1
|
Zhang X, He N, Zhang L, Dai T, Sun Z, Shi Y, Li S, Yu N. Application of high intensity focused ultrasound combined with nanomaterials in anti-tumor therapy. Drug Deliv 2024; 31:2342844. [PMID: 38659328 PMCID: PMC11047217 DOI: 10.1080/10717544.2024.2342844] [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: 04/03/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
High intensity focused ultrasound (HIFU) has demonstrated its safety, efficacy and noninvasiveness in the ablation of solid tumor. However, its further application is limited by its inherent deficiencies, such as postoperative recurrence caused by incomplete ablation and excessive intensity affecting surrounding healthy tissues. Recent research has indicated that the integration of nanomaterials with HIFU exhibits a promising synergistic effect in tumor ablation. The concurrent utilization of nanomaterials with HIFU can help overcome the limitations of HIFU by improving targeting and ablation efficiency, expanding operation area, increasing operation accuracy, enhancing stability and bio-safety during the process. It also provides a platform for multi-therapy and multi-mode imaging guidance. The present review comprehensively expounds upon the synergistic mechanism between nanomaterials and HIFU, summarizes the research progress of nanomaterials as cavitation nuclei and drug carriers in combination with HIFU for tumor ablation. Furthermore, this review highlights the potential for further exploration in the development of novel nanomaterials that enhance the synergistic effect with HIFU on tumor ablation.
Collapse
Affiliation(s)
- Xuehui Zhang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Liang Zhang
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zihan Sun
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Yuqing Shi
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Gong N, Alameh MG, El-Mayta R, Xue L, Weissman D, Mitchell MJ. Enhancing in situ cancer vaccines using delivery technologies. Nat Rev Drug Discov 2024; 23:607-625. [PMID: 38951662 DOI: 10.1038/s41573-024-00974-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/03/2024]
Abstract
In situ cancer vaccination refers to any approach that exploits tumour antigens available at a tumour site to induce tumour-specific adaptive immune responses. These approaches hold great promise for the treatment of many solid tumours, with numerous candidate drugs under preclinical or clinical evaluation and several products already approved. However, there are challenges in the development of effective in situ cancer vaccines. For example, inadequate release of tumour antigens from tumour cells limits antigen uptake by immune cells; insufficient antigen processing by antigen-presenting cells restricts the generation of antigen-specific T cell responses; and the suppressive immune microenvironment of the tumour leads to exhaustion and death of effector cells. Rationally designed delivery technologies such as lipid nanoparticles, hydrogels, scaffolds and polymeric nanoparticles are uniquely suited to overcome these challenges through the targeted delivery of therapeutics to tumour cells, immune cells or the extracellular matrix. Here, we discuss delivery technologies that have the potential to reduce various clinical barriers for in situ cancer vaccines. We also provide our perspective on this emerging field that lies at the interface of cancer vaccine biology and delivery technologies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, Center for BioAnalytical Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, George Mason University, Fairfax, VA, USA
| | - Rakan El-Mayta
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Penn institute for RNA innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Wang K, Zhang M, Geng Z, Zhang S, Deng Z, Tan J, Zhang Q, Jiao Z, Gu N. One-Step Preparation of Magnetic Lipid Bubbles: Magnetothermal Effect Induces the Simultaneous Formation of Gas Nuclei and Self-Assembly of Phospholipids. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30755-30765. [PMID: 38847111 DOI: 10.1021/acsami.4c03788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
In recent years, enveloped micro-nanobubbles have garnered significant attention in research due to their commendable stability, biocompatibility, and other notable properties. Currently, the preparation methods of enveloped micro-nanobubbles have limitations such as complicated preparation process, large bubble size, wide distribution range, low yield, etc. There exists an urgent demand to devise a simple and efficient method for the preparation of enveloped micro-nanobubbles, ensuring both high concentration and a uniform particle size distribution. Magnetic lipid bubbles (MLBs) are a multifunctional type of enveloped micro-nanobubble combining magnetic nanoparticles with lipid-coated bubbles. In this study, MLBs are prepared simply and efficiently by a magneto internal heat bubble generation process based on the interfacial self-assembly of iron oxide nanoparticles induced by the thermogenic effect in an alternating magnetic field. The mean hydrodynamic diameter of the MLBs obtained was 384.9 ± 8.5 nm, with a polydispersity index (PDI) of 0.248 ± 0.021, a zeta potential of -30.5 ± 1.0 mV, and a concentration of (7.92 ± 0.46) × 109 bubbles/mL. Electron microscopy results show that the MLBs have a regular spherical stable core-shell structure. The superparamagnetic iron oxide nanoparticles (SPIONs) and phospholipid layers adsorbed around the spherical gas nuclei of the MLBs, leading the particles to demonstrate commendable superparamagnetic and magnetic properties. In addition, the effects of process parameters on the morphology of MLBs, including phospholipid concentration, phospholipid proportiona, current intensity, magnetothermal time, and SPION concentration, were investigated and discussed to achieve controlled preparation of MLBs. In vitro imaging results reveal that the higher the concentration of MLBs loaded with iron oxide nanoparticles, the better the in vitro ultrasound (US) imaging and magnetic resonance imaging (MRI) results. This study proves that the magneto internal heat bubble generation process is a simple and efficient technique for preparing MLBs with high concentration, regular structure, and commendable properties. These findings lay a robust foundation for the mass production and application of enveloped micro-nanobubbles, particularly in biomedical fields and other related domains.
Collapse
Affiliation(s)
- Kailin Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Mengnan Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zejin Geng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
- Joint Research Institute of Southeast University and Monash University, Suzhou, Jiangsu 215123, PR China
| | - Shuo Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zhuang Deng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Jin Tan
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Zhen Jiao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
- Joint Research Institute of Southeast University and Monash University, Suzhou, Jiangsu 215123, PR China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu 211189, PR China
- Medical School, Nanjing University, Nanjing, Jiangsu 210093, PR China
| |
Collapse
|
4
|
Xu Z, Huang J, Zhang T, Xu W, Liao X, Wang Y, Wang G. RGD peptide modified RBC membrane functionalized biomimetic nanoparticles for thrombolytic therapy. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:18. [PMID: 37043085 PMCID: PMC10097782 DOI: 10.1007/s10856-023-06719-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 03/14/2023] [Indexed: 05/03/2023]
Abstract
In recent years, the fabrication of nano-drug delivery systems for targeted treatment of thrombus has become a research hotspot. In this study, we intend to construct a biomimetic nanomedicine for targeted thrombus treatment. The poly lactic-co-glycolic acid (PLGA) was selected as the nanocarrier material. Then, urokinase and perfluoro-n-pentane (PFP) were co-loaded into PLGA by the double emulsification solvent evaporation method to prepare phase change nanoparticles PPUNPs. Subsequently, the RGD peptide-modified red blood cell membrane (RBCM) was coated on the surface of PPUNPs to prepare a biomimetic nano-drug carrier (RGD-RBCM@PPUNPs). The as-prepared RGD-RBCM@PPUNPs possessed a "core-shell" structure, have good dispersibility, and inherited the membrane protein composition of RBCs. Under ultrasound stimulation, the loaded urokinase could be rapidly released. In vitro cell experiments showed that RGD-RBCM@PPUNPs had good hemocompatibility and cytocompatibility. Due to the coated RGD-RBC membrane, RGD-RBCM@PPUNPs could effectively inhibit the uptake of macrophages. In addition, RGD-RBCM@PPUNPs showed better thrombolytic function in vitro. Overall, the results suggested that this biomimetic nanomedicine provided a promising therapeutic strategy for the targeted therapy of thrombosis.
Collapse
Affiliation(s)
- Zichen Xu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Jinxia Huang
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Tao Zhang
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Wenfeng Xu
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China.
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
5
|
Lin Y, Huang J, Chen Y, Wen Z, Cao Y, Zhang L, Cai T, Yu C, He X. Evaluation of perfluoropropane (C 3F 8)-filled chitosan polyacrylic acid nanobubbles for ultrasound imaging of sentinel lymph nodes and tumors. Biomater Sci 2022; 10:6447-6459. [PMID: 36018299 DOI: 10.1039/d2bm01140a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Accurate sentinel lymph node (SLN) identification is an important prerequisite for sentinel lymph node biopsy (SLNB). However, existing SLN mapping techniques, mainly imaging-guided methods, are severely restricted by the high cost of the instruments, harmful radiation or unsatisfactory imaging depths. Herein, we prepared a new ultrasound contrast agent by filling perfluoropropane (C3F8) into chitosan polyacrylic acid nanobubbles for precise SLN identification. The obtained ultrasound contrast agent, coined C3F8-CS-PAA nanobubbles, presents a nanometer size with a diameter of approximately 120 nm. The C3F8-CS-PAA nanobubbles of desirable size are able to enter lymphatic vessels and accumulate in the sentinel lymph node to enhance ultrasound imaging. As a result, the injection of C3F8-CS-PAA nanobubbles can remarkably enhance the ultrasound imaging lymph system, providing image guidance for sentinel lymph node biopsy. Furthermore, it was shown that such C3F8-CS-PAA nanobubbles can effectively permeate into the tumor region via the tumor-enhanced permeability and retention (EPR) effect to enhance tumor ultrasound imaging for monitoring tumorigenesis. This work highlights a novel nanoscale ultrasound contrast agent for the lymphatic system and tumor imaging, with great promise for subsequent studies and clinical applications.
Collapse
Affiliation(s)
- Yi Lin
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Ju Huang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yinyin Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering & Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400010, China
| | - Ziwei Wen
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Tao Cai
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chaoqun Yu
- College of Pharmacy, Chongqing Medical University, Chongqing 400010, China.
| | - Xuemei He
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
6
|
Gao H, Wang Z, Tan M, Liu W, Zhang L, Huang J, Cao Y, Li P, Wang Z, Wen J, Shang T, Ran H. pH-Responsive Nanoparticles for Enhanced Antitumor Activity by High-Intensity Focused Ultrasound Therapy Combined with Sonodynamic Therapy. Int J Nanomedicine 2022; 17:333-350. [PMID: 35115772 PMCID: PMC8800590 DOI: 10.2147/ijn.s336632] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/24/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Therapeutic ultrasound (US) has been extensively explored for its inherent high tissue-penetrating capability and on-demand irradiation without radioactive damage. Although high-intensity focused ultrasound (HIFU) is evolved as such an outstanding US-based approach, its insufficient therapeutic effect and the high-intensity induced potential damage to surrounding normal tissues hindered its development towards practical application. As opposed to high intensity ultrasound, sonodynamic therapy (SDT) is a low intensity US-based method which exhibits certain therapeutic effects against cancer via sonosensitizers-generated reactive oxygen species (ROS) overproduction. METHODS Hematoporphyrin monomethyl ether (HMME) loaded CaCO3 nanoparticles (designated as Ca@H) were synthesized by a gas diffusion method. The pH-responsive performance, in vitro SDT, ex vivo HIFU therapy (HIFUT), photoacoustic (PA) imaging and in vivo HIFUT combined with SDT were investigated thoroughly. RESULTS Ca@H NPs gradually decomposed in acid tumor microenvironment, produced CO2 and released HMME. Both CO2 and HMME enhanced photoacoustic (PA) imaging. The generated CO2 bubbles also enhanced HIFUT by inducing an enlarged ablation area. The tumor ablation efficiency (61.04%) was significantly improved with a combination of HIFU therapy and SDT. CONCLUSION pH-responsive Ca@H NPs have been successfully constructed for PA imaging-guided/monitored HIFUT combined with SDT. With the assistance of pH-responsive Ca@H NPs, the combination of these two US-based therapies is expected to play a role in the treatment of non-invasive tumor in the future.
Collapse
Affiliation(s)
- Hui Gao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhaoxia Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Mixiao Tan
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Weiwei Liu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ju Huang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jiexin Wen
- Department of Ultrasound, Children’s Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Tingting Shang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
7
|
Highlights in ultrasound-targeted microbubble destruction-mediated gene/drug delivery strategy for treatment of malignancies. Int J Pharm 2021; 613:121412. [PMID: 34942327 DOI: 10.1016/j.ijpharm.2021.121412] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/05/2023]
Abstract
Ultrasound is one of the safest and most advanced medical imaging technologies that is widely used in clinical practice. Ultrasound microbubbles, traditionally used for contrast-enhanced imaging, are increasingly applied in Ultrasound-targeted Microbubble Destruction (UTMD) technology which enhances tissue and cell membrane permeability through cavitation and sonoporation, to result in a promising therapeutic gene/drug delivery strategy. Here, we review recent developments in the application of UTMD-mediated gene and drug delivery in the diagnosis and treatment of tumors, including the concept, mechanism of action, clinical application status, and advantages of UTMD. Furthermore, the future perspectives that should be paid more attention to in this field are prospected.
Collapse
|
8
|
Wang Y, Cong H, Wang S, Yu B, Shen Y. Development and application of ultrasound contrast agents in biomedicine. J Mater Chem B 2021; 9:7633-7661. [PMID: 34586124 DOI: 10.1039/d1tb00850a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the rapid development of molecular imaging, ultrasound (US) medicine has evolved from traditional imaging diagnosis to integrated diagnosis and treatment at the molecular level. Ultrasound contrast agents (UCAs) play a crucial role in the integration of US diagnosis and treatment. As the micro-bubbles (MBs) in UCAs can enhance the cavitation effect and promote the biological effect of US, UCAs have also been studied in the fields of US thrombolysis, mediated gene transfer, drug delivery, and high intensity focused US. The application range of UCAs is expanding, and the value of their applications is improving. This paper reviews the development and application of UCAs in biomedicine in recent years, and the existing problems and prospects are pointed out.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China.
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China.
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
9
|
Chattaraj R, Hammer DA, Lee D, Sehgal CM. Multivariable Dependence of Acoustic Contrast of Fluorocarbon and Xenon Microbubbles under Flow. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:2676-2691. [PMID: 34112553 PMCID: PMC8355047 DOI: 10.1016/j.ultrasmedbio.2021.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/21/2021] [Accepted: 04/25/2021] [Indexed: 06/12/2023]
Abstract
Microbubbles (MBs) are 1 to 10 µm gas particles stabilized by an amphiphilic shell capable of responding to biomedical ultrasound with strong acoustic signals, allowing them to be commonly used in ultrasound imaging and therapy. The composition of both the shell and the core determines their stability and acoustic properties. While there has been extensive characterization of the dissolution, oscillation, cavitation, collapse and therefore, ultrasound contrast of MBs under static conditions, few reports have examined such behavior under hydrodynamic flow. In this study, we evaluate the interplay of ultrasound parameters (five different mechanical indices [MIs]), MB shell parameter (shell stiffness), type of gas (perfluorocarbon for diagnostic imaging and xenon as a therapeutic gas), and a flow parameter (flow rate) on the ultrasound signal of phospholipid-stabilized MBs flowing through a latex tube embedded in a tissue-mimicking phantom. We find that the contrast gradient (CG), a metric of the rate of decay of contrast along the length of the tube, and the contrast peak (CP), the location where the maximum contrast is reached, depend on the conditions of flow, imaging, and MB material. For instance, while the contrast near the flow inlet of the field of view is highest for a softer shell (dipalmitoylphosphatidylcholine [DPPC], C16) than for stiffer shells (distearoylphosphatidylcholine [DSPC], C18, and dibehenoylphosphatidylcholine [DBPC], C22), the contrast decay is also faster; stiffer shells provide more resistance and hence lead to slower MB dissolution/destruction. At higher flow rates, the CG is low for a fixed length of time because each MB is exposed to ultrasound for a shorter period. The CG becomes high for low flow rates, especially at high incident pressures (high MI), causing more MB destruction closer to the inlet of the field of view. Also, the CP shifts toward the inlet at low flow rates, high MIs, and low shell stiffness. We also report the first demonstration of sustained ultrasound flow imaging of a water-soluble, therapeutic gas MB (xenon). We find that an increased MB concentration is necessary for obtaining the same signal magnitude for xenon MBs. In summary, this study builds a framework depicting how multiple variables simultaneously affect the evolution of MB ultrasound contrast under flow. Depending on the MB composition, imaging conditions, transducer positioning, and image processing, building on such a framework could potentially allow for extraction of additional diagnostic information than is commonly analyzed for physiological flow.
Collapse
Affiliation(s)
- Rajarshi Chattaraj
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel A Hammer
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daeyeon Lee
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chandra M Sehgal
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
10
|
Lin X, Chen W, Wei F. Technique Success, Technique Efficacy and Complications of HIFU Ablation for Palliation of Pain in Patients With Bone Lesions: A Meta-Analysis of 28 Feasibility Studies. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1182-1191. [PMID: 33583637 DOI: 10.1016/j.ultrasmedbio.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 06/12/2023]
Abstract
Several feasibility studies have reported that high-intensity focused ultrasound (HIFU) ablation can be applied to ease patients' bone pain. However, the effect of HIFU ablation to palliate bone pain remains unclear. To evaluate the technique's success, efficacy, minor complication and major complication on patients suffering from bone pain, we searched the PubMed, MEDLINE, EMBASE and Cochrane Library databases from January 1998 to March 2019. Clinical studies that have assessed the association between bone pain and HIFU ablation were involved. We filtered out 28 feasibility studies, which reported the association between HIFU ablation and bone pain, including a total of 717 patients and 736 bone lesions. Overall, our results indicate that the rate of technique success of HIFU ablation was 93% (95% confidence interval [CI] 89%-96%) for patients with bone lesions. The technique efficacy rate of HIFU ablation for palliation of pain from bone lesions was 80% (95% CI 74%-87%) in all the patients, 96% (91%-100%) in the subgroup of retrospective studies and 77% (69%-85%) in the subgroup of prospective studies. In regard to HIFU ablation treatment safety, the hazard ratio for minor complication was 12% (95% CI 7%-17%), and the hazard ratio for major complication was 2% (95% CI 1%-3%). In conclusion, the summary rates for various considerations of using HIFU ablation for the palliation of bone pain are as follows: technique success is 93%, technique efficacy is 77%, minor complication is 12% and major complication is 2%. Our results suggest that extracorporeal HIFU ablation is a promising method for palliation of pain in bone lesions, with high technique success and efficacy, but low adverse events.
Collapse
Affiliation(s)
- Xiaoti Lin
- Department of Breast, Fujian Provincial Maternity and Children's Hospital, Fujian Medical University, Fuzhou, China; Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Weiyu Chen
- Department of Physiology, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, China
| | - Fengqin Wei
- Department of Emergency, Fujian Provincial 2nd People's Hospital, Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
11
|
Wu H, Tong L, Wang Y, Yan H, Sun Z. Bibliometric Analysis of Global Research Trends on Ultrasound Microbubble: A Quickly Developing Field. Front Pharmacol 2021; 12:646626. [PMID: 33967783 PMCID: PMC8101552 DOI: 10.3389/fphar.2021.646626] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Microbubbles are widely used as highly effective contrast agents to improve the diagnostic capability of ultrasound imaging. Mounting evidence suggests that ultrasound coupled with microbubbles has promising therapeutic applications in cancer, cardiovascular, and neurological disorders by acting as gene or drug carriers. The aim of this study was to identify the scientific output and activity related to ultrasound microbubble through bibliometric approaches. Methods: The literature related to ultrasound microbubble published between 1998 and 2019 was identified and selected from the Science Citation Index Expanded of Web of Science Core Collection on February 21, 2021. The Scopus database was also searched to validate the results and provided as supplementary material. Quantitative variables including number of publications and citations, H-index, and journal citation reports were analyzed by using Microsoft Excel 2019 and GraphPad Prism 8.0 software. VOS viewer and CiteSpace V were used to perform coauthorship, citation, co-citation, and co-occurrence analysis for countries/regions, institutions, authors, and keywords. Results: A total of 6088 publications from the WoSCC were included. The United States has made the largest contribution in this field, with the majority of publications (2090, 34.3%), citations (90,741, 46.6%), the highest H-index (138), and close collaborations with China and Canada. The most contributive institution was the University of Toronto. Professors De Jong N and Dayton P A have made great achievements in this field. However, the research cooperation between institutions and authors was relatively weak. All the studies could be divided into four clusters: "ultrasound diagnosis study," "microbubbles' characteristics study," "gene therapy study," and "drug delivery study." The average appearing years (AAY) of keywords in the cluster "drug delivery study" was more recent than other clusters. For promising hot spots, "doxorubicin" showed a relatively latest AAY of 2015.49, followed by "nanoparticles" and "breast cancer." Conclusion: There has been an increasing amount of scientific output on ultrasound microbubble according to the global trends, and the United States is staying ahead in this field. Collaboration between research teams still needs to be strengthened. The focus gradually shifts from "ultrasound diagnosis study" to "drug delivery study." It is recommended to pay attention to the latest hot spots, such as "doxorubicin," "nanoparticles," and "breast cancer."
Collapse
Affiliation(s)
- Haiyang Wu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Linjian Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Yulin Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhiming Sun
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China.,Department of Orthopaedic Surgery, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
12
|
Oroojalian F, Beygi M, Baradaran B, Mokhtarzadeh A, Shahbazi MA. Immune Cell Membrane-Coated Biomimetic Nanoparticles for Targeted Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006484. [PMID: 33577127 DOI: 10.1002/smll.202006484] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Nanotechnology has provided great opportunities for managing neoplastic conditions at various levels, from preventive and diagnostic to therapeutic fields. However, when it comes to clinical application, nanoparticles (NPs) have some limitations in terms of biological stability, poor targeting, and rapid clearance from the body. Therefore, biomimetic approaches, utilizing immune cell membranes, are proposed to solve these issues. For example, macrophage or neutrophil cell membrane coated NPs are developed with the ability to interact with tumor tissue to suppress cancer progression and metastasis. The functionality of these particles largely depends on the surface proteins of the immune cells and their preserved function during membrane extraction and coating process on the NPs. Proteins on the outer surface of immune cells can render a wide range of activities to the NPs, including prolonged blood circulation, remarkable competency in recognizing antigens for enhanced targeting, better cellular interactions, gradual drug release, and reduced toxicity in vivo. In this review, nano-based systems coated with immune cells-derived membranous layers, their detailed production process, and the applicability of these biomimetic systems in cancer treatment are discussed. In addition, future perspectives and challenges for their clinical translation are also presented.
Collapse
Affiliation(s)
- Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
| | - Mohammad Beygi
- Department of Agricultural Engineering, Isfahan University of Technology (IUT), Isfahan, 84156-83111, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 51666-14731, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 51666-14731, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, 45139-56184, Iran
| |
Collapse
|
13
|
Li Y, Lu J, Zhang J, Zhu X, Liu J, Zhang Y. Phase-Change Nanotherapeutic Agents Based on Mesoporous Carbon for Multimodal Imaging and Tumor Therapy. ACS APPLIED BIO MATERIALS 2020; 3:8705-8713. [DOI: 10.1021/acsabm.0c01102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Yong Li
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
- School of Life Sciences, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| | - Jialin Lu
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| | - Jing Zhang
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| | - Xiaohui Zhu
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| | - Jinliang Liu
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| | - Yong Zhang
- School of Environmental and Chemical Engineering, Shanghai University, 99 Shangda Road, 200444 Shanghai, China
| |
Collapse
|
14
|
A review of ultrasound-mediated microbubbles technology for cancer therapy: a vehicle for chemotherapeutic drug delivery. JOURNAL OF RADIOTHERAPY IN PRACTICE 2020. [DOI: 10.1017/s1460396919000633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AbstractBackground:The unique behaviour of microbubbles under ultrasound acoustic pressure makes them useful agents for drug and gene delivery. Several studies have demonstrated the potential application of microbubbles as a non-invasive, safe and effective technique for targeted delivery of drugs and genes. The drugs can be incorporated into the microbubbles in several different approaches and then carried to the site of interest where it can be released by destruction of the microbubbles using ultrasound to achieve the required therapeutic effect.Methods:The objective of this article is to report on a review of the recent advances of ultrasound-mediated microbubbles as a vehicle for delivering drugs and genes and its potential application for the treatment of cancer.Conclusion:Ultrasound-mediated microbubble technology has the potential to significantly improve chemotherapy drug delivery to treatment sites with minimal side effects. Moreover, the technology can induce temporary and reversible changes in the permeability of cells and vessels, thereby allowing for drug delivery in a spatially localised region which can improve the efficiency of drugs with poor bioavailability due to their poor absorption, rapid metabolism and rapid systemic elimination.
Collapse
|
15
|
He H, Du L, Guo H, An Y, Lu L, Chen Y, Wang Y, Zhong H, Shen J, Wu J, Shuai X. Redox Responsive Metal Organic Framework Nanoparticles Induces Ferroptosis for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2001251. [PMID: 32677157 DOI: 10.1002/smll.202001251] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/05/2020] [Indexed: 06/11/2023]
Abstract
Ferroptosis is attracting significant attention due to its effectiveness in tumor treatment. The efficiency to produce toxic lipid peroxides (LPOs) at the tumor site plays a key role in ferroptosis. A hybrid PFP@Fe/Cu-SS metal organic framework (MOF) is synthesized and shown to increase intratumoral LPO content through redox reactions generating ·OH. In addition, glutathione (GSH) depletion through disulfide-thiol exchange leads to the inactivation of glutathione peroxide 4 (GPX4), which results in a further increase in LPO content. This MOF exhibits high inhibitory effect on the growth of xenografted Huh-7 tumors in mice. The coadministration of a ferroptosis inhibitor reduces the antitumor effect of the MOF, leading to a restoration of GPX4 activity and an increase in tumor growth. Moreover, the construction of Cu into mesoporous PFP@Fe/Cu-SS not only allows the MOF to be used as a contrast agent for T1 -weighted magnetic resonance imaging, but also renders its photothermal conversion capacity. Thus, near-infrared irradiation is able to induce photothermal therapy and transform the encapsulated liquid perfluoropentane into microbubbles for ultrasound imaging.
Collapse
Affiliation(s)
- Haozhe He
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Lihua Du
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Huanling Guo
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yongcheng An
- University of Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510275, China
| | - Liejing Lu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yali Chen
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yong Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Huihai Zhong
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xintao Shuai
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
16
|
Pellow C, O'Reilly MA, Hynynen K, Zheng G, Goertz DE. Simultaneous Intravital Optical and Acoustic Monitoring of Ultrasound-Triggered Nanobubble Generation and Extravasation. NANO LETTERS 2020; 20:4512-4519. [PMID: 32374617 DOI: 10.1021/acs.nanolett.0c01310] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ultrasound-activated nanobubbles are being widely investigated as contrast agents and therapeutic vehicles. Nanobubbles hold potential for accessing the tumor extravascular compartment, though this relies on clinically debated passive accumulation for which evidence to date is indirect. We recently reported ultrasound-triggered conversion of high payload porphyrin-encapsulated microbubbles to nanobubbles, with actively enhanced permeability for local delivery. This platform holds implications for optical/ultrasound-based imaging and therapeutics. While promising, it remains to be established how nanobubbles are generated and whether they extravasate intact. Here, insights into the conversion process are reported, complemented by novel simultaneous intravital and acoustic monitoring in tumor-affected functional circulation. The first direct acoustic evidence of extravascular intact nanobubbles are presented. These insights collectively advance this delivery platform and multimodal micro- and nanobubbles, extending their utility for imaging and therapeutics within and beyond the vasculature.
Collapse
Affiliation(s)
- Carly Pellow
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 2C1, Canada
| | - Meaghan A O'Reilly
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| | - Kullervo Hynynen
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| | - Gang Zheng
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 2C1, Canada
| | - David E Goertz
- University of Toronto, Department of Medical Biophysics, Toronto, M5G 1L7, Canada
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, M4N 3M5, Canada
| |
Collapse
|
17
|
Li J, Xi A, Qiao H, Liu Z. Ultrasound-mediated diagnostic imaging and advanced treatment with multifunctional micro/nanobubbles. Cancer Lett 2020; 475:92-98. [DOI: 10.1016/j.canlet.2020.01.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/10/2020] [Accepted: 01/28/2020] [Indexed: 12/20/2022]
|
18
|
Li J, Ji H, Jing Y, Wang S. pH- and acoustic-responsive platforms based on perfluoropentane-loaded protein nanoparticles for ovarian tumor-targeted ultrasound imaging and therapy. NANOSCALE RESEARCH LETTERS 2020; 15:31. [PMID: 32016619 PMCID: PMC6997325 DOI: 10.1186/s11671-020-3252-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/14/2020] [Indexed: 05/02/2023]
Abstract
In this study, we developed a multifunctional ultrasound (US) therapeutic agent that encapsulates perfluoropentane (PFP) into ferritin (FRT) and conjugates the tumor-targeting molecule folic acid (FA) (FA-FRT-PFP). The prepared FA-FRT-PFP had an average particle diameter of 42.8 ± 2.5 nm, a zeta potential of - 41.1 ± 1.7 mV and shows good stability in physiological solution and temperatures. FRT is a pH-sensitive cage protein that, at pH 5.0, disassembles to form pores that can load PFP. The adjustment to neutral pH closes the pores and encapsulates the PFP inside the FRT to form nanoparticles. At pH 5.0, 3 min of low-intensity focused ultrasound (LIFU, 2 W/cm2) significantly enhanced the US signal of FA-FRT-PFP through the acoustic droplet vaporization (ADV) effect. Under identical conditions, 4 min of LIFU irradiation caused the bubbles generated by FA-FRT-PFP to break. FA-FRT-PFP could be efficiently targeted into ovarian cancer cells and significantly enhanced the US contrast of FA-FRT-PFP after 3 min of LIFU irradiation. After 4 min of LIFU irradiation, cell viability significantly decreased due to necrosis, likely due to the FA-FRT-PFP mediated release of PFP in the acidic environment of lysosomes after entering the tumor cells. PFP is then transformed into bubbles that burst under LIFU irradiation, forming physical shock waves that lead to the destruction of the cell structure and necrosis, achieving tumor treatment. Taken together, this demonstrates that FA-FRT-PFP is both a novel and promising US theranostics agent for future clinic application.
Collapse
Affiliation(s)
- Jianping Li
- Department of Geriatric Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, 610041 Sichuan China
| | - Hong Ji
- Department of Geriatric Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, 610041 Sichuan China
| | - Yong Jing
- Department of Imaging, Eastern Hospital of Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, 610000 Sichuan China
| | - Shiguang Wang
- Department of Imaging, Eastern Hospital of Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, 610000 Sichuan China
| |
Collapse
|
19
|
Hameed S, Zhang M, Bhattarai P, Mustafa G, Dai Z. Enhancing cancer therapeutic efficacy through ultrasound‐mediated micro‐to‐nano conversion. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1604. [DOI: 10.1002/wnan.1604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Sadaf Hameed
- Department of Biomedical Engineering, College of Engineering Peking University Beijing China
| | - Miaomiao Zhang
- Department of Biomedical Engineering, College of Engineering Peking University Beijing China
| | - Pravin Bhattarai
- Department of Biomedical Engineering, College of Engineering Peking University Beijing China
- Phutung Research Institute Kathmandu Nepal
| | - Ghulam Mustafa
- Department of Sciences Bahria University Lahore Lahore Pakistan
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Engineering Peking University Beijing China
| |
Collapse
|
20
|
Xie Y, Wang J, Wang J, Hu Z, Hariri A, Tu N, Krug KA, Burkart MD, Gianneschi NC, Jokerst JV, Rinehart JD. Tuning the ultrasonic and photoacoustic response of polydopamine-stabilized perfluorocarbon contrast agents. J Mater Chem B 2019; 7:4833-4842. [PMID: 31389967 PMCID: PMC6690494 DOI: 10.1039/c9tb00928k] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Contrast-enhanced ultrasound (CEUS) offers the exciting prospect of retaining the ease of ultrasound imaging while enhancing imaging clarity, diagnostic specificity, and theranostic capability. To advance the capabilities of CEUS, the synthesis and understanding of new ultrasound contrast agents (UCAs) is a necessity. Many UCAs are nano- or micro-scale materials composed of a perfluorocarbon (PFC) and stabilizer that synergistically induce an ultrasound response that is both information-rich and easily differentiated from natural tissue. In this work, we probe the extent to which CEUS is modulated through variation in a PFC stabilized with fluorine-modified polydopamine nanoparticles (PDA NPs). The high level of synthetic tunability in this system allows us to study signal as a function of particle aggregation and PFC volatility in a systematic manner. Separation of aggregated and non-aggregated nanoparticles lead to a fundamentally different signal response, and for this system, PFC volatility has little effect on CEUS intensity despite a range of over 50 °C in boiling point. To further explore the imaging tunability and multimodality, Fe3+-chelation was employed to generate an enhanced photoacoustic (PA) signal in addition to the US signal. In vitro and in vivo results demonstrate that PFC-loaded PDA NPs show stronger PA signal than the non-PFC ones, indicating that the PA signal can be used for in situ differentiation between PFC-loading levels. In sum, these data evince the rich role synthetic chemistry can play in guiding new directions of development for UCAs.
Collapse
Affiliation(s)
- Yijun Xie
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yildirim A, Blum NT, Goodwin AP. Colloids, nanoparticles, and materials for imaging, delivery, ablation, and theranostics by focused ultrasound (FUS). Theranostics 2019; 9:2572-2594. [PMID: 31131054 PMCID: PMC6525987 DOI: 10.7150/thno.32424] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 12/15/2022] Open
Abstract
This review focuses on different materials and contrast agents that sensitize imaging and therapy with Focused Ultrasound (FUS). At high intensities, FUS is capable of selectively ablating tissue with focus on the millimeter scale, presenting an alternative to surgical intervention or management of malignant growth. At low intensities, FUS can be also used for other medical applications such as local delivery of drugs and blood brain barrier opening (BBBO). Contrast agents offer an opportunity to increase selective acoustic absorption or facilitate destructive cavitation processes by converting incident acoustic energy into thermal and mechanical energy. First, we review the history of FUS and its effects on living tissue. Next, we present different colloidal or nanoparticulate approaches to sensitizing FUS, for example using microbubbles, phase-shift emulsions, hollow-shelled nanoparticles, or hydrophobic silica surfaces. Exploring the science behind these interactions, we also discuss ways to make stimulus-responsive, or "turn-on" contrast agents for improved selectivity. Finally, we discuss acoustically-active hydrogels and membranes. This review will be of interest to those working in materials who wish to explore new applications in acoustics and those in acoustics who are seeking new agents to improve the efficacy of their approaches.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
- Present address: CEDAR, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239 USA
| | - Nicholas T. Blum
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
| |
Collapse
|
22
|
Hernandez C, Abenojar EC, Hadley J, de Leon AC, Coyne R, Perera R, Gopalakrishnan R, Basilion JP, Kolios MC, Exner AA. Sink or float? Characterization of shell-stabilized bulk nanobubbles using a resonant mass measurement technique. NANOSCALE 2019; 11:851-855. [PMID: 30601524 PMCID: PMC6350620 DOI: 10.1039/c8nr08763f] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/25/2018] [Indexed: 05/14/2023]
Abstract
Nano-sized shell-stabilized gas bubbles have applications in various fields ranging from environmental science to biomedical engineering. A resonant mass measurement (RMM) technique is demonstrated here as a new and only method capable of simultaneously measuring the size and concentration of buoyant and non-buoyant particles in a nanobubble sample used as a next-generation ultrasound contrast agent.
Collapse
Affiliation(s)
- Christopher Hernandez
- Department of Radiology
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
.
| | - Eric C. Abenojar
- Department of Radiology
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
.
| | | | - Al Christopher de Leon
- Department of Radiology
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
.
| | - Robert Coyne
- Malvern Panalytical
,
Westborough
, MA
, 01581 USA
| | - Reshani Perera
- Department of Radiology
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
.
| | | | - James P. Basilion
- Department of Radiology
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
.
- Department of Biomedical Engineering
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
| | - Michael C. Kolios
- Department of Physics
, Ryerson University
,
Toronto
, Ontario
, Canada M5B 2K3
| | - Agata A. Exner
- Department of Radiology
, Case Western Reserve University
,
Cleveland
, OH
, 44106 USA
.
| |
Collapse
|
23
|
Yildirim A, Shi D, Roy S, Blum NT, Chattaraj R, Cha JN, Goodwin AP. Nanoparticle-Mediated Acoustic Cavitation Enables High Intensity Focused Ultrasound Ablation Without Tissue Heating. ACS APPLIED MATERIALS & INTERFACES 2018; 10:36786-36795. [PMID: 30339360 PMCID: PMC6702128 DOI: 10.1021/acsami.8b15368] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
While thermal ablation of various solid tumors has been demonstrated using high intensity focused ultrasound (HIFU), the therapeutic outcomes of this technique are still unsatisfactory because of common recurrence of thermally ablated cancers and treatment side effects due to the high ultrasound intensity and acoustic pressure requirements. More precise ablation of tumors can be achieved by generating cavitating bubbles in the tissue using shorter pulses with higher acoustic pressures, which induce mechanical damage rather than thermal. However, it has remained as a challenge to safely deliver the acoustic pressures required for mechanical ablation of solid tumors. Here, we report a method to achieve mechanical ablation at lower acoustic pressures by utilizing phospholipid-stabilized hydrophobic mesoporous silica nanoparticles (PL-hMSN). The PL-hMSNs act as seeds for nucleation of cavitation events and thus significantly reduce the peak negative pressures and spatial-average temporal-average HIFU intensities needed to achieve mechanical ablation. Substantial mechanical damage was observed in the red blood cell or tumor spheroid containing tissue mimicking phantoms at PL-hMSN concentrations as low as 10 μg mL-1, after only 5 s of HIFU treatment with peak negative pressures ∼11 MPa and duty cycles ∼0.01%. Even the application of HIFU (peak negative pressure of 16.8 MPa and duty cycle of 0.017%) for 1 min in the presence of PL-hMSN (200 μg mL-1) did not cause any detectable temperature increase in tissue-mimicking phantoms. In addition, the mechanical effects of cavitation promoted by PL-hMSNs were observed up to 0.5 mm from the center of the cavitation events. This method may thus also improve delivery of therapeutics or nanoparticles to tumor environments with limited macromolecular transport.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
- (Adem Yildirim):
| | - Dennis Shi
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| | - Shambojit Roy
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| | - Nicholas T. Blum
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| | - Rajarshi Chattaraj
- Department of Mechanical Engineering, University of Colorado Boulder. Boulder, Colorado 80309, United States
| | - Jennifer N. Cha
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
- Corresponding Author (Andrew P. Goodwin):
| |
Collapse
|
24
|
McLaughlan JR. Controllable Nucleation of Cavitation from Plasmonic Gold Nanoparticles for Enhancing High Intensity Focused Ultrasound Applications. J Vis Exp 2018. [PMID: 30346394 PMCID: PMC6235418 DOI: 10.3791/58045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In this study, plasmonic gold nanoparticles were simultaneously exposed to pulsed near-infrared laser light and high intensity focused ultrasound (HIFU) for the controllable nucleation of cavitation in tissue-mimicking gel phantoms. This in vitro protocol was developed to demonstrate the feasibility of this approach, for both enhancement of imaging and therapeutic applications for cancer. The same apparatus can be used for both imaging and therapeutic applications by varying the exposure duration of the HIFU system. For short duration exposures (10 µs), broadband acoustic emissions were generated through the controlled nucleation of inertial cavitation around the gold nanoparticles. These emissions provide direct localization of nanoparticles. For future applications, these particles may be functionalized with molecular-targeting antibodies (e.g. anti-HER2 for breast cancer) and can provide precise localization of cancerous regions, complementing routine diagnostic ultrasound imaging. For continuous wave (CW) exposures, the cavitation activity was used to increase the localized heating from the HIFU exposures resulting in larger thermal damage in the gel phantoms. The acoustic emissions generated from inertial cavitation activity during these CW exposures was monitored using a passive cavitation detection (PCD) system to provide feedback of cavitation activity. Increased localized heating was only achieved through the unique combination of nanoparticles, laser light and HIFU. Further validation of this technique in pre-clinical models of cancer is necessary.
Collapse
Affiliation(s)
- James R McLaughlan
- School of Electronic and Electrical Engineering, University of Leeds; Leeds Institute of Cancer and Pathology, University of Leeds;
| |
Collapse
|
25
|
Microbubble-Mediated Ultrasound Outweighs Low-Intensity Pulsed Ultrasound on Osteogenesis and Neovascularization in a Rabbit Model of Steroid-Associated Osteonecrosis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4606791. [PMID: 30298135 PMCID: PMC6157205 DOI: 10.1155/2018/4606791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/04/2018] [Accepted: 07/30/2018] [Indexed: 12/27/2022]
Abstract
Microbubbles magnify the acoustic pressure of low-intensity pulsed ultrasound (LIPUS) and may enhance its bioeffect for diagnostic and therapeutic purposes. This study compared the effect of this novel microbubble-mediated ultrasound (MUS) with that of the traditional LIPUS on osteogenesis and neovascularization in a rabbit model of steroid-associated osteonecrosis. We hypothesized that MUS might outweigh LIPUS on promoting osteogenesis and neovascularization in steroid-associated osteonecrosis. The bilateral femoral head necrosis was induced by lipopolysaccharide and methylprednisolone in the rabbits. The indices of bone mineral density (BMD), trabecular number, maximal loading strength, and mineral apposition rate were analyzed, demonstrating that the animal model of steroid-associated osteonecrosis was successfully established. Both the MUS group (GM) and the LIPUS group (GL) were insonated 20 min daily for six weeks. GM received an extra intracapsular injection of microbubbles before insonation every other day. Fluorescence bone labeling, Micro-CT Analysis, biomechanical test, quantitative real-time PCR, Western blot analysis, and histological evaluation were performed for comparing GM with GL. The results demonstrated a 39% higher mineral apposition rate in GM compared with GL. The BMD and the maximal loading strength of femoral head of GM increased by 4.3% and 27.8% compared to those of GL, respectively. The mRNA and protein expression of BMP-2 and VEGF were also significantly higher in GM. The number of blood vessels of GM was 65% greater than that of GL. MUS is more potent than LIPUS in enhancing osteogenesis, neovascularization, and biomechanical strength of femoral head in the animal model of steroid-associated osteonecrosis. Without increasing the intensity of insonation or the risk of tissue damage, MUS is better for inhibiting the process of steroid-associated osteonecrosis.
Collapse
|
26
|
Tu J, Zhang H, Yu J, Liufu C, Chen Z. Ultrasound-mediated microbubble destruction: a new method in cancer immunotherapy. Onco Targets Ther 2018; 11:5763-5775. [PMID: 30254469 PMCID: PMC6140758 DOI: 10.2147/ott.s171019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immunotherapy provides a new treatment option for cancer. However, it may be therapeutically insufficient if only using the self-immune system alone to attack the tumor without any aiding methods. To overcome this drawback and improve the efficiency of therapy, new treatment methods are emerging. In recent years, ultrasound-mediated microbubble destruction (UMMD) has shown great potential in cancer immunotherapy. Using the combination of ultrasound and targeted microbubbles, molecules such as antigens or genes encoding antigens can be efficiently and specifically delivered into the tumor tissue. This review focuses on the recent progress in the application of UMMD in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiawei Tu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Hui Zhang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Chun Liufu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| |
Collapse
|
27
|
Canavese G, Ancona A, Racca L, Canta M, Dumontel B, Barbaresco F, Limongi T, Cauda V. Nanoparticle-assisted ultrasound: A special focus on sonodynamic therapy against cancer. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2018; 340:155-172. [PMID: 30881202 PMCID: PMC6420022 DOI: 10.1016/j.cej.2018.01.060] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
At present, ultrasound radiation is broadly employed in medicine for both diagnostic and therapeutic purposes at various frequencies and intensities. In this review article, we focus on therapeutically-active nanoparticles (NPs) when stimulated by ultrasound. We first introduce the different ultrasound-based therapies with special attention to the techniques involved in the oncological field, then we summarize the different NPs used, ranging from soft materials, like liposomes or micro/nano-bubbles, to metal and metal oxide NPs. We therefore focus on the sonodynamic therapy and on the possible working mechanisms under debate of NPs-assisted sonodynamic treatments. We support the idea that various, complex and synergistics physical-chemical processes take place during acoustic cavitation and NP activation. Different mechanisms are therefore responsible for the final cancer cell death and strongly depends not only on the type and structure of NPs or nanocarriers, but also on the way they interact with the ultrasonic pressure waves. We conclude with a brief overview of the clinical applications of the various ultrasound therapies and the related use of NPs-assisted ultrasound in clinics, showing that this very innovative and promising approach is however still at its infancy in the clinical cancer treatment.
Collapse
Affiliation(s)
- Giancarlo Canavese
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
- Center for Sustainable Future Technologies CSFT@Polito, Istituto Italiano di Tecnologia, Corso Trento 21, 10129, Turin, Italy
| | - Andrea Ancona
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Luisa Racca
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Marta Canta
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Bianca Dumontel
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Federica Barbaresco
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Tania Limongi
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
- Center for Sustainable Future Technologies CSFT@Polito, Istituto Italiano di Tecnologia, Corso Trento 21, 10129, Turin, Italy
| |
Collapse
|
28
|
Warzecha KT, Bartneck M, Möckel D, Appold L, Ergen C, Al Rawashdeh W, Gremse F, Niemietz PM, Jahnen-Dechent W, Trautwein C, Kiessling F, Lammers T, Tacke F. Targeting and Modulation of Liver Myeloid Immune Cells by Hard-Shell Microbubbles. ACTA ACUST UNITED AC 2018; 2. [PMID: 29876517 DOI: 10.1002/adbi.201800002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Poly n-butylcyanoacrylate (PBCA)-based hard-shell microbubbles (MB) have manifold biomedical applications, including targeted drug delivery or contrast agents for ultrasound (US)-based liver imaging. MB and their fragments accumulate in phagocytes, especially in the liver, but it is unclear if MB affect the function of these immune cells. We herein show that human primary monocytes internalize different PBCA-MB by phagocytosis, which transiently inhibits monocyte migration in vertical chemotaxis assays and renders monocytes susceptible to cytotoxic effects of MB during US-guided destruction. Conversely, human macrophage viability and function, including cytokine release and polarization, remain unaffected after MB uptake. After i.v. injection in mice, MB predominantly accumulate in liver, especially in hepatic phagocytes (monocytes and Kupffer cells). Despite efficiently targeting myeloid immune cells in liver, MB or MB after US-elicited burst do not cause overt hepatotoxicity or inflammation. Furthermore, MB application with or without US-guided burst does not aggravate the course of experimental liver injury in mice or the inflammatory response to liver injury in vivo. In conclusion, PBCA-MB have immunomodulatory effects on primary human myeloid cells in vitro, but do not provoke hepatotoxicity, inflammation or altered response to liver injury in vivo, suggesting the safety of these MB for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Klaudia T Warzecha
- Department of Medicine III, Medical Faculty, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Matthias Bartneck
- Department of Medicine III, Medical Faculty, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Diana Möckel
- Department of Experimental Molecular Imaging, University Hospital and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Lia Appold
- Department of Experimental Molecular Imaging, University Hospital and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Can Ergen
- Department of Medicine III, Medical Faculty, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Wáel Al Rawashdeh
- Department of Experimental Molecular Imaging, University Hospital and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Felix Gremse
- Department of Experimental Molecular Imaging, University Hospital and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Patricia M Niemietz
- Department of Medicine III, Medical Faculty, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Willi Jahnen-Dechent
- Helmholtz-Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, Medical Faculty, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Fabian Kiessling
- Department of Experimental Molecular Imaging, University Hospital and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Department of Experimental Molecular Imaging, University Hospital and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany; Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Frank Tacke
- Department of Medicine III, Medical Faculty, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| |
Collapse
|
29
|
Li C, Zhang Y, Li Z, Mei E, Lin J, Li F, Chen C, Qing X, Hou L, Xiong L, Hao H, Yang Y, Huang P. Light-Responsive Biodegradable Nanorattles for Cancer Theranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1706150. [PMID: 29271515 DOI: 10.1002/adma.201706150] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/15/2017] [Indexed: 05/25/2023]
Abstract
Cancer nanotheranostics, integrating both diagnostic and therapeutic functions into nanoscale agents, are advanced solutions for cancer management. Herein, a light-responsive biodegradable nanorattle-based perfluoropentane-(PFP)-filled mesoporous-silica-film-coated gold nanorod (GNR@SiO2 -PFP) is strategically designed and prepared for enhanced ultrasound (US)/photoacoustic (PA) dual-modality imaging guided photothermal therapy of melanoma. The as-prepared nanorattles are composed of a thin mesoporous silica film as the shell, which endows the nanoplatform with flexible morphology and excellent biodegradability, as well as large cavity for PFP filling. Upon 808 nm laser irradiation, the loaded PFP will undergo a liquid-gas phase transition due to the heat generation from GNRs, thus generating nanobubbles followed by the coalescence into microbubbles. The conversion of nanobubbles to microbubbles can improve the intratumoral permeation and retention in nonmicrovascular tissue, as well as enhance the tumor-targeted US imaging signals. This nanotheranostic platform exhibits excellent biocompatibility and biodegradability, distinct gas bubbling phenomenon, good US/PA imaging contrast, and remarkable photothermal efficiency. The results demonstrate that the GNR@SiO2 -PFP nanorattles hold great potential for cancer nanotheranostics.
Collapse
Affiliation(s)
- Chunxiao Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yifan Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zhiming Li
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
| | - Enci Mei
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
| | - Jing Lin
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Fan Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Cunguo Chen
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
| | - Xialing Qing
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Liyue Hou
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
| | - Lingling Xiong
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, P. R. China
| | - Hui Hao
- Nanomaterials and Chemistry Key Laboratory, Wenzhou University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Yun Yang
- Nanomaterials and Chemistry Key Laboratory, Wenzhou University, Wenzhou, Zhejiang, 325027, P. R. China
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, P. R. China
| |
Collapse
|
30
|
Zullino S, Argenziano M, Stura I, Guiot C, Cavalli R. From Micro- to Nano-Multifunctional Theranostic Platform: Effective Ultrasound Imaging Is Not Just a Matter of Scale. Mol Imaging 2018; 17:1536012118778216. [PMID: 30213222 PMCID: PMC6144578 DOI: 10.1177/1536012118778216] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/20/2018] [Accepted: 04/08/2018] [Indexed: 12/20/2022] Open
Abstract
Ultrasound Contrast Agents (UCAs) consisting of gas-filled-coated Microbubbles (MBs) with diameters between 1 and 10 µm have been used for a number of decades in diagnostic imaging. In recent years, submicron contrast agents have proven to be a viable alternative to MBs for ultrasound (US)-based applications for their capability to extravasate and accumulate in the tumor tissue via the enhanced permeability and retention effect. After a short overview of the more recent approaches to ultrasound-mediated imaging and therapeutics at the nanoscale, phase-change contrast agents (PCCAs), which can be phase-transitioned into highly echogenic MBs by means of US, are here presented. The phenomenon of acoustic droplet vaporization (ADV) to produce bubbles is widely investigated for both imaging and therapeutic applications to develop promising theranostic platforms.
Collapse
Affiliation(s)
- Sara Zullino
- Department of Neuroscience, University of Turin, Turin, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Ilaria Stura
- Department of Clinical and Biological Science, University of Turin, Turin, Italy
| | - Caterina Guiot
- Department of Neuroscience, University of Turin, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| |
Collapse
|
31
|
McLaughlan JR, Cowell DMJ, Freear S. Gold nanoparticle nucleated cavitation for enhanced high intensity focused ultrasound therapy. Phys Med Biol 2017; 63:015004. [PMID: 29098986 DOI: 10.1088/1361-6560/aa97e9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High intensity focused ultrasound (HIFU) or focused ultrasound surgery is a non-invasive technique for the treatment of cancerous tissue, which is limited by difficulties in getting real-time feedback on treatment progress and long treatment durations. The formation and activity of acoustic cavitation, specifically inertial cavitation, during HIFU exposures has been demonstrated to enhance heating rates. However, without the introduction of external nuclei its formation an activity can be unpredictable, and potentially counter-productive. In this study, a combination of pulse laser illumination (839 nm), HIFU exposures (3.3 MHz) and plasmonic gold nanorods (AuNR) was demonstrated as a new approach for the guidance and enhancement of HIFU treatments. For imaging, short duration HIFU pulses (10 μs) demonstrated broadband acoustic emissions from AuNR nucleated cavitation with a signal-to-noise ranging from 5-35 dB for peak negative pressures between 1.19-3.19 ± 0.01 MPa. In the absence of either AuNR or laser illumination these emissions were either not present or lower in magnitude (e.g. 5 dB for 3.19 MPa). Continuous wave (CW) HIFU exposures for 15 s, were then used to generate thermal lesions for peak negative pressures from 0.2-2.71 ± 0.01 MPa at a fluence of 3.4 mJ [Formula: see text]. Inertial cavitation dose (ICD) was monitored during all CW exposures, where exposures combined with both laser illumination and AuNRs resulted in the highest level of detectable emissions. This parameter was integrated over the entire exposure to give a metric to compare with measured thermal lesion area, where it was found that a minimum total ICD of [Formula: see text] a.u. was correlated with the formation of thermal lesions in gel phantoms. Furthermore, lesion area (mm2) was increased for equivalent exposures without either AuNRs or laser illumination. Once combined with cancer targeting AuNRs this approach could allow for the future theranostic use of HIFU, such as providing the ability to identify and treat small multi-focal cancerous regions with minimal damage to surrounding healthy tissue.
Collapse
Affiliation(s)
- J R McLaughlan
- School of Electronic and Electrical Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom. Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | | | | |
Collapse
|
32
|
Pellow C, Goertz DE, Zheng G. Breaking free from vascular confinement: status and prospects for submicron ultrasound contrast agents. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10:e1502. [PMID: 29148219 DOI: 10.1002/wnan.1502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/23/2017] [Accepted: 09/30/2017] [Indexed: 12/11/2022]
Abstract
The development of encapsulated microbubbles (~1-6 μm) has expanded the utility of ultrasound from soft tissue anatomical imaging to not only functional intravascular imaging, but therapeutic interventions, with compelling studies of elicited biological effects. The large diameter of these bubbles has confined their utility to the vasculature, but converging interdisciplinary research pathways are giving rise to new submicron ultrasound contrast agents capable of extending their effects beyond the vascular compartment. This article reviews the status and prospects of exogenous agents including nanobubbles, echogenic liposomes, gas vesicles, cavitation seeds, and nanodroplets, and assesses outstanding criticisms preventing their advance. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Carly Pellow
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Toronto, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - David E Goertz
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Toronto, Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
33
|
Yildirim A, Chattaraj R, Blum NT, Shi D, Kumar K, Goodwin AP. Phospholipid Capped Mesoporous Nanoparticles for Targeted High Intensity Focused Ultrasound Ablation. Adv Healthc Mater 2017; 6:10.1002/adhm.201700514. [PMID: 28699308 PMCID: PMC5627974 DOI: 10.1002/adhm.201700514] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/25/2017] [Indexed: 01/20/2023]
Abstract
The mechanical effects of cavitation can be effective for therapy but difficult to control, thus potentially leading to off-target side effects in patients. While administration of ultrasound active agents such as fluorocarbon microbubbles and nanodroplets can locally enhance the effects of high intensity focused ultrasound (HIFU), it has been challenging to prepare ultrasound active agents that are small and stable enough to accumulate in tumors and internalize into cancer cells. Here, this paper reports the synthesis of 100 nm nanoparticle ultrasound agents based on phospholipid-coated, mesoporous, hydrophobically functionalized silica nanoparticles that can internalize into cancer cells and remain acoustically active. The ultrasound agents produce bubbles when subjected to short HIFU pulses (≈6 µs) with peak negative pressure as low as ≈7 MPa and at particle concentrations down to 12.5 µg mL-1 (7 × 109 particles mL-1 ). Importantly, ultrasound agents are effectively uptaken by cancer cells without cytotoxic effects, but HIFU insonation causes destruction of the cells by the acoustically generated bubbles, as demonstrated by (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) and lactate dehydrogenase assays and flow cytometry. Finally, it is showed that the HIFU dose required to effectively eliminate cancer cells in the presence of ultrasound agents causes only a small temperature increase of ≈3.5 °C.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Rajarshi Chattaraj
- Department of Chemical Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Nicholas T Blum
- Department of Chemical Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Dennis Shi
- Department of Chemical Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Kaushlendra Kumar
- Department of Chemical Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Andrew P Goodwin
- Department of Chemical Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303, USA
| |
Collapse
|