1
|
Abeid BA, Fabiilli ML, Aliabouzar M, Estrada JB. Experimental & numerical investigations of ultra-high-speed dynamics of optically induced droplet cavitation in soft materials. J Mech Behav Biomed Mater 2024; 160:106776. [PMID: 39488187 DOI: 10.1016/j.jmbbm.2024.106776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/26/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Perfluorocarbon (PFC) droplets represent a novel class of phase-shift contrast agent with promise in applications in biomedical and bioengineering fields. PFC droplets undergo a fast liquid-gas transition upon exposure to acoustic or optical triggering, offering a potential adaptable and versatile tool as contrast agent in diagnostic imaging and localized drug delivery vehicles in therapeutics systems. In this paper, we utilize advanced imaging techniques to investigate ultra-high-speed inertial dynamics and rectified quasi-static (low-speed) diffusion evolution of optically induced PFC droplet vaporization within three different hydrogels, each of different concentrations, examining effects such as droplet size and PFC core on bubble dynamics and material viscoelastic properties. Gelatin hydrogels reveal concentration-dependent impacts on bubble expansion and material elasticity. Embedding PFC droplets in gelatin increases internal pressure, resulting in higher equilibrium radius and continuous bubble growth during quasi-static evolution. Similar trends are observed in fibrin and polyacrylamide matrices, with differences in bubble behavior attributed to matrix properties and droplet presence. Interestingly, droplet size exhibits minimal impact on bubble expansion during inertial dynamics but influences quasi-static evolution, with larger droplets leading to continuous growth beyond 60 s. Furthermore, the core composition of PFC droplets significantly affects bubble behavior, with higher boiling point droplets exhibiting higher maximum expansion and faster quasi-static dissolution rates. Overall, the study sheds light on the intricate interplay between droplet characteristics, matrix properties, and multi-timescale bubble dynamics, offering valuable insights into their behavior within biomimetic hydrogels.
Collapse
Affiliation(s)
- Bachir A Abeid
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Mitra Aliabouzar
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan B Estrada
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Bader KB, Padilla F, Haworth KJ, Ellens N, Dalecki D, Miller DL, Wear KA. Overview of Therapeutic Ultrasound Applications and Safety Considerations: 2024 Update. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024. [PMID: 39526313 DOI: 10.1002/jum.16611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
A 2012 review of therapeutic ultrasound was published to educate researchers and physicians on potential applications and concerns for unintended bioeffects (doi: 10.7863/jum.2012.31.4.623). This review serves as an update to the parent article, highlighting advances in therapeutic ultrasound over the past 12 years. In addition to general mechanisms for bioeffects produced by therapeutic ultrasound, current applications, and the pre-clinical and clinical stages are outlined. An overview is provided for image guidance methods to monitor and assess treatment progress. Finally, other topics relevant for the translation of therapeutic ultrasound are discussed, including computational modeling, tissue-mimicking phantoms, and quality assurance protocols.
Collapse
Affiliation(s)
- Kenneth B Bader
- Department of Radiology, University of Chicago, Chicago, Illinois, USA
| | - Frederic Padilla
- Gene Therapy Program, Focused Ultrasound Foundation, Charlottesville, Virginia, USA
- Department of Radiology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Kevin J Haworth
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States
- Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Diane Dalecki
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Douglas L Miller
- Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Keith A Wear
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
3
|
Su X, Geng X, Zhang Y, Shi Y, Zhao L. Microenvironmental pH modulating oxygen self-boosting microalgal prodrug carboxymethyl chitosan/hyaluronic acid/puerarin hydrogel for accelerating wound healing in diabetic rats. Int J Biol Macromol 2024; 282:136669. [PMID: 39437940 DOI: 10.1016/j.ijbiomac.2024.136669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Chronic diabetic wounds are characterized by a range of detrimental features, including hypoxia, elevated levels of reactive oxygen species, impaired angiogenesis, chronic inflammation, and an increased susceptibility to bacterial infections. We have developed an innovative multifunctional hydrogel system based on carboxymethyl chitosan, which incorporates embedded microalgae PCC7942 along with hyaluronic acid and puerarin, termed PCC7942@carboxymethyl chitosan/hyaluronic acid/puerarin hydrogel. It demonstrated outstanding capabilities in exudate absorption, mechanical flexibility, hemostatic action, and antibacterial efficacy. Furthermore, it effectively modulated the pH of wound microenvironment through the hydrolysis of amide bonds, thereby establishing a favorable low-pH microenvironment. Microalgae in hydrogel covered in the wound exhibited stable and continuous oxygen production within 24 h, with more efficiency in dissolved oxygen penetration through skin. Furthermore, prodrugs such as hyaluronic acid and puerarin from hydrogel displayed the controlled release behavior and facilitated the fast and enhanced accumulation of drugs at wound site, thereby accelerating the process of wound healing via enhanced angiogenesis and anti-inflammation effects. In summary, the healing-promoting effect of PCC7942@carboxymethyl chitosan/hyaluronic acid/puerarin hydrogel in type 1 diabetic rats can be attributed to the synergistic effects of microalgae, hyaluronic acid, and puerarin, which collectively accelerated wound healing rate and improved the quality of wound recovery.
Collapse
Affiliation(s)
- Xiangchen Su
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China
| | - Xinrong Geng
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China
| | - Yifei Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou, Liaoning, China; Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
4
|
Huang A, Jia Z, Wu H, Feng K, Zhang C, Wan M, Zong Y. Exploring the Postactivation Behavioral Patterns of Intratumorally Injected Theranostic Nanodroplets: An Ultrasound-Only Extravascular Monitoring Technique. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:1186-1198. [PMID: 39196736 DOI: 10.1109/tuffc.2024.3450885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Phase-change nanodroplets (PCNDs) are customizable and controllable theranostic agents of particular interest in extravascular therapies such as drug delivery and histotripsy. High-bulk-boiling-point (HBP) PCNDs are preferred for their enhanced thermal stability under physiological temperature to achieve on-demand therapeutic effects on target sites-mainly in tumor tissue. However, the behavioral patterns of high-concentration, heterogeneously distributed HBP PCNDs in vivo have rarely been explored-the foci of PCND-related therapies mostly fall on the final therapeutic effect rather than the detailed behaviors of PCNDs, which may hamper the development and improvement of in vivo treatments with PCNDs. To fill the gap, we demonstrate an ultrasound-only extravascular monitoring technique to analyze the underlying behavioral patterns of intratumorally injected HBP PCNDs. In our hypothesis, recondensation and coalescence are the two predominant patterns influencing the trend of the postactivation signal of PCNDs. A "blink map" method was, thus, proposed to separate the two parts of the signal by recognizing the unique signal pattern of stochastic recondensation, and four derivative metrics were calculated for further analysis. The results revealed the postactivation patterns of PCNDs at different activation-pulse durations and activation stages throughout the activation-imaging period, and several general trends were observed and explained by existing theories, suggesting the feasibility of our extravascular monitoring technique. Overall, this work enriches the knowledge of the characteristics of HBP PCNDs as extravascular theranostic agents, and the monitoring results have the potential to provide timely feedback on PCND-related treatments underway, which may help adjust the treatment strategy and improve the therapeutic efficacy.
Collapse
|
5
|
Maciulevičius M, Palepšienė R, Vykertas S, Raišutis R, Rafanavičius A, Krilavičius T, Šatkauskas S. The comparison of the dynamics of Ca 2+ and bleomycin intracellular delivery after cell sonoporation and electroporation in vitro. Bioelectrochemistry 2024; 158:108708. [PMID: 38636366 DOI: 10.1016/j.bioelechem.2024.108708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
Ca2+, in combination with SP or EP, induces cell cytotoxicity much faster compared to BLM. The application of BLM in combination with, SP or EP, reaches the level of cell death, induced by similar combination with Ca2+, only after 72 h. The methods of SP and EP were calibrated according to the level of differential cytotoxicity, determined after 6 days (using cell clonogenic assay). The combination of Ca2+ SP induces cell death faster than Ca2+ EP - after Ca2+ SP it increases to a maximum level after 15 min and remains constant for up to 6 days, while the cytotoxic efficiency after Ca2+ EP increases to the level of Ca2+ SP only after 72 h. The combination of BLM SP shows a very similar dynamics to BLM EP - both reach maximal level of cytotoxicity after 48-72 h. Ca2+ and BLM in combination with SP have shown similar levels of cytotoxicity at higher acoustic pressures (≥250 kPa); therefore, Ca2+ SP can be used to induce immediate and maximal level of cytotoxic effect. The faster cytotoxic efficiency of Ca2+ in combination with SP than EP was determined to be due to the involvement of microbubble inertial cavitation.
Collapse
Affiliation(s)
- Martynas Maciulevičius
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania; Ultrasound Research Institute, Kaunas University of Technology, K. Baršausko st. 59, LT-51423 Kaunas, Lithuania.
| | - Rūta Palepšienė
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Salvijus Vykertas
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Renaldas Raišutis
- Ultrasound Research Institute, Kaunas University of Technology, K. Baršausko st. 59, LT-51423 Kaunas, Lithuania; Department of Electrical Power Systems, Faculty of Electrical and Electronics Engineering, Kaunas University of Technology, Studentų st. 48, LT-51367 Kaunas, Lithuania.
| | - Aras Rafanavičius
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Tomas Krilavičius
- Faculty of Informatics, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Saulius Šatkauskas
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| |
Collapse
|
6
|
Jayasankar G, Koilpillai J, Narayanasamy D. A Systematic Study on Long-acting Nanobubbles: Current Advancement and Prospects on Theranostic Properties. Adv Pharm Bull 2024; 14:278-301. [PMID: 39206408 PMCID: PMC11347731 DOI: 10.34172/apb.2024.042] [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: 08/23/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 09/04/2024] Open
Abstract
Delivery of diagnostic drugs via nanobubbles (NBs) has shown to be an emerging field of study. Due to their small size, NBs may more easily travel through constricted blood vessels and precisely target certain bodily parts. NB is considered the major treatment for cancer treatment and other diseases which are difficult to diagnose. The field of NBs is dynamic and continues to grow as researchers discover new properties and seek practical applications in various fields. The predominant usage of NBs in novel drug delivery is to enhance the bioavailability, and controlled drug release along with imaging properties NBs are important because they may change interfacial characteristics including surface force, lubrication, and absorption. The quick diffusion of gas into the water was caused by a hypothetical film that was stimulated and punctured by a strong acting force at the gas/water contact of the bubble. In this article, various prominent aspects of NBs have been discussed, along with the long-acting nature, and the theranostical aspect which elucidates the potential marketed drugs along with clinical trial products. The article also covers quality by design aspects, different production techniques that enable method-specific therapeutic applications, increasing the floating time of the bubble, and refining its properties to enhance the prepared NB's quality. NB containing both analysis and curing properties makes it special from other nano-carriers. This work includes all the possible methods of preparing NB, its application, all marketed drugs, and products in clinical trials.
Collapse
Affiliation(s)
| | | | - Damodharan Narayanasamy
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institution of Science and Technology, Kattankulathur, Chengalpattu, India
| |
Collapse
|
7
|
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
|
8
|
Mochizuki K, Mitova V, Makino K, Terada H, Takeuchi I, Troev K. pH-Sensitive Amphiphilic Diblock Polyphosphoesters with Lactate Units: Synthesis and Application as Drug Carriers. Int J Mol Sci 2024; 25:4518. [PMID: 38674103 PMCID: PMC11049995 DOI: 10.3390/ijms25084518] [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: 03/13/2024] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
pH-sensitive amphiphilic diblock polyphosphoesters containing lactic acid units were synthesized by multistep one-pot polycondensation reactions. They comprise acid-labile P(O)-O-C and C(O)-O-C bonds, the cleavage of which depends on the pH of the medium. The structure of these copolymers was characterized by 1H, 13C {H}, 31P NMR, and size exclusion chromatography (SEC). The newly synthesized polymers self-assembled into the micellar structure in an aqueous solution. The effects of the molecular weight of the copolymer and the length of the hydrophobic chain on micelle formation and stabilityand micelle size were studied via dynamic light scattering (DLS). Drug loading and encapsulation efficiency tests using doxorubicin revealed that hydrophobic drugs can be delivered by copolymers. It was established that the molecular weight of the copolymer, length of the hydrophobic chain and content of lactate units affects the size of the micelles, drug loading, and efficiency of encapsulation. A copolymer with 10.7% lactate content has drug loading (3.2 ± 0.3) and efficiency of encapsulation (57.4 ± 3.2), compared to the same copolymer with 41.8% lactate content (1.63%) and (45.8%), respectively. It was demonstrated that the poly[alkylpoly(ethylene glycol) phosphate-b-alkylpoly(ethylene glycol)lactate phosphate] DOX system has a pH-sensitive response capability in the result in which DOX was selectively accumulated into the tumor, where pH is acidic. The results obtained indicate that amphiphilic diblock polyphosphoesters have potential as drug carriers.
Collapse
Affiliation(s)
- Kasumi Mochizuki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan; (K.M.); (K.M.); (H.T.); (I.T.)
| | - Violeta Mitova
- Institute of Polymers, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Kimiko Makino
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan; (K.M.); (K.M.); (H.T.); (I.T.)
| | - Hiroshi Terada
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan; (K.M.); (K.M.); (H.T.); (I.T.)
| | - Issei Takeuchi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan; (K.M.); (K.M.); (H.T.); (I.T.)
- Faculty of Pharmaceutical Science, Josai International University, 1 Gumyo, Togane 283-8555, Chiba, Japan
| | - Kolio Troev
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan; (K.M.); (K.M.); (H.T.); (I.T.)
- Institute of Polymers, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| |
Collapse
|
9
|
Chen J, Zhao C, Liu H, Wang Z, Ma L, Zhang J, Xu N, Hu K, Duan L. Integrated micro/nano drug delivery system based on magnetically responsive phase-change droplets for ultrasound theranostics. Front Bioeng Biotechnol 2024; 12:1323056. [PMID: 38665816 PMCID: PMC11043469 DOI: 10.3389/fbioe.2024.1323056] [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: 10/17/2023] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Phase-change droplets (PCDs) are intelligent responsive micro and nanomaterials developed based on micro/nano bubbles. Subject to external energy inputs such as temperature and ultrasound, the core substance, perfluorocarbon (PFC), undergoes a phase transition from liquid to gas. This transformation precipitates alterations in the PCDs' structure, size, ultrasound imaging capabilities, drug delivery efficiency, and other pertinent characteristics. This gives them the ability to exhibit "intelligent responses". This study utilized lipids as the membrane shell material and perfluorohexane (PFH) as the core to prepare lipid phase-change droplets. Superparamagnetic nanoparticles (PEG-functionalized Fe3O4 nanoparticles) and the anti-tumor drug curcumin (Cur) were loaded into the membrane shell, forming magnetic drug-loaded phase-change droplets (Fe-Cur-NDs). These nanoscale phase-change droplets exhibited excellent magnetic resonance/ultrasound imaging capabilities and thermal/ultrasound-mediated drug release. The Fe-Cur-NDs showed excellent anti-tumor efficacy for the MCF-7 cells under low-intensity focused ultrasound (LIFU) guidance in vitro. Therefore, Fe-Cur-NDs represent a promising smart responsive theranostic integrated micro/nano drug delivery system.
Collapse
Affiliation(s)
- Jieying Chen
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Chan Zhao
- Department of Clinical Medical Engineering, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Liu
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Zhangchao Wang
- Stomatological College, Nanjing Medical University, Nanjing, China
| | - Luyao Ma
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jiamin Zhang
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Ning Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ke Hu
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Lei Duan
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Martínez-Orts M, Pujals S. Responsive Supramolecular Polymers for Diagnosis and Treatment. Int J Mol Sci 2024; 25:4077. [PMID: 38612886 PMCID: PMC11012635 DOI: 10.3390/ijms25074077] [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: 03/01/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Stimuli-responsive supramolecular polymers are ordered nanosized materials that are held together by non-covalent interactions (hydrogen-bonding, metal-ligand coordination, π-stacking and, host-guest interactions) and can reversibly undergo self-assembly. Their non-covalent nature endows supramolecular polymers with the ability to respond to external stimuli (temperature, light, ultrasound, electric/magnetic field) or environmental changes (temperature, pH, redox potential, enzyme activity), making them attractive candidates for a variety of biomedical applications. To date, supramolecular research has largely evolved in the development of smart water-soluble self-assemblies with the aim of mimicking the biological function of natural supramolecular systems. Indeed, there is a wide variety of synthetic biomaterials formulated with responsiveness to control and trigger, or not to trigger, aqueous self-assembly. The design of responsive supramolecular polymers ranges from the use of hydrophobic cores (i.e., benzene-1,3,5-tricarboxamide) to the introduction of macrocyclic hosts (i.e., cyclodextrins). In this review, we summarize the most relevant advances achieved in the design of stimuli-responsive supramolecular systems used to control transport and release of both diagnosis agents and therapeutic drugs in order to prevent, diagnose, and treat human diseases.
Collapse
Affiliation(s)
| | - Silvia Pujals
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain;
| |
Collapse
|
11
|
Shakya G, Cattaneo M, Guerriero G, Prasanna A, Fiorini S, Supponen O. Ultrasound-responsive microbubbles and nanodroplets: A pathway to targeted drug delivery. Adv Drug Deliv Rev 2024; 206:115178. [PMID: 38199257 DOI: 10.1016/j.addr.2023.115178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Ultrasound-responsive agents have shown great potential as targeted drug delivery agents, effectively augmenting cell permeability and facilitating drug absorption. This review focuses on two specific agents, microbubbles and nanodroplets, and provides a sequential overview of their drug delivery process. Particular emphasis is given to the mechanical response of the agents under ultrasound, and the subsequent physical and biological effects on the cells. Finally, the state-of-the-art in their pre-clinical and clinical implementation are discussed. Throughout the review, major challenges that need to be overcome in order to accelerate their clinical translation are highlighted.
Collapse
Affiliation(s)
- Gazendra Shakya
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Marco Cattaneo
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Giulia Guerriero
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Anunay Prasanna
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Samuele Fiorini
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Outi Supponen
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|
12
|
Howells AR, Welch PJ, Kim J, Forest CR, Shi C, Lian XL. A drug-selectable acoustic reporter gene system for human cell ultrasound imaging. Bioeng Transl Med 2024; 9:e10584. [PMID: 38435822 PMCID: PMC10905554 DOI: 10.1002/btm2.10584] [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: 01/20/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 03/05/2024] Open
Abstract
A promising new field of genetically encoded ultrasound contrast agents in the form of gas vesicles has recently emerged, which could extend the specificity of medical ultrasound imaging. However, given the delicate genetic nature of how these genes are integrated and expressed, current methods of producing gas vesicle-expressing mammalian cell lines requires significant cell processing time to establish a clonal/polyclonal line that robustly expresses the gas vesicles sufficiently enough for ultrasound contrast. Here, we describe an inducible and drug-selectable acoustic reporter gene system that can enable gas vesicle expression in mammalian cell lines, which we demonstrate using HEK293T cells. Our drug-selectable construct design increases the stability and proportion of cells that successfully integrate all plasmids into their genome, thus reducing the amount of cell processing time required. Additionally, we demonstrate that our drug-selectable strategy forgoes the need for single-cell cloning and fluorescence-activated cell sorting, and that a drug-selected mixed population is sufficient to generate robust ultrasound contrast. Successful gas vesicle expression was optically and ultrasonically verified, with cells expressing gas vesicles exhibiting an 80% greater signal-to-noise ratio compared to negative controls and a 500% greater signal-to-noise ratio compared to wild-type HEK293T cells. This technology presents a new reporter gene paradigm by which ultrasound can be harnessed to visualize specific cell types for applications including cellular reporting and cell therapies.
Collapse
Affiliation(s)
| | - Phoebe J. Welch
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - John Kim
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Craig R. Forest
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Chengzhi Shi
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGeorgiaUSA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Xiaojun Lance Lian
- Department of Biomedical EngineeringPennsylvania State UniversityPennsylvaniaUSA
- Huck Institutes of the Life Sciences, Pennsylvania State UniversityPennsylvaniaUSA
- Department of BiologyPennsylvania State UniversityPennsylvaniaUSA
| |
Collapse
|
13
|
Bouakaz A, Michel Escoffre J. From concept to early clinical trials: 30 years of microbubble-based ultrasound-mediated drug delivery research. Adv Drug Deliv Rev 2024; 206:115199. [PMID: 38325561 DOI: 10.1016/j.addr.2024.115199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Ultrasound mediated drug delivery, a promising therapeutic modality, has evolved remarkably over the past three decades. Initially designed to enhance contrast in ultrasound imaging, microbubbles have emerged as a main vector for drug delivery, offering targeted therapy with minimized side effects. This review addresses the historical progression of this technology, emphasizing the pivotal role microbubbles play in augmenting drug extravasation and targeted delivery. We explore the complex mechanisms behind this technology, from stable and inertial cavitation to diverse acoustic phenomena, and their applications in medical fields. While the potential of ultrasound mediated drug delivery is undeniable, there are still challenges to overcome. Balancing therapeutic efficacy and safety and establishing standardized procedures are essential areas requiring attention. A multidisciplinary approach, gathering collaborations between researchers, engineers, and clinicians, is important for exploiting the full potential of this technology. In summary, this review highlights the potential of using ultrasound mediated drug delivery in improving patient care across various medical conditions.
Collapse
Affiliation(s)
- Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | | |
Collapse
|
14
|
Xu W, Zhao Y, Zhang C, Huo M, Wang L, Wu X, Zhang Y, Li Q, Gai Y. Bimetallic nanoplatform for synergistic sonodynamic-calcium overload therapy utilizing self-supplied hydrogen peroxide and relieved hypoxia. Biomater Sci 2024; 12:1171-1184. [PMID: 38205509 DOI: 10.1039/d3bm01430d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Sonodynamic therapy (SDT) has emerged as a potential alternative to traditional cancer treatments as it offers deep cellular penetration and reduced invasivity. Sonosensitizers generate reactive oxygen species (ROS) under ultrasound activation, focusing the ultrasound energy on malignant sites located deep in tissues and causing cell apoptosis and necrosis. However, due to tumor hypoxia and the limited levels of intracellular endogenous hydrogen peroxide (H2O2 is a fundamental species for supplying oxygen via catalase activity), SDT efficacy is still insufficient. In this study, a bimetallic and multifunctional system (Fe3O4-TAPP@PVP-CaO2) was prepared by using ferrosoferric oxide (Fe3O4) as a carrier loaded with 5,10,15,20-tetrakis(4-aminophenyl), porphyrin (TAPP), that was then coated with polyvinyl pyrrolidone (PVP) and calcium peroxide (CaO2). The CaO2 layer elevated the levels of H2O2 and Ca2+ in the tumor microenvironment when exposed to intracellular acidity, providing essential elements for oxygen generation. Intracellular hypoxia was alleviated via the catalase-like activity of Fe3O4 inducing calcium overload. Under ultrasonic irradiation, SDT generated toxic reactive oxygen species (ROS, singlet oxygen) and activated calcium influx through acoustic cavitation. Meanwhile, calcium overload therapy efficiently induced cell apoptosis at the moment of uncontrollable cellular accumulation of Ca2+. In addition, we modified the PVP on the surface to make it more stable. This study presents a bimetallic nanoplatform that can efficiently induce cancer cell death by synergistic sonodynamic-calcium overload therapy via modulation of O2/ROS/Ca2+ species, indicating its potential for multi-modality cancer therapy.
Collapse
Affiliation(s)
- Wenqian Xu
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Yisheng Zhao
- School of Pharmaceutical Sciences, Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, P.R. China
| | - Chao Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, P.R. China
| | - Mengping Huo
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Lei Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Xuewu Wu
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Nephro-Urological Clinical Center, Lanzhou 730000, P.R. China
| | - Yang Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Qiao Li
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Yonghao Gai
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| |
Collapse
|
15
|
Yu J, Liu Y, Zhang Y, Ran R, Kong Z, Zhao D, Liu M, Zhao W, Cui Y, Hua Y, Gao L, Zhang Z, Yang Y. Smart nanogels for cancer treatment from the perspective of functional groups. Front Bioeng Biotechnol 2024; 11:1329311. [PMID: 38268937 PMCID: PMC10806105 DOI: 10.3389/fbioe.2023.1329311] [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: 10/28/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Introduction: Cancer remains a significant health challenge, with chemotherapy being a critical treatment modality. However, traditional chemotherapy faces limitations due to non-specificity and toxicity. Nanogels, as advanced drug carriers, offer potential for targeted and controlled drug release, improving therapeutic efficacy and reducing side effects. Methods: This review summarizes the latest developments in nanogel-based chemotherapy drug delivery systems, focusing on the role of functional groups in drug loading and the design of smart hydrogels with controlled release mechanisms. We discuss the preparation methods of various nanogels based on different functional groups and their application in cancer treatment. Results: Nanogels composed of natural and synthetic polymers, such as chitosan, alginate, and polyacrylic acid, have been developed for chemotherapy drug delivery. Functional groups like carboxyl, disulfide, and hydroxyl groups play crucial roles in drug encapsulation and release. Smart hydrogels have been engineered to respond to tumor microenvironmental cues, such as pH, redox potential, temperature, and external stimuli like light and ultrasound, enabling targeted drug release. Discussion: The use of functional groups in nanogel preparation allows for the creation of multifunctional nanogels with high drug loading capacity, controllable release, and good targeting. These nanogels have shown promising results in preclinical studies, with enhanced antitumor effects and reduced systemic toxicity compared to traditional chemotherapy. Conclusion: The development of smart nanogels with functional group-mediated drug delivery and controlled release strategies represents a promising direction in cancer therapy. These systems offer the potential for improved patient outcomes by enhancing drug targeting and minimizing adverse effects. Further research is needed to optimize nanogel design, evaluate their safety and efficacy in clinical trials, and explore their potential for personalized medicine.
Collapse
Affiliation(s)
- Jiachen Yu
- General Hospital of Northern Theater Command, China Medical University, Shenyang, China
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yuting Liu
- General Hospital of Northern Theater Command, China Medical University, Shenyang, China
- Shenyang Traditional Chinese Medicine Hospital, China Medical University, Shenyang, China
| | - Yingchun Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Rong Ran
- Department of Anesthesia, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Zixiao Kong
- China Medical University, Shenyang, Liaoning, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Minda Liu
- Department of Oral-maxillofacial Head and Neck, Oral Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Lianbo Gao
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
| | - Yingxin Yang
- General Hospital of Northern Theater Command, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Pellow C, Jafari Sojahrood A, Zhao X, Kolios MC, Exner AA, Goertz DE. Synchronous Intravital Imaging and Cavitation Monitoring of Antivascular Focused Ultrasound in Tumor Microvasculature Using Monodisperse Low Boiling Point Nanodroplets. ACS NANO 2024; 18:410-427. [PMID: 38147452 PMCID: PMC10786165 DOI: 10.1021/acsnano.3c07711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.
Collapse
Affiliation(s)
- Carly Pellow
- Sunnybrook Research Institute, Toronto M4N 3M5, Canada
| | - Amin Jafari Sojahrood
- Sunnybrook Research Institute, Toronto M4N 3M5, Canada
- Department of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Xiaoxiao Zhao
- Sunnybrook Research Institute, Toronto M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - David E Goertz
- Sunnybrook Research Institute, Toronto M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| |
Collapse
|
17
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
18
|
Polydorou AE, May JP, Makris K, Ferri S, Wu Q, Stride E, Carugo D, Evans ND. An investigation into the cytotoxic effects of microbubbles and their constituents on osteosarcoma and bone marrow stromal cells. Biochim Biophys Acta Gen Subj 2023; 1867:130481. [PMID: 37802372 DOI: 10.1016/j.bbagen.2023.130481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Ultrasound-responsive microbubbles offer a means of achieving minimally invasive, localised drug delivery in applications including regenerative medicine. To facilitate their use, however, it is important to determine any cytotoxic effects they or their constituents may have. The aim of this study was to test the hypothesis that phospholipid-shelled microbubbles are non-toxic to human bone-derived cells at biologically-relevant concentrations. METHODS Microbubbles were fabricated using combinations of 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-dibehenoyl-sn-glycero-3-phosphocholine (DBPC), polyoxyethylene(40) stearate (PEG40S) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene-glycol)-2000] (DSPE-PEG2000). Microbubble size and concentration were measured as a function of time and temperature by optical microscopy. Effects on MG63 osteosarcoma and human bone marrow stromal cells (BMSCs) were measured for up to 72 h by assay for viability, metabolic activity and proliferation. RESULTS DBPC:DSPE-PEG2000 microbubbles were significantly more stable than DSPC:PEG40S microbubbles under all conditions tested. Serum-containing medium had no detrimental effect on microbubble stability, but storage at 37 °C compared to at 4 °C reduced stability for both preparations, with almost complete dissolution of microbubbles at times ≥24 h. DSPC:PEG40S microbubbles had greater inhibitory effects on cell metabolism and growth than DBPC:DSPE-PEG2000 microbubbles, with PEG40S found to be the principle inhibitory component. These effects were only evident at high microbubble concentrations (≥20% (v/v)) or with prolonged culture (≥24 h). Increasing cell-microbubble contact by inversion culture in a custom-built device had no inhibitory effect on metabolism. CONCLUSIONS These data indicate that, over a broad range of concentrations and incubation times, DBPC:DSPE-PEG2000 and DSPC:PEG40S microbubbles have little effect on osteoblastic cell viability and growth, and that PEG40S is the principle inhibitory component in the formulations investigated.
Collapse
Affiliation(s)
- A E Polydorou
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - J P May
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - K Makris
- Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - S Ferri
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom
| | - Q Wu
- Institute of Biomedical Engineering, University of Oxford, United Kingdom
| | - E Stride
- Institute of Biomedical Engineering, University of Oxford, United Kingdom; Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, United Kingdom
| | - D Carugo
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, United Kingdom
| | - N D Evans
- Centre for Human Development, Stem Cells and Regenerative Medicine, Bone and Joint Research group, University of Southampton, United Kingdom; Bioengineering Sciences Group, Institute for Life Sciences, University of Southampton, United Kingdom.
| |
Collapse
|
19
|
Deng L, Lea-Banks H, Jones RM, O’Reilly MA, Hynynen K. Three-dimensional super resolution ultrasound imaging with a multi-frequency hemispherical phased array. Med Phys 2023; 50:7478-7497. [PMID: 37702919 PMCID: PMC10872837 DOI: 10.1002/mp.16733] [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: 04/26/2023] [Accepted: 08/27/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND High resolution imaging of the microvasculature plays an important role in both diagnostic and therapeutic applications in the brain. However, ultrasound pulse-echo sonography imaging the brain vasculatures has been limited to narrow acoustic windows and low frequencies due to the distortion of the skull bone, which sacrifices axial resolution since it is pulse length dependent. PURPOSE To overcome the detect limit, a large aperture 256-module sparse hemispherical transmit/receive array was used to visualize the acoustic emissions of ultrasound-vaporized lipid-coated decafluorobutane nanodroplets flowing through tube phantoms and within rabbit cerebral vasculature in vivo via passive acoustic mapping and super resolution techniques. METHODS Nanodroplets were vaporized with 55 kHz burst-mode ultrasound (burst length = 145 μs, burst repetition frequency = 9-45 Hz, peak negative acoustic pressure = 0.10-0.22 MPa), which propagates through overlying tissues well without suffering from severe distortions. The resulting emissions were received at a higher frequency (612 or 1224 kHz subarray) to improve the resulting spatial resolution during passive beamforming. Normal resolution three-dimensional images were formed using a delay, sum, and integrate beamforming algorithm, and super-resolved images were extracted via Gaussian fitting of the estimated point-spread-function to the normal resolution data. RESULTS With super resolution techniques, the mean lateral (axial) full-width-at-half-maximum image intensity was 16 ± 3 (32 ± 6) μm, and 7 ± 1 (15 ± 2) μm corresponding to ∼1/67 of the normal resolution at 612 and 1224 kHz, respectively. The mean positional uncertainties were ∼1/350 (lateral) and ∼1/180 (axial) of the receive wavelength in water. In addition, a temporal correlation between nanodroplet vaporization and the transmit waveform shape was observed, which may provide the opportunity to enhance the signal-to-noise ratio in future studies. CONCLUSIONS Here, we demonstrate the feasibility of vaporizing nanodroplets via low frequency ultrasound and simultaneously performing spatial mapping via passive beamforming at higher frequencies to improve the resulting spatial resolution of super resolution imaging techniques. This method may enable complete four-dimensional vascular mapping in organs where a hemispherical array could be positioned to surround the target, such as the brain, breast, or testicles.
Collapse
Affiliation(s)
- Lulu Deng
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, M4N 3M5, Canada
| | - Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, M4N 3M5, Canada
| | - Ryan M. Jones
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, M4N 3M5, Canada
| | - Meaghan A. O’Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3E2, Canada
| |
Collapse
|
20
|
Mendes MIP, Coelho CDF, Schaberle FA, Moreno MJ, Calvete MJF, Arnaut LG. Nanodroplet vaporization with pulsed-laser excitation repeatedly amplifies photoacoustic signals at low vaporization thresholds. RSC Adv 2023; 13:35040-35049. [PMID: 38046627 PMCID: PMC10690495 DOI: 10.1039/d3ra05639b] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023] Open
Abstract
Nanodroplets' explosive vaporization triggered by absorption of laser pulses produces very large volume changes. These volume changes are two orders of magnitude higher than those of thermoelastic expansion generated by equivalent laser pulses, and should generate correspondingly higher photoacoustic waves (PAW). The generation of intense PAWs is desirable in photoacoustic tomography (PAT) to increase sensitivity. The biocompatibility and simplicity of nanodroplets obtained by sonication of perfluoropentane (PFP) in an aqueous solution of bovine serum albumin (BSA) containing a dye make them particularly appealing for use as contrast agents in clinical applications of PAT. Their usefulness depends on stability and reproducible vaporization of nanodroplets (liquid PFP inside) to microbubbles (gaseous PFP inside), and reversible condensation to nanodroplets. This work incorporates porphyrins with fluorinated chains and BSA labelled with fluorescent probes in PFP nanodroplets to investigate the structure and properties of such nanodroplets. Droplets prepared with average diameters in the 400-1000 nm range vaporize when exposed to nanosecond laser pulses with fluences above 3 mJ cm-2 and resist coalescence. The fluorinated chains are likely responsible for the low vaporization threshold, ∼2.5 mJ cm-2, which was obtained from the laser fluence dependence of the photoacoustic wave amplitudes. Only ca. 10% of the droplets incorporate fluorinated porphyrins. Nevertheless, PAWs generated with nanodroplets are ten times higher than those generated by aqueous BSA solutions containing an equivalent amount of porphyrin. Remarkably, successive laser pulses result in similar amplification, indicating that the microbubbles revert back to nanodroplets at a rate faster than the laser repetition rate (10 Hz). PFP nanodroplets are promising contrast agents for PAT and their performance increases with properly designed dyes.
Collapse
Affiliation(s)
- Maria Inês P Mendes
- CQC-IMS, Chemistry Department, University of Coimbra 3004-535 Coimbra Portugal
- LaserLeap Technologies Rua Coronel Júlio Veiga Simão, Edifício B, CTCV, S/N 3025-307 Coimbra Portugal
| | - Carlos D F Coelho
- CQC-IMS, Chemistry Department, University of Coimbra 3004-535 Coimbra Portugal
| | - Fábio A Schaberle
- CQC-IMS, Chemistry Department, University of Coimbra 3004-535 Coimbra Portugal
| | - Maria João Moreno
- CQC-IMS, Chemistry Department, University of Coimbra 3004-535 Coimbra Portugal
| | - Mário J F Calvete
- CQC-IMS, Chemistry Department, University of Coimbra 3004-535 Coimbra Portugal
| | - Luis G Arnaut
- CQC-IMS, Chemistry Department, University of Coimbra 3004-535 Coimbra Portugal
| |
Collapse
|
21
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
22
|
Shi Z, Hu Y, Li X. Polymer mechanochemistry in drug delivery: From controlled release to precise activation. J Control Release 2023; 365:S0168-3659(23)00703-4. [PMID: 39491171 DOI: 10.1016/j.jconrel.2023.10.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/08/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Controlled drug delivery systems that can respond to mechanical force offer a unique solution for on-demand activation and release under physiological conditions. Compression, tension, and shear forces encompass the most commonly utilized mechanical stimuli for controlled drug activation and release. While compression and tension forces have been extensively explored for designing mechanoresponsive drug release systems through object deformation, ultrasound (US) holds advantages in achieving spatiotemporally controlled drug release from micro-/nanocarriers such as microbubbles, liposomes, and micelles. Unlike light-based methods, the US bypasses drawbacks such as phototoxicity and limited tissue penetration. Conventional US-triggered drug release primarily relies on heat-induced phase transitions or chemical transformations in the nano-/micro-scale range. In contrast, the cutting-edge approach of "Sonopharmacology" leverages polymer mechanochemistry, where US-induced shear force activates latent sites containing active pharmaceutical ingredients incorporated into polymer chains more readily than other bonds within the polymeric structure. This article provides a brief overview of controlled drug release systems based on compression and tension, followed by recent significant studies on drug activation using the synergistic effects of US and polymer mechanochemistry. The remaining challenges and potential future directions in this subfield are also discussed. PROGRESS AND POTENTIAL: The precise spatiotemporal control of drug activity using exogenous signals holds great promise for achieving precise disease treatment with minimal side effects. Ultrasound, known for its safety, has found widespread application in clinical settings and offers adjustable tissue penetration depth and drug release control. However, challenges persist in achieving precise control over drug activity using ultrasound. In recent years, ultrasound-induced drug release utilizing the principle of polymer mechanochemistry (Sonopharmacology) has made significant progress and demonstrated its potential in achieving precise drug activation and release. These systems enable drug release at the sub-molecular level, allowing for selective control over drug activation. Sonopharmacology offers a unique advantage by integrating both chemical and biomedical perspectives, positioning it as a promising field with broad implications in polymer chemistry, nanoscience and technology, and pharmaceutics. This review article aims to examine recent advancements in ultrasound-triggered drug activation systems based on polymeric materials and with an focus on polymer mechanochemistry, identify remaining challenges, and propose potential perspectives in this rapidly evolving field. By providing a comprehensive understanding of the progress and potential of sonopharmacology, this article aims to guide future research and inspire the development of innovative drug delivery systems that offer enhanced selectivity and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyuan Shi
- School of Pharmaceutical Science and Technology, Tianjin University, 300072 Tianjin, China.
| | - Yong Hu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China.
| | - Xin Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 201804 Shanghai, China.
| |
Collapse
|
23
|
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
|
24
|
Arıbal E. Future of Breast Radiology. Eur J Breast Health 2023; 19:262-266. [PMID: 37795010 PMCID: PMC10546805 DOI: 10.4274/ejbh.galenos.2023.2023-8-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
The landscape of breast imaging has transformed significantly since mammography's introduction in the 1960s, accelerated by ultrasound and imageguided biopsies in the 1990s. The emergence of magnetic resonance imaging (MRI) in the 2000s added a valuable dimension to advanced imaging. Multimodality and multiparametric imaging have firmly established breast radiology's pivotal role in managing breast disorders. A shift from conventional to digital radiology emerged in the late 20th and early 21st centuries, enabling advanced techniques like digital breast tomosynthesis, contrast-enhanced mammography, and artificial intelligence (AI) integration. AI's impending integration into breast radiology may enhance diagnostics and workflows. It involves computer-aided diagnosis (CAD) algorithms, workflow support algorithms, and data processing algorithms. CAD systems, developed since the 1980s, optimize cancer detection rates by addressing false positives and negatives. Radiologists' roles will evolve into specialized clinicians collaborating with AI for efficient patient care and utilizing advanced techniques with multiparametric imaging and radiomics. Wearable technologies, non-contrast MRI, and innovative modalities like photoacoustic imaging show potential to enhance diagnostics. Imaging-guided therapy, notably cryotherapy, and theranostics, gains traction. Theranostics, integrating therapy and diagnostics, holds potential for precise treatment. Advanced imaging, AI, and novel therapies will revolutionize breast radiology, offering refined diagnostics and personalized treatments. Personalized screening, AI's role, and imaging-guided therapies will shape the future of breast radiology.
Collapse
Affiliation(s)
- Erkin Arıbal
- Acıbadem University Faculty of Medicine, Department of Radiology, İstanbul, Turkey
| |
Collapse
|
25
|
Guo S, Gu D, Yang Y, Tian J, Chen X. Near-infrared photodynamic and photothermal co-therapy based on organic small molecular dyes. J Nanobiotechnology 2023; 21:348. [PMID: 37759287 PMCID: PMC10523653 DOI: 10.1186/s12951-023-02111-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Near-infrared (NIR) organic small molecule dyes (OSMDs) are effective photothermal agents for photothermal therapy (PTT) due to their advantages of low cost and toxicity, good biodegradation, and strong NIR absorption over a wide wavelength range. Nevertheless, OSMDs have limited applicability in PTT due to their low photothermal conversion efficiency and inadequate destruction of tumor regions that are nonirradiated by NIR light. However, they can also act as photosensitizers (PSs) to produce reactive oxygen species (ROS), which can be further eradicated by using ROS-related therapies to address the above limitations of PTT. In this review, the synergistic mechanism, composition, and properties of photodynamic therapy (PDT)-PTT nanoplatforms were comprehensively discussed. In addition, some specific strategies for further improving the combined PTT and PDT based on OSMDs for cancer to completely eradicate cancer cells were outlined. These strategies include performing image-guided co-therapy, enhancing tumor infiltration, increasing H2O2 or O2 in the tumor microenvironment, and loading anticancer drugs onto nanoplatforms to enable combined therapy with phototherapy and chemotherapy. Meanwhile, the intriguing prospects and challenges of this treatment modality were also summarized with a focus on the future trends of its clinical application.
Collapse
Affiliation(s)
- Shuang Guo
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Dongyu Gu
- College of Marine Science and Environment, Dalian Ocean University, Dalian, 116023, China
| | - Yi Yang
- School of Light Industry and Chemical Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Jing Tian
- School of Biological Engineering, Dalian Polytechnic University, Dalian, 116034, China.
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
26
|
Lin S, Zhu L, Li Z, Yue S, Wang Z, Xu Y, Zhang Y, Gao Q, Chen J, Yin T, Niu L, Geng J. Ultrasound-responsive glycopolymer micelles for targeted dual drug delivery in cancer therapy. Biomater Sci 2023; 11:6149-6159. [PMID: 37548310 DOI: 10.1039/d3bm01101a] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Controlled drug release of nanoparticles was achieved by irreversibly disrupting polymer micelles through high-intensity focused ultrasound (HIFU) induction. An ultrasound-responsive block copolymer was synthesized, comprising an end-functional Eosin Y fluorophore, 2-tetrahydropyranyl acrylate (THPA), and acrylate mannose (MAN). The block copolymer was then self-assembled to produce micelles. The chemotherapy drug dasatinib (DAS) and the sonodynamic therapy agent methylene blue (MB) were encapsulated by the self-assembly of the block copolymer. This targeted nanoparticle enables sonodynamic therapy through high-intensity focused ultrasound while triggering nanoparticle disassembly for controlled drug release. The ultrasound-mediated, non-invasive strategy provides external spatiotemporal control for targeted tumour treatment.
Collapse
Affiliation(s)
- Shanmeng Lin
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liwei Zhu
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhiying Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Siyuan Yue
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Zhaohan Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Youwei Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yichuan Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Quan Gao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Jie Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Ting Yin
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Lili Niu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Jin Geng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
27
|
Wang T, Wu C, Hu Y, Zhang Y, Ma J. Stimuli-responsive nanocarrier delivery systems for Pt-based antitumor complexes: a review. RSC Adv 2023; 13:16488-16511. [PMID: 37274408 PMCID: PMC10233443 DOI: 10.1039/d3ra00866e] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/30/2023] [Indexed: 06/06/2023] Open
Abstract
Platinum-based anticancer drugs play a crucial role in the clinical treatment of various cancers. However, the application of platinum-based drugs is heavily restricted by their severe toxicity and drug resistance/cross resistance. Various drug delivery systems have been developed to overcome these limitations of platinum-based chemotherapy. Stimuli-responsive nanocarrier drug delivery systems as one of the most promising strategies attract more attention. And huge progress in stimuli-responsive nanocarrier delivery systems of platinum-based drugs has been made. In these systems, a variety of triggers including endogenous and extracorporeal stimuli have been employed. Endogenous stimuli mainly include pH-, thermo-, enzyme- and redox-responsive nanocarriers. Extracorporeal stimuli include light-, magnetic field- and ultrasound responsive nanocarriers. In this review, we present the recent advances in stimuli-responsive drug delivery systems with different nanocarriers for improving the efficacy and reducing the side effects of platinum-based anticancer drugs.
Collapse
Affiliation(s)
- Tianshuai Wang
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Chen Wu
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Yanggen Hu
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Yan Zhang
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| | - Junkai Ma
- Hubei Key Lab of Wudang Local Chinese Medicine Research, Hubei University of Medicine Shiyan 442000 Hubei China
- College of Pharmaceutical Sciences, Hubei University of Medicine Shiyan 442000 Hubei China
| |
Collapse
|
28
|
Alcaraz PE, Davidson SJ, Shreeve E, Meuschke R, Romanowski M, Witte RS, Porter TR, Matsunaga TO. Thermal and Acoustic Stabilization Of Volatile Phase-Change Contrast Agents Via Layer-By-Layer Assembly. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1058-1069. [PMID: 36797095 PMCID: PMC10050125 DOI: 10.1016/j.ultrasmedbio.2022.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 05/11/2023]
Abstract
OBJECTIVE Phase-change contrast agents (PCCAs) are perfluorocarbon nanodroplets (NDs) that have been widely studied for ultrasound imaging in vitro, pre-clinical studies, and most recently incorporated a variant of PCCAs, namely a microbubble-conjugated microdroplet emulsion, into the first clinical studies. Their properties also make them attractive candidates for a variety of diagnostic and therapeutic applications including drug-delivery, diagnosis and treatment of cancerous and inflammatory diseases, as well as tumor-growth tracking. However, control over the thermal and acoustic stability of PCCAs both in vivo and in vitro has remained a challenge for expanding the potential utility of these agents in novel clinical applications. As such, our objective was to determine the stabilizing effects of layer-by-layer assemblies and its effect on both thermal and acoustic stability. METHODS We utilized layer-by-layer (LBL) assemblies to coat the outer PCCA membrane and characterized layering by measuring zeta potential and particle size. Stability studies were conducted by; 1) incubating the LBL-PCCAs at atmospheric pressure at 37∘C and 45∘C followed by; 2) ultrasound-mediated activation at 7.24 MHz and peak-negative pressures ranging from 0.71 - 5.48 MPa to ascertain nanodroplet activation and resultant microbubble persistence. The thermal and acoustic properties of decafluorobutane gas-condensed nanodroplets (DFB-NDs) layered with 6 and 10 layers of charge-alternating biopolymers, (LBL6NDs and LBL10NDs) respectively, were studied and compared to non-layered DFB-NDs. Half-life determinations were conducted at both 37∘C and 45∘C with acoustic droplet vaporization (ADV) measurements occurring at 23∘C. DISCUSSION Successful application of up to 10 layers of alternating positive and negatively charged biopolymers onto the surface membrane of DFB-NDs was demonstrated. Two major claims were substantiated in this study; namely, (1) biopolymeric layering of DFB-NDs imparts a thermal stability up to an extent; and, (2) both LBL6NDs and LBL10NDs did not appear to alter particle acoustic vaporization thresholds, suggesting that the thermal stability of the particle may not necessarily be coupled with particle acoustic vaporization thresholds. CONCLUSION Results demonstrate that the layered PCCAs had higher thermal stability, where the half-lifes of the LBLxNDs are significantly increased after incubation at 37∘C and 45∘C. Furthermore, the acoustic vaporization profiles the DFB-NDs, LBL6NDs, and LBL10NDs show that there is no statistically significant difference between the acoustic vaporization energy required to initiate acoustic droplet vaporization.
Collapse
Affiliation(s)
- Pedro Enrique Alcaraz
- College of Optical Sciences, University of Arizona, 1630 E University Blvd., Tucson, AZ 85721 United States; Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Medical Imaging, University of Arizona, Tucson, AZ. 85719 United States
| | - Skylar J Davidson
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Evan Shreeve
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Rainee Meuschke
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Marek Romanowski
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85719 United States
| | - Russell S Witte
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Materials Science and Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Medical Imaging, University of Arizona, Tucson, AZ. 85719 United States
| | - Thomas R Porter
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Terry O Matsunaga
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85719 United States; Department of Medical Imaging, University of Arizona, Tucson, AZ. 85719 United States.
| |
Collapse
|
29
|
Mechanotransduction in tumor dynamics modeling. Phys Life Rev 2023; 44:279-301. [PMID: 36841159 DOI: 10.1016/j.plrev.2023.01.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Mechanotherapy is a groundbreaking approach to impact carcinogenesis. Cells sense and respond to mechanical stimuli, translating them into biochemical signals in a process known as mechanotransduction. The impact of stress on tumor growth has been studied in the last three decades, and many papers highlight the role of mechanics as a critical self-inducer of tumor fate at the in vitro and in vivo biological levels. Meanwhile, mathematical models attempt to determine laws to reproduce tumor dynamics. This review discusses biological mechanotransduction mechanisms and mathematical-biomechanical models together. The aim is to provide a common framework for the different approaches that have emerged in the literature from the perspective of tumor avascularity and to provide insight into emerging mechanotherapies that have attracted interest in recent years.
Collapse
|
30
|
Collado-Lara G, Heymans SV, Rovituso M, Sterpin E, D'hooge J, Vos HJ, Abeele KVD, de Jong N. Analytic prediction of droplet vaporization events to estimate the precision of ultrasound-based proton range verification. Med Phys 2023. [PMID: 36856326 DOI: 10.1002/mp.16327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND The safety and efficacy of proton therapy is currently hampered by range uncertainties. The combination of ultrasound imaging with injectable radiation-sensitive superheated nanodroplets was recently proposed for in vivo range verification. The proton range can be estimated from the distribution of nanodroplet vaporization events, which is stochastically related to the stopping distribution of protons, as nanodroplets are vaporized by protons reaching their maximal LET at the end of their range. PURPOSE Here, we aim to estimate the range estimation precision of this technique. As for any stochastic measurement, the precision will increase with the sample size, that is, the number of detected vaporizations. Thus, we first develop and validate a model to predict the number of vaporizations, which is then applied to estimate the range verification precision for a set of conditions (droplet size, droplet concentration, and proton beam parameters). METHODS Starting from the thermal spike theory, we derived a model that predicts the expected number of droplet vaporizations in an irradiated sample as a function of the droplet size, concentration, and number of protons. The model was validated by irradiating phantoms consisting of size-sorted perfluorobutane droplets dispersed in an aqueous matrix. The number of protons was counted with an ionization chamber, and the droplet vaporizations were recorded and counted individually using high frame rate ultrasound imaging. After validation, the range estimate precision was determined for different conditions using a Monte Carlo algorithm. RESULTS A good agreement between theory and experiments was observed for the number of vaporizations, especially for large (5.8 ± 2.2 µm) and medium (3.5 ± 1.1 µm) sized droplets. The number of events was lower than expected in phantoms with small droplets (2.0 ± 0.7 µm), but still within the same order of magnitude. The inter-phantom variability was considerably larger (up to 30x) than predicted by the model. The validated model was then combined with Monte Carlo simulations, which predicted a theoretical range retrieval precision improving with the square-root of the number of vaporizations, and degrading at high beam energies due to range straggling. For single pencil beams with energies between 70 and 240 MeV, a range verification precision below 1% of the range required perfluorocarbon concentrations in the order of 0.3-2.4 µM. CONCLUSION We proposed and experimentally validated a model to provide a quick estimate of the number of vaporizations for a given set of conditions (droplet size, droplet concentration, and proton beam parameters). From this model, promising range verification performances were predicted for realistic perfluorocarbon concentrations. These findings are an incentive to move towards preclinical studies, which are critical to assess the achievable droplet distribution in and around the tumor, and hence the in vivo range verification precision.
Collapse
Affiliation(s)
- Gonzalo Collado-Lara
- Biomedical Engineering, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sophie V Heymans
- Biomedical Engineering, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Physics, KU Leuven Campus Kulak, Kortrijk, Belgium.,Department of Cardiovascular Sciences, Leuven KU, Leuven, Belgium
| | | | - Edmond Sterpin
- Department of Oncology, Leuven KU, Leuven, Belgium.,Center of Molecular Imaging, Radiotherapy and Oncology, IREC Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jan D'hooge
- Department of Cardiovascular Sciences, Leuven KU, Leuven, Belgium
| | - Hendrik J Vos
- Biomedical Engineering, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Nico de Jong
- Biomedical Engineering, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
31
|
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: 32] [Impact Index Per Article: 32.0] [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
|
32
|
Recent progress in theranostic microbubbles. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
33
|
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: 1.0] [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
|
34
|
Xin L, Zhang C, Chen J, Jiang Y, Liu Y, Jin P, Wang X, Wang G, Huang P. Ultrasound-Activatable Phase-Shift Nanoparticle as a Targeting Antibacterial Agent for Efficient Eradication of Pseudomonas aeruginosa Biofilms. ACS APPLIED MATERIALS & INTERFACES 2022; 14:47420-47431. [PMID: 36222290 DOI: 10.1021/acsami.2c13166] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Biofilms are physical barriers composed of extracellular polymeric substances (EPS) that enable planktonic bacteria to resist host responses and antibacterial treatments, complicating efforts to clear bacteria such as Pseudomonas aeruginosa (P. aeruginosa) and thereby contributing to persistently chronic infections. As such, it is critical to develop a robust antimicrobial strategy capable of effectively eradicating P. aeruginosa biofilms and to further address aggressive clinical infection. In this study, ultrasound-activatable targeted nanoparticles were designed by using poly(lactic-co-glycolic acid) (PLGA) nanoparticles to encapsulate phase-transformable perfluoropentane (PFP) and the antibiotic meropenem via a double emulsion approach, followed by conjugation with anti-P. aeruginosa antibodies. In this strategy, ultrasound exposure can trigger PFP to produce microbubbles, inducing ultrasonic cavitation effects that can disrupt EPS components and allow nanoparticles to release meropenem to kill P. aeruginosa directly and accelerate the associated wound healing. These nanoparticles eradicated biofilms effectively and cleared bacteria in vitro as well as exhibited potent anti-infective activity in vivo. In summary, this study demonstrates the efficacy of a sonobactericidal strategy as a means of effectively and reliably eliminating biofilms.
Collapse
Affiliation(s)
- Lei Xin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Chao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Yifan Jiang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Yajing Liu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Xue Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou311215, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou310053, China
| |
Collapse
|
35
|
Huang J, Wang H, Huang L, Zhou Y. Phospholipid-mimicking block, graft, and block-graft copolymers for phase-transition microbubbles as ultrasound contrast agents. Front Pharmacol 2022; 13:968835. [PMCID: PMC9606805 DOI: 10.3389/fphar.2022.968835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Lipid and polymer microbubbles (MBs) are widely used as ultrasound contrast agents in clinical diagnosis, and possess great potential in ultrasound-mediated therapy due to their drug loading function. However, overcoming the limitations of stability and echo enhancement of MBs are still a considerable challenge.Methods: A series novel block, graft and block-graft copolymers was proposed and prepared in this work, and these copolymers were used as shells to encapsulate perfluoropentane as ultrasound contrast agents. First, block, graft and block-graft copolymers with different topological structures were prepared. Then, these copolymers were prepared into block copolymer phase-transition MBs, graft copolymer phase-transition MBs, and block-graft copolymer phase-transition MBs, respectively. Finally, the dexamethasone was used for drug-loaded phase-transition microbubbles model to explore the potential of theranostic microbubbles.Results: Finally, these three resulting copolymer MBs with average size of 4–5 μm exhibited well enhancement of ultrasound imaging under the influence of different frequencies and mechanical index, and they exhibited a longer contrast-enhanced ultrasound imaging time and higher resistance to mechanical index compared with SonoVue in vitro and in vivo. In vitro drug release results also showed that these copolymer MBs could encapsulate dexamethasone drugs, and the drug release could be enhanced by ultrasonic triggering. These copolymer MBs were therapeutic MBs for targeted triggering drug release.Conclusion: Therefore, the feasibility of block, graft, and block-graft copolymers as ultrasonic contrast agents was verified, and their ultrasonic enhancement performance in vitro and in vivo was compared. The ultrasound contrast agents developed in this work have excellent development potential in comprehensive diagnosis and treatment.
Collapse
Affiliation(s)
- Jianbo Huang
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wang
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hong Wang,
| | - Lei Huang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Yuqing Zhou
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Cen J, Ye X, Liu X, Pan W, Zhang L, Zhang G, He N, Shen A, Hu J, Liu S. Fluorinated Copolypeptide‐Stabilized Microbubbles with Maleimide‐Decorated Surfaces as Long‐Term Ultrasound Contrast Agents. Angew Chem Int Ed Engl 2022; 61:e202209610. [DOI: 10.1002/anie.202209610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Jie Cen
- Department of Ultrasound Imaging & Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China 17 Lujiang Road Hefei, Anhui Province 230001 China
- CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China 96 Jinzhai Road Hefei, Anhui Province 230026 China
| | - Xianjun Ye
- Department of Ultrasound Imaging & Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China 17 Lujiang Road Hefei, Anhui Province 230001 China
| | - Xiao Liu
- Department of Ultrasound Imaging & Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China 17 Lujiang Road Hefei, Anhui Province 230001 China
| | - Wenhao Pan
- CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China 96 Jinzhai Road Hefei, Anhui Province 230026 China
| | - Lei Zhang
- Department of Ultrasound Imaging & Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China 17 Lujiang Road Hefei, Anhui Province 230001 China
| | - Guoying Zhang
- CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China 96 Jinzhai Road Hefei, Anhui Province 230026 China
| | - Nianan He
- Department of Ultrasound Imaging & Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China 17 Lujiang Road Hefei, Anhui Province 230001 China
| | - Aizong Shen
- Department of Ultrasound Imaging & Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Science and Technology of China 17 Lujiang Road Hefei, Anhui Province 230001 China
| | - Jinming Hu
- CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China 96 Jinzhai Road Hefei, Anhui Province 230026 China
| | - Shiyong Liu
- CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China 96 Jinzhai Road Hefei, Anhui Province 230026 China
| |
Collapse
|
37
|
Rasel MSI, Mohona FA, Akter W, Kabir S, Chowdhury AA, Chowdhury JA, Hassan MA, Al Mamun A, Ghose DK, Ahmad Z, Khan FS, Bari MF, Rahman MS, Amran MS. Exploration of Site-Specific Drug Targeting-A Review on EPR-, Stimuli-, Chemical-, and Receptor-Based Approaches as Potential Drug Targeting Methods in Cancer Treatment. JOURNAL OF ONCOLOGY 2022; 2022:9396760. [PMID: 36284633 PMCID: PMC9588330 DOI: 10.1155/2022/9396760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
Cancer has been one of the most dominant causes of mortality globally over the last few decades. In cancer treatment, the selective targeting of tumor cells is indispensable, making it a better replacement for conventional chemotherapies by diminishing their adverse side effects. While designing a drug to be delivered selectively in the target organ, the drug development scientists should focus on various factors such as the type of cancer they are dealing with according to which drug, targeting moieties, and pharmaceutical carriers should be targeted. All published articles have been collected regarding cancer and drug-targeting approaches from well reputed databases including MEDLINE, Embase, Cochrane Library, CENTRAL and ClinicalTrials.gov, Science Direct, PubMed, Scopus, Wiley, and Springer. The articles published between January 2010 and December 2020 were considered. Due to the existence of various mechanisms, it is challenging to choose which one is appropriate for a specific case. Moreover, a combination of more than one approach is often utilized to achieve optimal drug effects. In this review, we have summarized and highlighted central mechanisms of how the targeted drug delivery system works in the specific diseased microenvironment, along with the strategies to make an approach more effective. We have also included some pictorial illustrations to have a precise idea about different types of drug targeting. The core contribution of this work includes providing a cancer drug development scientist with a broad preliminary idea to choose the appropriate approach among the various targeted drug delivery mechanisms. Also, the study will contribute to improving anticancer treatment approaches by providing a pathway for lesser side effects observed in conventional chemotherapeutic techniques.
Collapse
Affiliation(s)
- Md. Shamiul Islam Rasel
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Farhana Afrin Mohona
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Wahida Akter
- College of Pharmacy, University of Houston, Houston, USA
| | - Shaila Kabir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Abu Asad Chowdhury
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Md. Abul Hassan
- Department of Science & Technology, Tokushima University Graduate School, Tokushima, Japan
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Dipayon Krisna Ghose
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, King Khalid University, Abha 61413, Saudi Arabia
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Md. Fazlul Bari
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Shah Amran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| |
Collapse
|
38
|
Yeingst TJ, Arrizabalaga JH, Hayes DJ. Ultrasound-Induced Drug Release from Stimuli-Responsive Hydrogels. Gels 2022; 8:554. [PMID: 36135267 PMCID: PMC9498906 DOI: 10.3390/gels8090554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/16/2022] Open
Abstract
Stimuli-responsive hydrogel drug delivery systems are designed to release a payload when prompted by an external stimulus. These platforms have become prominent in the field of drug delivery due to their ability to provide spatial and temporal control for drug release. Among the different external triggers that have been used, ultrasound possesses several advantages: it is non-invasive, has deep tissue penetration, and can safely transmit acoustic energy to a localized area. This review summarizes the current state of understanding about ultrasound-responsive hydrogels used for drug delivery. The mechanisms of inducing payload release and activation using ultrasound are examined, along with the latest innovative formulations and hydrogel design strategies. We also report on the most recent applications leveraging ultrasound activation for both cancer treatment and tissue engineering. Finally, the future perspectives offered by ultrasound-sensitive hydrogels are discussed.
Collapse
Affiliation(s)
- Tyus J. Yeingst
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
| | - Julien H. Arrizabalaga
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
| | - Daniel J. Hayes
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
- Materials Research Institute, Millennium Science Complex, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
- The Huck Institute of the Life Sciences, Millennium Science Complex, The Pennsylvania State University, University Park, Centre County, PA 16802, USA
| |
Collapse
|
39
|
Falatah HA, Lacerda Q, Chaga M, Wessner CE, Forsberg F, Leeper DB, Eisenbrey JR. Activation of Phase Change Contrast Agents Using Ionizing Radiation. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:2365-2371. [PMID: 34866197 PMCID: PMC9793720 DOI: 10.1002/jum.15910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
The feasibility of activating phase change contrast agents (PCCA) made from Definity (Lantheus Medical Imaging) using X-rays was investigated. A 10 mL of Definity PCCA (0.1 mL PCCA/mL) were injected into gelatin phantoms and irradiated using doses of 0, 30, 50, and 100 Gy. Size distribution and PCCA activation were measured after irradiation. Definity PCCAs were activated at a threshold of 50 Gy. Changes were visible with microscopy, visual inspection of T-flasks, and ultrasound imaging of gelatin phantoms. Moreover, increasing the radiation dose above 50 Gy appeared to further activate PCCA. These results indicate Definity PCCAs may be useful for ultrasound-based radiation dosimetry.
Collapse
Affiliation(s)
- Hebah A Falatah
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Quezia Lacerda
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Michael Chaga
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Corinne E Wessner
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dennis B Leeper
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
40
|
Chiang PH, Fan CH, Jin Q, Yeh CK. Enhancing Doxorubicin Delivery in Solid Tumor by Superhydrophobic Amorphous Calcium Carbonate-Doxorubicin Silica Nanoparticles with Focused Ultrasound. Mol Pharm 2022; 19:3894-3905. [PMID: 36018041 DOI: 10.1021/acs.molpharmaceut.2c00384] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The current approach of delivering chemotherapy via pH-sensitive amorphous calcium carbonate-doxorubicin silica nanoparticles (ADS NPs) faces the challenge of insufficient drug dose due to drug instability within the bloodstream and poor tumor penetration. To overcome these long-standing obstacles, we proposed a superhydrophobic coating on the surface of the ADS NPs that could be easily modified via fluorination (ADSF NPs). The surface of fluorinated ADS NPs was further modified with a phospholipid layer to reduce aggregation and improve biocompatibility (ADSFL NPs). The contact angle and mean size of ADSFL NPs were 30.2 ± 4.4° and 353.1 ± 54.2 nm, respectively. The superhydrophobic layer generated interfacial nanobubbles on the outer shell of the NPs that reduced water-induced leakage of doxorubicin (DOX) sevenfold compared with the uncoated group and induced a cavitation effect upon ultrasound (US) sonication. Moreover, release of DOX from the ADSFL NPs could be triggered by US, and this release was further improved 1.6-fold in acidic aqueous conditions, indicating that the ADSFL NPs retained pH responsiveness. Enhanced sonography contrast and histological examination demonstrated that US could trigger cavitation activities from ADSFL NPs in vivo to induce vessel disruption and enhance the fluorescence intensity of DOX within the tumor region threefold under US imaging guidance compared with the ADSFL NPs-only group.
Collapse
Affiliation(s)
- Pei-Hua Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Road, Tainan 701, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, No. 1, University Road, Tainan 701, Taiwan
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan 430022, Hubei, China
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan
| |
Collapse
|
41
|
Cen J, Ye X, Liu X, Pan W, Zhang L, Zhang G, He N, Shen A, Hu J, Liu S. Fluorinated Copolypeptide‐Stabilized Microbubbles with Maleimide‐Decorated Surfaces as Long‐Term Ultrasound Contrast Agents. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Jie Cen
- China University of Science and Technology Department of Polymer Science and Engineering CHINA
| | - Xianjun Ye
- China University of Science and Technology Department of Ultrasound Imaging CHINA
| | - Xiao Liu
- China University of Science and Technology Department of Ultrasound Imaging CHINA
| | - Wenhao Pan
- China University of Science and Technology Department of Polymer Science and Engineering CHINA
| | - Lei Zhang
- China University of Science and Technology Department of Pharmacy CHINA
| | - Guoying Zhang
- China University of Science and Technology Department of Polymer Science and Engineering CHINA
| | - Nianan He
- China University of Science and Technology Department of Ultrasound Imaging CHINA
| | - Aizong Shen
- China University of Science and Technology Department of Pharmacy CHINA
| | - Jinming Hu
- China University of Science and Technology Department of Polymer Science and Engineering 96 Jinzhai RoadDepartment of Polymer Science and EngineeringUniversity of Science and Technology of China 230026 Hefei CHINA
| | - Shiyong Liu
- University of Science and Technology of China Department of Polymer Science and Engineering 96 Jinzhai Road 230026 Hefei CHINA
| |
Collapse
|
42
|
A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
43
|
Maciulevičius M, Tamošiūnas M, Navickaitė D, Šatkauskas S, Venslauskas MS. Free- and liposomal- doxorubicin delivery via microbubble inertial cavitation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Maciulevičius M, Tamošiūnas M, Jurkonis R, Šatkauskas S. Dosimetric Assessment of Antitumor Treatment by enhanced Bleomycin Delivery via Electroporation and Sonoporation. Bioelectrochemistry 2022; 146:108153. [DOI: 10.1016/j.bioelechem.2022.108153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/15/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
|
45
|
Durham PG, Kim J, Eltz KM, Caskey CF, Dayton PA. Polyvinyl Alcohol Cryogels for Acoustic Characterization of Phase-Change Contrast Agents. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:954-960. [PMID: 35246338 PMCID: PMC9012345 DOI: 10.1016/j.ultrasmedbio.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 05/03/2023]
Abstract
Phase-change contrast agents (PCCAs) consisting of lipid-encapsulated low-boiling-point perfluorocarbons can be used in conjunction with ultrasound for diagnostic and therapeutic applications. One benefit of PCCAs is site-specific activation, whereby the liquid core is acoustically vaporized into a bubble detectable via ultrasound imaging. For further evaluation of PCCAs in a variety of applications, it is useful to disperse these nanodroplets into an acoustically compatible stationary matrix. However, many traditional phantom preparations require heating, which causes premature thermal activation of low-boiling-point PCCAs. Polyvinyl alcohol (PVA) cryogels do not require heat to set. Here we propose a simple method for the incorporation of the low-boiling-point PCCAs using octafluoropropane (OFP) and decafluorobutane (DFB) into PVA cryogels for a variety of acoustic characterization applications. We determined the utility of the phantoms by activating droplets with a focused transducer, visualizing the lesions with ultrasound imaging. At 1 MHz, droplet activation was consistently observed at 2.0 and 4.0 MPa for OFP and DFB, respectively.
Collapse
Affiliation(s)
- Phillip G Durham
- Department of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, USA; Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, North Carolina, USA.
| | - Jinwook Kim
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Katherine M Eltz
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Charles F Caskey
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, Tennessee, USA; Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Paul A Dayton
- Department of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, North Carolina, USA; Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, North Carolina, USA
| |
Collapse
|
46
|
Zamani R, Bizari D, Heiat M. Synthesis and characterization of phase shift dextran stabilized nanodroplets for ultrasound-induced cancer therapy: A novel nanobiotechnology approach. J Biotechnol 2022; 350:17-23. [DOI: 10.1016/j.jbiotec.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/26/2022] [Accepted: 04/08/2022] [Indexed: 11/25/2022]
|
47
|
Delaney LJ, Isguven S, Eisenbrey JR, Hickok NJ, Forsberg F. Making waves: how ultrasound-targeted drug delivery is changing pharmaceutical approaches. MATERIALS ADVANCES 2022; 3:3023-3040. [PMID: 35445198 PMCID: PMC8978185 DOI: 10.1039/d1ma01197a] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/23/2022] [Indexed: 05/06/2023]
Abstract
Administration of drugs through oral and intravenous routes is a mainstay of modern medicine, but this approach suffers from limitations associated with off-target side effects and narrow therapeutic windows. It is often apparent that a controlled delivery of drugs, either localized to a specific site or during a specific time, can increase efficacy and bypass problems with systemic toxicity and insufficient local availability. To overcome some of these issues, local delivery systems have been devised, but most are still restricted in terms of elution kinetics, duration, and temporal control. Ultrasound-targeted drug delivery offers a powerful approach to increase delivery, therapeutic efficacy, and temporal release of drugs ranging from chemotherapeutics to antibiotics. The use of ultrasound can focus on increasing tissue sensitivity to the drug or actually be a critical component of the drug delivery. The high spatial and temporal resolution of ultrasound enables precise location, targeting, and timing of drug delivery and tissue sensitization. Thus, this noninvasive, non-ionizing, and relatively inexpensive modality makes the implementation of ultrasound-mediated drug delivery a powerful method that can be readily translated into the clinical arena. This review covers key concepts and areas applied in the design of different ultrasound-mediated drug delivery systems across a variety of clinical applications.
Collapse
Affiliation(s)
- Lauren J Delaney
- Department of Radiology, Thomas Jefferson University 132 S. 10th Street, Main 763 Philadelphia PA 19107 USA +1 (215) 955-4870
| | - Selin Isguven
- Department of Radiology, Thomas Jefferson University 132 S. 10th Street, Main 763 Philadelphia PA 19107 USA +1 (215) 955-4870
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street Philadelphia PA 19107 USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University 132 S. 10th Street, Main 763 Philadelphia PA 19107 USA +1 (215) 955-4870
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Thomas Jefferson University, 1015 Walnut Street Philadelphia PA 19107 USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University 132 S. 10th Street, Main 763 Philadelphia PA 19107 USA +1 (215) 955-4870
| |
Collapse
|
48
|
Athanassiadis AG, Ma Z, Moreno-Gomez N, Melde K, Choi E, Goyal R, Fischer P. Ultrasound-Responsive Systems as Components for Smart Materials. Chem Rev 2022; 122:5165-5208. [PMID: 34767350 PMCID: PMC8915171 DOI: 10.1021/acs.chemrev.1c00622] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Indexed: 02/06/2023]
Abstract
Smart materials can respond to stimuli and adapt their responses based on external cues from their environments. Such behavior requires a way to transport energy efficiently and then convert it for use in applications such as actuation, sensing, or signaling. Ultrasound can carry energy safely and with low losses through complex and opaque media. It can be localized to small regions of space and couple to systems over a wide range of time scales. However, the same characteristics that allow ultrasound to propagate efficiently through materials make it difficult to convert acoustic energy into other useful forms. Recent work across diverse fields has begun to address this challenge, demonstrating ultrasonic effects that provide control over physical and chemical systems with surprisingly high specificity. Here, we review recent progress in ultrasound-matter interactions, focusing on effects that can be incorporated as components in smart materials. These techniques build on fundamental phenomena such as cavitation, microstreaming, scattering, and acoustic radiation forces to enable capabilities such as actuation, sensing, payload delivery, and the initiation of chemical or biological processes. The diversity of emerging techniques holds great promise for a wide range of smart capabilities supported by ultrasound and poses interesting questions for further investigations.
Collapse
Affiliation(s)
- Athanasios G. Athanassiadis
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Zhichao Ma
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Nicolas Moreno-Gomez
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
- Institute
of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Kai Melde
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Eunjin Choi
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
- Institute
of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Rahul Goyal
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
| | - Peer Fischer
- Micro,
Nano, and Molecular Systems Group, Max Planck
Institute for Intelligent Systems, Heisenbergstrasse 3, 70569 Stuttgart, Germany
- Institute
of Physical Chemistry, University of Stuttgart, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| |
Collapse
|
49
|
Xavierselvan M, Cook J, Duong J, Diaz N, Homan K, Mallidi S. Photoacoustic nanodroplets for oxygen enhanced photodynamic therapy of cancer. PHOTOACOUSTICS 2022; 25:100306. [PMID: 34917471 PMCID: PMC8666552 DOI: 10.1016/j.pacs.2021.100306] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/07/2021] [Accepted: 09/21/2021] [Indexed: 05/20/2023]
Abstract
Photodynamic therapy (PDT) is a well-known cancer therapy that utilizes light to excite a photosensitizer and generate cytotoxic reactive oxygen species (ROS). The efficacy of PDT primarily depends on the photosensitizer and oxygen concentration in the tumor. Hypoxia in solid tumors promotes treatment resistance, resulting in poor PDT outcomes. Hence, there is a need to combat hypoxia while delivering sufficient photosensitizer to the tumor for ROS generation. Here we showcase our unique theranostic perfluorocarbon nanodroplets as a triple agent carrier for oxygen, photosensitizer, and indocyanine green that enables light triggered spatiotemporal delivery of oxygen to the tumors. We evaluated the characteristics of the nanodroplets and validated their ability to deliver oxygen via photoacoustic monitoring of blood oxygen saturation and subsequent PDT efficacy in a murine subcutaneous tumor model. The imaging results were validated with an oxygen sensing probe, which showed a 9.1 fold increase in oxygen content inside the tumor, following systemic administration of the nanodroplets. These results were also confirmed with immunofluorescence. In vivo studies showed that nanodroplets held higher rates of treatment efficacy than a clinically available benzoporphyrin derivative formulation. Histological analysis showed higher necrotic area within the tumor with perfluoropentane nanodroplets. Overall, the photoacoustic nanodroplets can significantly enhance image-guided PDT and has demonstrated substantial potential as a valid theranostic option for patient-specific photodynamic therapy-based treatments.
Collapse
Key Words
- 1O2, singlet oxygen
- BPD, benzoporphyrin derivative
- DLS, dynamic light scattering
- DPPC, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine
- DSPE-mPEG, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]
- H&E, hematoxylin and eosin
- HbT, total hemoglobin
- Hypoxia
- ICG, indocyanine green
- IF, immunofluorescence
- Image guided PDT
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- NIR, near infrared radiation
- PA, photoacoustic
- PBS, phosphate buffered saline
- PDT, photodynamic therapy
- PFC, perfluorocarbon
- PFP, perfluoropentane
- PS, photosensitizer
- Perfluorocarbon nanodroplets
- Photoacoustic imaging
- Photodynamic therapy
- ROS, reactive oxygen species
- SOSG, singlet oxygen sensor green
- StO2, oxygen saturation
- TBAI, tertbutylammonium iodide
- pO2, partial pressure of oxygen
Collapse
Affiliation(s)
- Marvin Xavierselvan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | - Jeanne Duong
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Nashielli Diaz
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | | | - Srivalleesha Mallidi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
50
|
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: 13.0] [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
|