi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Kohan
- Department of Science, University of Kurdistan, Kurdistan, Sanandaj, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | | |
Collapse
|
25
|
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
|
26
|
Drug Delivery by Ultrasound-Responsive Nanocarriers for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13081135. [PMID: 34452096 PMCID: PMC8397943 DOI: 10.3390/pharmaceutics13081135] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Conventional cancer chemotherapies often exhibit insufficient therapeutic outcomes and dose-limiting toxicity. Therefore, there is a need for novel therapeutics and formulations with higher efficacy, improved safety, and more favorable toxicological profiles. This has promoted the development of nanomedicines, including systems for drug delivery, but also for imaging and diagnostics. Nanoparticles loaded with drugs can be designed to overcome several biological barriers to improving efficiency and reducing toxicity. In addition, stimuli-responsive nanocarriers are able to release their payload on demand at the tumor tissue site, preventing premature drug loss. This review focuses on ultrasound-triggered drug delivery by nanocarriers as a versatile, cost-efficient, non-invasive technique for improving tissue specificity and tissue penetration, and for achieving high drug concentrations at their intended site of action. It highlights aspects relevant for ultrasound-mediated drug delivery, including ultrasound parameters and resulting biological effects. Then, concepts in ultrasound-mediated drug delivery are introduced and a comprehensive overview of several types of nanoparticles used for this purpose is given. This includes an in-depth compilation of the literature on the various in vivo ultrasound-responsive drug delivery systems. Finally, toxicological and safety considerations regarding ultrasound-mediated drug delivery with nanocarriers are discussed.
Collapse
|
27
|
Gupta I, Su X, Jonnalagadda US, Das D, Pramanik M, Kwan JJ. Investigating the Acoustic Response and Contrast Enhancement of Drug-Loadable PLGA Microparticles with Various Shapes and Morphologies. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1844-1856. [PMID: 33810888 DOI: 10.1016/j.ultrasmedbio.2021.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/07/2021] [Accepted: 02/16/2021] [Indexed: 06/12/2023]
Abstract
Polymer nanoparticles and microparticles have been used primarily for drug delivery. There is now growing interest in further developing polymer-based solid cavitation agents to also enhance ultrasound imaging. We previously reported on a facile method to produce hollow poly(lactic-co-glycolic acid) (PLGA) microparticles with different diameters and degrees of porosity. Here, we investigate the cavitation response from these PLGA microparticles with both therapeutic and diagnostic ultrasound transducers. Interestingly, all formulations exhibited stable cavitation; larger porous and multicavity particles also provided inertial cavitation at elevated acoustic pressure amplitudes. These larger particles also achieved contrast enhancement comparable to that of commercially available ultrasound contrast agents, with a maximum recorded contrast-to-tissue ratio of 28 dB. Therefore, we found that multicavity PLGA microparticles respond to both therapeutic and diagnostic ultrasound and may be applied as a theranostic agent.
Collapse
Affiliation(s)
- Ipshita Gupta
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - Xiaoqian Su
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - Umesh Sai Jonnalagadda
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - Dhiman Das
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - Manojit Pramanik
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - James J Kwan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459.
| |
Collapse
|
28
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
29
|
Snipstad S, Hanstad S, Bjørkøy A, Mørch Ý, de Lange Davies C. Sonoporation Using Nanoparticle-Loaded Microbubbles Increases Cellular Uptake of Nanoparticles Compared to Co-Incubation of Nanoparticles and Microbubbles. Pharmaceutics 2021; 13:640. [PMID: 33946327 PMCID: PMC8146007 DOI: 10.3390/pharmaceutics13050640] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022] Open
Abstract
Therapeutic agents can benefit from encapsulation in nanoparticles, due to improved pharmacokinetics and biodistribution, protection from degradation, increased cellular uptake and sustained release. Microbubbles in combination with ultrasound have been shown to improve the delivery of nanoparticles and drugs to tumors and across the blood-brain barrier. Here, we evaluate two different microbubbles for enhancing the delivery of polymeric nanoparticles to cells in vitro: a commercially available lipid microbubble (Sonazoid) and a microbubble with a shell composed of protein and nanoparticles. Various ultrasound parameters are applied and confocal microscopy is employed to image cellular uptake. Ultrasound enhanced cellular uptake depending on the pressure and duty cycle. The responsible mechanisms are probably sonoporation and sonoprinting, followed by uptake, and to a smaller degree enhanced endocytosis. The use of commercial Sonazoid microbubbles leads to significantly lower uptake than when using nanoparticle-loaded microbubbles, suggesting that proximity between cells, nanoparticles and microbubbles is important, and that mainly nanoparticles in the shell are taken up, rather than free nanoparticles in solution.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
- Cancer Clinic, St. Olav’s Hospital, Prinsesse Kristinas Gate 1, 7030 Trondheim, Norway
| | - Sigurd Hanstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| |
Collapse
|
30
|
Lawanprasert A, Chau A, Sloand JN, Hannifin S, Medina SH. Tuning the Interfacial Properties of Fluorous Colloids Toward Ultrasound Programmable Bioactivity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:5989-5998. [PMID: 33522791 DOI: 10.1021/acsami.0c20352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Liquid-in-liquid emulsions are kinetically stable colloids that undergo liquid-to-gas phase transitions in response to thermal or acoustic stimuli. Perfluorocarbons (PFCs) are preferred species as their highly fluorinated nature imparts unique properties that are unparalleled by nonfluorinated counterparts. However, traditional methods to prepare PFC emulsions lack the ability to precisely tune the thermodynamic stability of the fluorous-water interphase and consequently control their vaporization behavior. Here, we report a privileged fluoroalkanoic acid that undergoes concentration-dependent assembly on the surfaces of fluorous droplets to modulate interfacial tension. This allows for the rational formulation of orthogonal PFC droplets that can be programmed to vaporize at specified ultrasound powers. We exploit this behavior in two exemplary biomedical settings by developing emulsions that aid ultrasound-mediated hemostasis and enable bioorthogonal delivery of molecular sensors to mammalian cells. Mechanistic insights gained from these studies can be generalized to tune the thermodynamic interfacial equilibria of PFC emulsions toward designing controllable tools for precision medicine.
Collapse
Affiliation(s)
- Atip Lawanprasert
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Alda Chau
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Janna N Sloand
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| | - Sean Hannifin
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
- Immunology Graduate Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Scott H Medina
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
- Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
31
|
Trout CJ, Clapp JA, Griepenburg JC. Plasmonic carriers responsive to pulsed laser irradiation: a review of mechanisms, design, and applications. NEW J CHEM 2021. [DOI: 10.1039/d1nj02062e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review focuses on interactions which govern release from plasmonic carrier systems including liposomes, polymersomes, and nanodroplets under pulsed irradiation.
Collapse
Affiliation(s)
- Cory J. Trout
- Department of Physics, Rutgers University-Camden, 227 Penn Street, Camden, NJ 08102, USA
- Department of Applied Physics, Rutgers University-Newark, 101 Warren St., Newark, NJ 07102, USA
| | - Jamie A. Clapp
- Center for Computational and Integrative Biology, Rutgers University-Camden, NJ 08102, USA
| | - Julianne C. Griepenburg
- Department of Physics, Rutgers University-Camden, 227 Penn Street, Camden, NJ 08102, USA
- Center for Computational and Integrative Biology, Rutgers University-Camden, NJ 08102, USA
| |
Collapse
|
32
|
Liao AH, Lin WT, Chen HK, Shih CP, Wang CH, Chu YH. Synergistic effects of combined treatment with ultrasound-mediated cisplatin-loaded microbubbles and atorvastatin on head and neck cancer. Head Neck 2020; 43:15-26. [PMID: 32954561 DOI: 10.1002/hed.26445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/19/2020] [Accepted: 08/14/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Previously, we used ultrasound (US)-mediated cisplatin (CDDP)-loaded microbubbles (CDDP-MBs) to increase intratumoral CDDP level while decreasing systemic cytotoxicity. Statins have shown antitumorigenic properties. Our study investigated the effects of atorvastatin with CDDP-MBs and US on head neck cancer. METHODS Cell viability analysis with CDDP-MBs and atorvastatin combined with US in FaDu cell line were tested. Cell proliferation and glutathione level were also evaluated. RESULTS Both CDDP and atorvastatin reduced cell's viability. Coadministration of CDDP and atorvastatin resulted in synergistic inhibitory effect. After US sonication, cell viability with atorvastatin and CDDP was significantly reduced for CDDP combined with MBs (65.98% to 49.13%) and for CDDP-MBs (86.17% to 50.15%). CDDP-MBs combined with atorvastatin and US inhibited the proliferation of cells: 19.61% for CDDP-MBs + atorvastatin + US, 36.28% for CDDP + atorvastatin, and 71.73% for atorvastatin alone. Also, CDDP-MBs + atorvastatin + US induced apoptosis by decreasing cellular level of glutathione. CONCLUSIONS Atorvastatin combined with MB-conjugated CDDP exerts synergistic inhibitory effect on head neck cancer.
Collapse
Affiliation(s)
- Ai-Ho Liao
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan.,Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Wan-Ting Lin
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Hang-Kang Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Ping Shih
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Hung Wang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Yueng-Hsiang Chu
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
33
|
Melich R, Bussat P, Morici L, Vivien A, Gaud E, Bettinger T, Cherkaoui S. Microfluidic preparation of various perfluorocarbon nanodroplets: Characterization and determination of acoustic droplet vaporization (ADV) threshold. Int J Pharm 2020; 587:119651. [DOI: 10.1016/j.ijpharm.2020.119651] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022]
|
34
|
Yoshinaga K, Delage-Laurin L, Swager TM. Fluorous phthalocyanines and subphthalocyanines. J PORPHYR PHTHALOCYA 2020. [DOI: 10.1142/s1088424620500182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Incorporating fluorine atoms into a molecule can endow it with various unique properties that enable materials applications. Selective solubility in fluorous solvents is achieved by a high fluorine content and selective partitioning into perfluorinated liquids over organic and aqueous phases provides orthogonal opportunities for chemistry and materials assembly. Although there is a growing number of partially fluorinated molecules, there are insufficient structural design principles to produce diverse fluorous soluble dyes. Herein, we report the synthesis of six fluorous phthalocyanine and subphthalocyanine dyes, and study their properties in the fluorous phase. Phthalocyanines generally display limited solubility and we also observed apparent aggregation in the fluorous phase. However, the nonplanar subphthalocyanines showed greater solubility. Subphthalocyanines also displayed fluorescence in selected solvents, and their emissive properties were investigated. The materials described expand the library of fluorous dyes and provide insights for the design of new molecules with fluorous solubility.
Collapse
Affiliation(s)
- Kosuke Yoshinaga
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Leo Delage-Laurin
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Timothy M. Swager
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| |
Collapse
|
35
|
Zhu Y, Zhang G, Li M, Ma L, Huang J, Qiu L. Ultrasound-Augmented Phase Transition Nanobubbles for Targeted Treatment of Paclitaxel-Resistant Cancer. Bioconjug Chem 2020; 31:2008-2020. [PMID: 32628454 DOI: 10.1021/acs.bioconjchem.0c00364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yi Zhu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Ultrasound, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Guonan Zhang
- Department of Gynecological Oncology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Meiying Li
- Department of Biochemistry & Molecular Biology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Lang Ma
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianming Huang
- Department of Biochemistry & Molecular Biology, the Affiliated Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Cancer Hospital & Institute, Chengdu 610041, China
| | - Li Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
36
|
Sloand JN, Nguyen TT, Zinck SA, Cook EC, Zimudzi TJ, Showalter SA, Glick AB, Simon JC, Medina SH. Ultrasound-Guided Cytosolic Protein Delivery via Transient Fluorous Masks. ACS NANO 2020; 14:4061-4073. [PMID: 32134630 DOI: 10.1021/acsnano.9b08745] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The inability to spatiotemporally guide proteins in tissues and efficiently deliver them into cells remains a key barrier to realizing their full potential in precision medicine. Here, we report ultrasound-sensitive fluoro-protein nanoemulsions which can be acoustically tracked, guided, and activated for on-demand cytosolic delivery of proteins, including antibodies, using clinically relevant diagnostic ultrasound. This advance is accessed through the discovery of a family of fluorous tags, or FTags, that transiently mask proteins to mediate their efficient dispersion into ultrasound-sensitive liquid perfluorocarbons, a phenomenon akin to dissolving an egg in liquid Teflon. We identify the biochemical basis for protein fluorous masking and confirm FTag coatings are shed during delivery, without disrupting the protein structure or function. Harnessing the ultrasound sensitivity of fluorous emulsions, real-time imaging is used to simultaneously monitor and activate FTag-protein complexes to enable controlled cytosolic antibody delivery in vitro and in vivo. These findings may advance the development of image-guided, protein-based biosensing and therapeutic modalities.
Collapse
Affiliation(s)
- Janna N Sloand
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Theodore T Nguyen
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Scott A Zinck
- Graduate Program in Acoustics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Erik C Cook
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Tawanda J Zimudzi
- Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Scott A Showalter
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Adam B Glick
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Julianna C Simon
- Graduate Program in Acoustics, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Scott H Medina
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
37
|
Lambert E, Gorantla VS, Janjic JM. Pharmaceutical design and development of perfluorocarbon nanocolloids for oxygen delivery in regenerative medicine. Nanomedicine (Lond) 2019; 14:2697-2712. [PMID: 31657273 DOI: 10.2217/nnm-2019-0260] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Perfluorocarbons (PFCs) have been investigated as oxygen carriers for several decades in varied biomedical applications. PFCs are chemically and biologically inert, temperature and storage stable, pose low to no infectious risk, can be commercially manufactured, and have well established gas transport properties. In this review, we highlight design and development strategies for their successful application in regenerative medicine, transplantation and organ preservation. Effective tissue preservation strategies are key to improving outcomes of extremity salvage and organ transplantation. Maintaining tissue integrity requires adequate oxygenation to support aerobic metabolism. The use of whole blood for oxygen delivery is fraught with limitations of poor shelf stability, infectious risk, religious exclusions and product shortages. Other agents also face clinical challenges in their implementation. As a solution, we discuss new ways of designing and developing PFC-based artificial oxygen carriers by implementing modern pharmaceutical quality by design and scale up manufacturing methodologies.
Collapse
Affiliation(s)
- Eric Lambert
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.,Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA 15282, USA
| | - Vijay S Gorantla
- Department of Surgery, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA.,AIRMED Program, 59th Medical Wing, United States Air Force, United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Jelena M Janjic
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.,Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA 15282, USA.,AIRMED Program, 59th Medical Wing, United States Air Force, United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| |
Collapse
|
38
|
Basha SA, Salkho N, Dalibalta S, Husseini GA. Liposomes in Active, Passive and Acoustically-Triggered Drug Delivery. Mini Rev Med Chem 2019; 19:961-969. [PMID: 30961495 DOI: 10.2174/1389557519666190408155251] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 02/17/2018] [Accepted: 11/06/2018] [Indexed: 12/23/2022]
Abstract
Cancer has become one of the most deadly noncommunicable diseases globally. Several modalities used to treat cancer patients exist today yet many have failed to prove high efficacy with low side effects. The most common example of such modalities is the use of chemotherapeutic drugs to treat cancerous cells and deter their uncontrolled proliferation. In addition to the destruction of cancerous tissues, chemotherapy destroys healthy tissues as it lacks the specificity to annihilate cancerous cells only and preferentially, which result in adverse side effects including nausea, hair fall and myocardial infarction. To prevent the side effects of non-selective chemotherapy, cancer therapy research has been focused on the implementation of nanocarrier systems that act as vehicles to encapsulate drugs and selectively transport their agent to the tumor site. In this paper, we shed light on liposomes along with three anticancer drug delivery approaches: passive, active and ultrasound-triggered drug delivery.
Collapse
Affiliation(s)
- Sara Al Basha
- Department of Chemistry, Biology and Environmental Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| | - Najla Salkho
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Sarah Dalibalta
- Department of Chemistry, Biology and Environmental Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb Adnan Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
39
|
Carvalho CR, Silva-Correia J, Oliveira JM, Reis RL. Nanotechnology in peripheral nerve repair and reconstruction. Adv Drug Deliv Rev 2019; 148:308-343. [PMID: 30639255 DOI: 10.1016/j.addr.2019.01.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/20/2018] [Accepted: 01/05/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Cristiana R Carvalho
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joaquim M Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, AvePark, 4805-017 Barco, Guimarães, Portugal.
| |
Collapse
|
40
|
Nevozhay D, Weiger M, Friedl P, Sokolov KV. Spatiotemporally controlled nano-sized third harmonic generation agents. BIOMEDICAL OPTICS EXPRESS 2019; 10:3301-3316. [PMID: 31360600 PMCID: PMC6640828 DOI: 10.1364/boe.10.003301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 05/09/2023]
Abstract
Here, we present a new class of third harmonic generation (THG) imaging probes that can be activated with precise spatiotemporal control using non-linear excitation. These probes consist of lipid-coated perfluorocarbon nanodroplets with embedded visible chromophores. The droplets undergo phase transition from liquid to gas upon heating mediated by two-photon absorption of NIR light by the embedded dyes. Resulting microbubbles provide a sharp, local refractive index mismatch, which makes an excellent source of THG signal. Potential applications of these probes include activatable THG agents for biological imaging and "on-demand" delivery of various compounds under THG monitoring.
Collapse
Affiliation(s)
- Dmitry Nevozhay
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- School of Biomedicine, Far Eastern Federal University, 8 Sukhanova Street, Vladivostok, 690950, Russia
- Equal contribution
| | - Michael Weiger
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Equal contribution
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Cancer Genomics Centre, (CGC.nl), 3584 Utrecht, Netherlands
| | - Konstantin V. Sokolov
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
- Department of Bioengineering, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street, Austin, TX 78712, USA
| |
Collapse
|
41
|
Hallam KA, Emelianov SY. Toward optimization of blood brain barrier opening induced by laser-activated perfluorocarbon nanodroplets. BIOMEDICAL OPTICS EXPRESS 2019; 10:3139-3151. [PMID: 31360596 PMCID: PMC6640833 DOI: 10.1364/boe.10.003139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 05/09/2023]
Abstract
The blood brain barrier (BBB), a component of the brain's natural defense system, is often a roadblock for the monitoring and treatment of neurological disorders. Recently, we introduced a technique to open the blood brain barrier through the use of laser-activated perfluorohexane nanodroplets (PFHnDs), a phase-change nanoagent that undergoes repeated vaporization and recondensation when excited by a pulsed laser. Laser-activated PFHnDs were shown to enable noninvasive and localized opening of the BBB, allowing extravasation of various sized agents into the brain tissue. In this current work, the laser-activated PFHnD-induced BBB opening is further explored. In particular, laser fluence and the number of laser pulses used for the PFHnD-induced BBB opening are examined and evaluated both qualitatively and quantitatively to determine the effect of these parameters on BBB opening. The results of these studies show trends between increased laser fluence and an increased BBB opening as well as between an increased number of laser pulses and an increased BBB opening, however, with limitations on the extent of the BBB opening after a certain number of pulses. Overall, the results of these studies serve as a guideline to choosing suitable laser parameters for safe and effective BBB opening.
Collapse
Affiliation(s)
- Kristina A. Hallam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Stanislav Y. Emelianov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
42
|
Synchronized Optical and Acoustic Droplet Vaporization for Effective Sonoporation. Pharmaceutics 2019; 11:pharmaceutics11060279. [PMID: 31197090 PMCID: PMC6631315 DOI: 10.3390/pharmaceutics11060279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 01/05/2023] Open
Abstract
Inertial cavitation-based sonoporation has been utilized to enhance treatment delivery efficacy. In our previous study, we demonstrated that tumor therapeutic efficacy can be enhanced through vaporization-assisted sonoporation with gold nanodroplets (AuNDs). Specifically, the AuNDs were vaporized both acoustically (i.e., acoustic droplet vaporization, ADV) and optically (i.e., optical droplet vaporization, ODV). A continuous wave (CW) laser was used for ODV in combination with an ultrasound pulse for ADV. Although effective for vaporization, the use of a CW laser is not energy efficient and may create unwanted heating and concomitant tissue damage. In this study, we propose the use of a pulsed wave (PW) laser to replace the CW laser. In addition, the PW laser was applied at the rarefaction phase of the ultrasound pulse so that the synergistic effects of ADV and ODV can be expected. Therefore, a significantly lower laser average power can be expected to achieve the vaporization threshold. Compared to the CW laser power at 2 W/cm2 from the previous approach, the PW laser power was reduced to only 0.2404 W/cm2. Furthermore, we also demonstrate in vitro that the sonoporation rate was increased when the PW laser was applied at the rarefaction phase. Specifically, the vaporization signal, the inertial cavitation signal, and the sonoporation rate all displayed a 1-µs period, which corresponded to the period of the 1-MHz acoustic wave used for ADV, as a function of the relative laser delay. The increased sonoporation rate indicates that this technique has the potential to enhance sonoporation-directed drug delivery and tumor therapy with a lower laser power while keeping the cell death rate at the minimum. Photoacoustic imaging can also be performed at the same time since a PW laser is used for the ODV.
Collapse
|
43
|
Zhang Y, Yong L, Luo Y, Ding X, Xu D, Gao X, Yan S, Wang Q, Luo J, Pu D, Zou J. Enhancement of HIFU ablation by sonosensitizer-loading liquid fluorocarbon nanoparticles with pre-targeting in a mouse model. Sci Rep 2019; 9:6982. [PMID: 31061456 PMCID: PMC6502828 DOI: 10.1038/s41598-019-43416-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
High intensity focused ultrasound (HIFU) is a noninvasive thermal ablation technique for the treatment of benign and malignant solid masses. To improve the efficacy of HIFU ablation, we developed poly (lactide-co-glycolide) (PLGA) nanoparticles encapsulating perfluoropentane (PFP) and hematoporphyrin monomethyl ether (HMME) as synergistic agents (HMME+PFP/PLGA). Two-step biotin-avidin pre-targeting technique was applied for the HIFU ablation. We further modified the nanoparticles with streptavidin (HMME+PFP/PLGA-SA). HMME+PFP/PLGA-SA were highly dispersed with spherical morphology (477.8 ± 81.8 nm in diameter). The encapsulation efficiency of HMME and PFP were 46.6 ± 3.3% and 40.1 ± 2.6%, respectively. The binding efficiency of nanoparticles to streptavidin was 95.5 ± 2.5%. The targeting ability of the HMME+PFP/PLGA-SA nanoparticles was tested by parallel plate flow chamber in vitro. In the pre-targeting group (HMME+PFP/PLGA-SA), a large number of nanoparticles bound to the peripheral and surface of the cell. In the HIFU ablation experiment in vivo, compared with the other groups, the largest gray-scale changes and coagulation necrosis areas were observed in the pre-targeting (HMME+PFP/PLGA-SA) group, with the lowest energy efficiency factor value. Moreover, the microvessel density and proliferation index declined, while the apoptotic index increased, in the tumor tissue surrounding the coagulation necrosis area in the pre-targeting group. Meanwhile, the survival time of the tumor-bearing nude mice in the pre-targeting group was significantly longer than that in the HIFU treatment group. These results suggest that HMME+PFP/PLGA-SA have high potential to act as synergistic agents in HIFU ablation.
Collapse
Affiliation(s)
- Yong Zhang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Lijun Yong
- Department of Obstetrics, Chongqing Health Center for Women and Children, Chongqing, 401147, China
| | - Yong Luo
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
| | - Xiaoya Ding
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
| | - Die Xu
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
| | - Xuan Gao
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
| | - Sijing Yan
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
- Department of Ultrasound, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Qi Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China
| | - Jie Luo
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Darong Pu
- Department of Ultrasound, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Jianzhong Zou
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing Collaborative Innovation Center for Minimally-invasive and Noninvasive Medicine, Chongqing, 400016, China.
| |
Collapse
|
44
|
Yang Q, Li P, Ran H, Wan J, Chen H, Chen H, Wang Z, Zhang L. Polypyrrole-coated phase-change liquid perfluorocarbon nanoparticles for the visualized photothermal-chemotherapy of breast cancer. Acta Biomater 2019; 90:337-349. [PMID: 30936037 DOI: 10.1016/j.actbio.2019.03.056] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
Abstract
A theranostic nanoplatform (DTX/PFH@PPy-FA) for multi-modal imaging-guided photothermal-chemotherapy has been constructed. Lipid-perfluorohexane (PFH) nanodroplet loaded with docetaxel (DTX) was coated with a polypyrrole (PPy) shell. Then the folic acid (FA) molecule with active tumor-targeting function was modified on the surface of PPy shell. Due to the good photothermal conversion performance, PPy shell can raise the temperature under the near infrared laser irradiation, which not only produces photothermal effect to kill tumor cells, but also promotes liquid-gas phase change of PFH, and produces ultrasound imaging effect. The results of photothermal experiment and imaging experiment confirmed that the obtained DTX/PFH@PPy-FA possessed good photothermal, photoacoustic imaging and ultrasound imaging effects in vitro and in vivo. The results of in vitro cell experiments showed that DTX/PFH@PPy-FA had a active targeting ability to tumor cells, and its photothermal-chemotherapy synergistically inhibited the proliferation of tumor cells. In vivo study on 4T1-bearing BALB/c mice indicated that the photothermal-chemotherapy of DTX/PFH@PPy-FA not only effectively inhibited the growth of 4T1 breast cancer, but also inhibited lung metastasis. This multifunctional nanoparticle is expected to become a new nanoplatform for the visualized photothermal-chemotherapy of cancer. STATEMENT OF SIGNIFICANCE: In this work, we presented a multi-modal imaging-guided photothermal-chemotherapy theranostic nanoplatform (DTX/PFH@PPy-FA) for visualized treatment of breast cancer. The docetaxel (DTX) loaded perfluorohexane (PFH) nanodroplets (DTX/PFH@SPC) were firstly prepared and then coated with polypyrrole shell (PPy). Then, PEGylated folic acid was covalently modified to obtain the folate-targeted multifunctional nanoparticle (DTX/PFH@PPy-FA). Due to the good photothermal conversion performance, PPy shell can raise the temperature under the near infrared laser irradiation, which not only produces photothermal effect to kill tumor cells, but also promotes liquid-gas phase change of PFH, and produces good ultrasound imaging effect. The PPy shell also imparts photoacoustic imaging characteristics to the nanoparticles. Experimental results show that our prepared DTX/PFH@PPy-FA possesses folic acid-mediated tumor targeting ability, ultrasound and photoacoustic imaging, and photothermal-chemotherapy synergistic effect. This multi-functional nanoparticle is expected to become a new platform for the visualized photothermal-chemotherapy of breast cancer.
Collapse
Affiliation(s)
- Qiang Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Chongqing Medical University, Chongqing 400016, PR China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Chongqing Medical University, Chongqing 400016, PR China
| | - Jingyuan Wan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Huan Chen
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Huali Chen
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Chongqing Medical University, Chongqing 400016, PR China
| | - Liangke Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
45
|
Santiesteban DY, Hallam KA, Yarmoska SK, Emelianov SY. Color-coded perfluorocarbon nanodroplets for multiplexed ultrasound and Photoacoustic imaging. NANO RESEARCH 2019; 12:741-747. [PMID: 31572565 PMCID: PMC6768563 DOI: 10.1007/s12274-019-2279-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/19/2018] [Accepted: 12/25/2018] [Indexed: 05/19/2023]
Abstract
Laser-activated perfluorocarbon nanodroplets are an emerging class of phase change, dual-contrast agents that can be utilized in ultrasound and photoacoustic imaging. Through the ability to differentiate subpopulations of nanodroplets via laser activation at different wavelengths of near-infrared light, optically-triggered color-coded perfluorocarbon nanodroplets present themselves as an attractive tool for multiplexed ultrasound and photoacoustic imaging. In particular, laser-activated droplets can be used to provide quantitative spatiotemporal information regarding distinct biological targets, allowing for their potential use in a wide range of diagnos tic and therapeutic applications. In the work presented, laser-activated color-coded perfluorocarbon nanodroplets are synthesized to selectively respond to laser irradiation at corresponding wavelengths. The dynamic ultrasound and photoacoustic signals produced by laser-activated perfluorocarbon nanodroplets are evaluated in situ prior to implementation in a murine model. In vivo, these particles are used to distinguish unique particle trafficking mechanisms and are shown to provide ultrasound and photoacoustic contrast for up to 72 hours within lymphatics. Overall, the conducted studies show that laser-activated color-coded perfluorocarbon nanodroplets are a promising agent for multiplexed ultrasound and photoacoustic imaging.
Collapse
Affiliation(s)
- Daniela Y. Santiesteban
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Kristina A. Hallam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Steven K. Yarmoska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Stanislav Y. Emelianov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
- School of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Corresponding author,
| |
Collapse
|
46
|
Mannaris C, Bau L, Grundy M, Gray M, Lea-Banks H, Seth A, Teo B, Carlisle R, Stride E, Coussios CC. Microbubbles, Nanodroplets and Gas-Stabilizing Solid Particles for Ultrasound-Mediated Extravasation of Unencapsulated Drugs: An Exposure Parameter Optimization Study. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:954-967. [PMID: 30655109 DOI: 10.1016/j.ultrasmedbio.2018.10.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 06/09/2023]
Abstract
Ultrasound-induced cavitation has been proposed as a strategy to tackle the challenge of inadequate extravasation, penetration and distribution of therapeutics into tumours. Here, the ability of microbubbles, droplets and solid gas-trapping particles to facilitate mass transport and extravasation of a model therapeutic agent following ultrasound-induced cavitation is investigated. Significant extravasation and penetration depths on the order of millimetres are achieved with all three agents, including the range of pressures and frequencies achievable with existing clinical ultrasound systems. Deeper but highly directional extravasation was achieved with frequencies of 1.6 and 3.3 MHz compared with 0.5 MHz. Increased extravasation was observed with increasing pulse length and exposure time, while an inverse relationship is observed with pulse repetition frequency. No significant cell death or any haemolytic activity in human blood was observed at clinically relevant concentrations for any of the agents. Overall, solid gas-trapping nanoparticles were found to enable the most extensive extravasation for the lowest input acoustic energy, followed by microbubbles and then droplets. The ability of these agents to produce sustained inertial cavitation activity whilst being small enough to follow the drug out of the circulation and into diseased tissue, combined with a good safety profile and the possibility of real-time monitoring, offers considerable potential for enhanced drug delivery of unmodified drugs in oncological and other biomedical applications.
Collapse
Affiliation(s)
- Christophoros Mannaris
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Megan Grundy
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Harriet Lea-Banks
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Anjali Seth
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Boon Teo
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom
| | - Constantin C Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Headington, Oxford, United Kingdom.
| |
Collapse
|
47
|
Zhong Q, Yoon BC, Aryal M, Wang JB, Ilovitsh T, Baikoghli MA, Hosseini-Nassab N, Karthik A, Cheng RH, Ferrara KW, Airan RD. Polymeric perfluorocarbon nanoemulsions are ultrasound-activated wireless drug infusion catheters. Biomaterials 2019; 206:73-86. [PMID: 30953907 DOI: 10.1016/j.biomaterials.2019.03.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 01/04/2023]
Abstract
Catheter-based intra-arterial drug therapies have proven effective for a range of oncologic, neurologic, and cardiovascular applications. However, these procedures are limited by their invasiveness and relatively broad drug spatial distribution. The ideal technique for local pharmacotherapy would be noninvasive and would flexibly deliver a given drug to any region of the body with high spatial and temporal precision. Combining polymeric perfluorocarbon nanoemulsions with existent clinical focused ultrasound systems could in principle meet these needs, but it has not been clear whether these nanoparticles could provide the necessary drug loading, stability, and generalizability across a range of drugs, beyond a few niche applications. Here, we develop polymeric perfluorocarbon nanoemulsions into a generalized platform for ultrasound-targeted delivery of hydrophobic drugs with high potential for clinical translation. We demonstrate that a wide variety of drugs may be effectively uncaged with ultrasound using these nanoparticles, with drug loading increasing with hydrophobicity. We also set the stage for clinical translation by delineating production protocols that are scalable and yield sterile, stable, and optimized ultrasound-activated drug-loaded nanoemulsions. Finally, we exhibit a new potential application of these nanoemulsions for local control of vascular tone. This work establishes the power of polymeric perfluorocarbon nanoemulsions as a clinically-translatable platform for efficacious, noninvasive, and localized ultrasonic drug uncaging for myriad targets in the brain and body.
Collapse
Affiliation(s)
- Q Zhong
- Department of Radiology, Stanford University, Stanford, CA 94305, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - B C Yoon
- Department of Radiology, Stanford University, Stanford, CA 94305, USA; Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - M Aryal
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - J B Wang
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - T Ilovitsh
- Department of Radiology, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - M A Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - N Hosseini-Nassab
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - A Karthik
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - R H Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - K W Ferrara
- Department of Radiology, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R D Airan
- Department of Radiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
48
|
Lea-Banks H, O'Reilly MA, Hynynen K. Ultrasound-responsive droplets for therapy: A review. J Control Release 2019; 293:144-154. [PMID: 30503398 PMCID: PMC6459400 DOI: 10.1016/j.jconrel.2018.11.028] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/21/2022]
Abstract
The last two decades have seen the development of acoustically activated droplets, also known as phase-change emulsions, from a diagnostic tool to a therapeutic agent. Through bubble effects and triggered drug release, these superheated agents have found potential applications from oncology to neuromodulation. The aim of this review is to summarise the key developments in therapeutic droplet design and use, to discuss the current challenges slowing clinical translation, and to highlight the new frontiers progressing towards clinical implementation. The literature is summarised by addressing the droplet design criteria and by carrying out a multiparametric study of a range of droplet formulations and their associated vaporisation thresholds.
Collapse
Affiliation(s)
- H Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| | - M A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - K Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
49
|
Ho YJ, Wu CC, Hsieh ZH, Fan CH, Yeh CK. Thermal-sensitive acoustic droplets for dual-mode ultrasound imaging and drug delivery. J Control Release 2018; 291:26-36. [DOI: 10.1016/j.jconrel.2018.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 12/23/2022]
|
50
|
Snipstad S, Sulheim E, de Lange Davies C, Moonen C, Storm G, Kiessling F, Schmid R, Lammers T. Sonopermeation to improve drug delivery to tumors: from fundamental understanding to clinical translation. Expert Opin Drug Deliv 2018; 15:1249-1261. [PMID: 30415585 DOI: 10.1080/17425247.2018.1547279] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Ultrasound in combination with microbubbles can make cells and tissues more accessible for drugs, thereby achieving improved therapeutic outcomes. In this review, we introduce the term 'sonopermeation', covering mechanisms such as pore formation (traditional sonoporation), as well as the opening of intercellular junctions, stimulated endocytosis/transcytosis, improved blood vessel perfusion and changes in the (tumor) microenvironment. Sonopermeation has gained a lot of interest in recent years, especially for delivering drugs through the otherwise impermeable blood-brain barrier, but also to tumors. AREAS COVERED In this review, we summarize various in vitro assays and in vivo setups that have been employed to unravel the fundamental mechanisms involved in ultrasound-enhanced drug delivery, as well as clinical trials that are ongoing in patients with brain, pancreatic, liver and breast cancer. We summarize the basic principles of sonopermeation, describe recent findings obtained in (pre-) clinical trials, and discuss future directions. EXPERT OPINION We suggest that an improved mechanistic understanding, and microbubbles and ultrasound equipment specialized for drug delivery (and not for imaging) are key aspects to create more effective treatment regimens by sonopermeation. Real-time feedback and tools to predict therapeutic outcome and which tumors/patients will benefit from sonopermeation-based interventions will be important to promote clinical translation.
Collapse
Affiliation(s)
- Sofie Snipstad
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Einar Sulheim
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway.,b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,c Cancer Clinic , St. Olavs Hospital , Trondheim , Norway
| | - Catharina de Lange Davies
- a Department of Physics , Norwegian University of Science and Technology (NTNU) , Trondheim , Norway
| | - Chrit Moonen
- d Imaging Division , University Medical Center , Utrecht , The Netherlands
| | - Gert Storm
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands
| | - Fabian Kiessling
- g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| | - Ruth Schmid
- b Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway
| | - Twan Lammers
- e Department of Pharmaceutics , Utrecht University , Utrecht , The Netherlands.,f Department of Targeted Therapeutics , University of Twente , Enschede , The Netherlands.,g Institute for Experimental Molecular Imaging , RWTH Aachen University , Aachen , Germany
| |
Collapse
|