1
|
Al Refaai KA, AlSawaftah NA, Abuwatfa W, Husseini GA. Drug Release via Ultrasound-Activated Nanocarriers for Cancer Treatment: A Review. Pharmaceutics 2024; 16:1383. [PMID: 39598507 PMCID: PMC11597164 DOI: 10.3390/pharmaceutics16111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Conventional cancer chemotherapy often struggles with safely and effectively delivering anticancer therapeutics to target tissues, frequently leading to dose-limiting toxicity and suboptimal therapeutic outcomes. This has created a need for novel therapies that offer greater efficacy, enhanced safety, and improved toxicological profiles. Nanocarriers are nanosized particles specifically designed to enhance the selectivity and effectiveness of chemotherapy drugs while reducing their toxicity. A subset of drug delivery systems utilizes stimuli-responsive nanocarriers, which enable on-demand drug release, prevent premature release, and offer spatial and temporal control over drug delivery. These stimuli can be internal (such as pH and enzymes) or external (such as ultrasound, magnetic fields, and light). This review focuses on the mechanics of ultrasound-induced drug delivery and the various nanocarriers used in conjunction with ultrasound. It will also provide a comprehensive overview of key aspects related to ultrasound-induced drug delivery, including ultrasound parameters and the biological effects of ultrasound waves.
Collapse
Affiliation(s)
- Khaled Armouch Al Refaai
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - Nour A. AlSawaftah
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
2
|
Rajankar N, Aalhate M, Mahajan S, Maji I, Gupta U, Nair R, Paul P, Singh PK. Unveiling multifaceted avenues of echogenic liposomes: Properties, preparation, and potential applications. J Drug Deliv Sci Technol 2024; 99:105931. [DOI: 10.1016/j.jddst.2024.105931] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Jayasankar G, Koilpillai J, Narayanasamy D. A Systematic Study on Long-acting Nanobubbles: Current Advancement and Prospects on Theranostic Properties. Adv Pharm Bull 2024; 14:278-301. [PMID: 39206408 PMCID: PMC11347731 DOI: 10.34172/apb.2024.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 09/04/2024] Open
Abstract
Delivery of diagnostic drugs via nanobubbles (NBs) has shown to be an emerging field of study. Due to their small size, NBs may more easily travel through constricted blood vessels and precisely target certain bodily parts. NB is considered the major treatment for cancer treatment and other diseases which are difficult to diagnose. The field of NBs is dynamic and continues to grow as researchers discover new properties and seek practical applications in various fields. The predominant usage of NBs in novel drug delivery is to enhance the bioavailability, and controlled drug release along with imaging properties NBs are important because they may change interfacial characteristics including surface force, lubrication, and absorption. The quick diffusion of gas into the water was caused by a hypothetical film that was stimulated and punctured by a strong acting force at the gas/water contact of the bubble. In this article, various prominent aspects of NBs have been discussed, along with the long-acting nature, and the theranostical aspect which elucidates the potential marketed drugs along with clinical trial products. The article also covers quality by design aspects, different production techniques that enable method-specific therapeutic applications, increasing the floating time of the bubble, and refining its properties to enhance the prepared NB's quality. NB containing both analysis and curing properties makes it special from other nano-carriers. This work includes all the possible methods of preparing NB, its application, all marketed drugs, and products in clinical trials.
Collapse
Affiliation(s)
| | | | - Damodharan Narayanasamy
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institution of Science and Technology, Kattankulathur, Chengalpattu, India
| |
Collapse
|
4
|
Dehghan Z, Darya G, Mehdinejadiani S, Derakhshanfar A. Comparison of two methods of sperm- and testis-mediated gene transfer in production of transgenic animals: A systematic review. Anim Genet 2024; 55:328-343. [PMID: 38361185 DOI: 10.1111/age.13404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/19/2023] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
Transgenic (Tg) animal technology is one of the growing areas in biology. Various Tg technologies, each with its own advantages and disadvantages, are available for generating Tg animals. These include zygote microinjection, electroporation, viral infection, embryonic stem cell or spermatogonial stem cell-mediated production of Tg animals, sperm-mediated gene transfer (SMGT), and testis-mediated gene transfer (TMGT). However, there are currently no comprehensive studies comparing SMGT and TMGT methods, selecting appropriate gene delivery carriers (such as nanoparticles and liposomes), and determining the optimal route for gene delivery (SMGT and TMGT) for producing Tg animal. Here we aim to provide a comprehensive assessment comparing SMGT and TMGT methods, and to introduce the best carriers and gene transfer methods to sperm and testis to generate Tg animals in different species. From 2010 to 2022, 47 studies on SMGT and 25 studies on TMGT have been conducted. Mice and rats were the most commonly used species in SMGT and TMGT. Regarding the SMGT approach, nanoparticles, streptolysin-O, and virus packaging were found to be the best gene transfer methods for generating Tg mice. In the TMGT method, the best gene transfer methods for generating Tg mice and rats were virus packaging, dimethyl sulfoxide, electroporation, and liposome. Our study has shown that the efficiency of producing Tg animals varies depending on the species, gene carrier, and method of gene transfer.
Collapse
Affiliation(s)
- Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamhossein Darya
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayesteh Mehdinejadiani
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Derakhshanfar
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Maharati A, Moghbeli M. Role of microRNA-505 during tumor progression and metastasis. Pathol Res Pract 2024; 258:155344. [PMID: 38744001 DOI: 10.1016/j.prp.2024.155344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Late diagnosis of cancer in advanced stages due to the lack of screening methods is considered as the main cause of poor prognosis and high mortality rate among these patients. Therefore, it is necessary to investigate the molecular tumor biology in order to introduce biomarkers that can be used in cancer screening programs and early diagnosis. MicroRNAs (miRNAs) have key roles in regulation of the cellular pathophysiological processes. Due to the high stability of miRNAs in body fluids, they are widely used as the non-invasive tumor markers. According to the numerous reports about miR-505 deregulation in a wide range of cancers, we investigated the role of miR-505 during tumor progression. It was shown that miR-505 mainly has the tumor suppressor functions through the regulation of signaling pathways, chromatin remodeling, and cellular metabolism. This review has an effective role in introducing miR-505 as a suitable marker for the early cancer diagnosis.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Moradi Kashkooli F, Jakhmola A, Hornsby TK, Tavakkoli JJ, Kolios MC. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions. J Control Release 2023; 355:552-578. [PMID: 36773959 DOI: 10.1016/j.jconrel.2023.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The application of biocompatible nanocarriers in medicine has provided several benefits over conventional treatment methods. However, achieving high treatment efficacy and deep penetration of nanocarriers in tumor tissue is still challenging. To address this, stimuli-responsive nano-sized drug delivery systems (DDSs) are an active area of investigation in delivering anticancer drugs. While ultrasound is mainly used for diagnostic purposes, it can also be applied to affect cellular function and the delivery/release of anticancer drugs. Therapeutic ultrasound (TUS) has shown potential as both a stand-alone anticancer treatment and a method to induce targeted drug release from nanocarrier systems. TUS approaches have been used to overcome various physiological obstacles, including endothelial barriers, the tumor microenvironment (TME), and immunological hurdles. Combining nanomedicine and ultrasound as a smart DDS can increase in situ drug delivery and improve access to impermeable tissues. Furthermore, smart DDSs can perform targeted drug release in response to distinctive TMEs, external triggers, or dual/multi-stimulus. This results in enhanced treatment efficacy and reduced damage to surrounding healthy tissue or organs at risk. Integrating DDSs and ultrasound is still in its early stages. More research and clinical trials are required to fully understand ultrasound's underlying physical mechanisms and interactions with various types of nanocarriers and different types of cells and tissues. In the present review, ultrasound-mediated nano-sized DDS, specifically focused on cancer treatment, is presented and discussed. Ultrasound interaction with nanoparticles (NPs), drug release mechanisms, and various types of ultrasound-sensitive NPs are examined. Additionally, in vitro, in vivo, and clinical applications of TUS are reviewed in light of the critical challenges that need to be considered to advance TUS toward an efficient, secure, straightforward, and accessible cancer treatment. This study also presents effective TUS parameters and safety considerations for this treatment modality and gives recommendations about system design and operation. Finally, future perspectives are considered, and different TUS approaches are examined and discussed in detail. This review investigates drug release and delivery through ultrasound-mediated nano-sized cancer treatment, both pre-clinically and clinically.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Sumi N, Nagahiro S, Nakata E, Watanabe K, Ohtsuki T. Ultrasound-dependent RNAi using TatU1A-rose bengal conjugate. Bioorg Med Chem Lett 2022; 68:128767. [PMID: 35513220 DOI: 10.1016/j.bmcl.2022.128767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Tat-U1A-rose bengal conjugate (TatU1A-RB) was prepared as an ultrasound-sensitive RNA carrier molecule. This molecule consists of Tat cell-penetrating peptide, U1A RNA-binding protein, and rose bengal as a sonosensitizer. We demonstrated that TatU1A-RB delivered RNA via the endocytosis pathway, which was followed by ultrasound-dependent endosomal escape and cytosolic dispersion of the RNA. A short hairpin RNA (shRNA) delivered by TatU1A-RB mediated RNA interference (RNAi) ultrasound-dependently. Even by ultrasound irradiation through blood cells, RNAi could be induced with TatU1A-RB and the shRNA. This ultrasound-dependent cytosolic RNA delivery method will serve as the basis for a new approach to nucleic acid therapeutics.
Collapse
Affiliation(s)
- Nanako Sumi
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama 700-8530, Japan
| | - Shota Nagahiro
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama 700-8530, Japan
| | - Eiji Nakata
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Kazunori Watanabe
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama 700-8530, Japan
| | - Takashi Ohtsuki
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 3-1-1 Tsushimanaka, Okayama 700-8530, Japan.
| |
Collapse
|
8
|
Singh P, Muhammad I, Nelson NE, Tran KTM, Vinikoor T, Chorsi MT, D’Orio E, Nguyen TD. Transdermal delivery for gene therapy. Drug Deliv Transl Res 2022; 12:2613-2633. [PMID: 35538189 PMCID: PMC9089295 DOI: 10.1007/s13346-022-01138-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
Gene therapy is a critical constituent of treatment approaches for genetic diseases and has gained tremendous attention. Treating and preventing diseases at the genetic level using genetic materials such as DNA or RNAs could be a new avenue in medicine. However, delivering genes is always a challenge as these molecules are sensitive to various enzymes inside the body, often produce systemic toxicity, and suffer from off-targeting problems. In this regard, transdermal delivery has emerged as an appealing approach to enable a high efficiency and low toxicity of genetic medicines. This review systematically summarizes outstanding transdermal gene delivery methods for applications in skin cancer treatment, vaccination, wound healing, and other therapies.
Collapse
Affiliation(s)
- Parbeen Singh
- Department of Mechanical Engineering, University of Connecticut, Storrs, USA
| | - I’jaaz Muhammad
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Nicole E. Nelson
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Khanh T. M. Tran
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Tra Vinikoor
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Meysam T. Chorsi
- Department of Mechanical Engineering, University of Connecticut, Storrs, USA ,Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| | - Ethan D’Orio
- Department of Biomedical Engineering, University of Connecticut, Storrs, USA ,Department of Biomedical Engineering and Department of Advanced Manufacturing for Energy Systems, Storrs, USA
| | - Thanh D. Nguyen
- Department of Mechanical Engineering, University of Connecticut, Storrs, USA ,Department of Biomedical Engineering, University of Connecticut, Storrs, USA
| |
Collapse
|
9
|
Wang J, Mo J, Xie Y, Wang C. Ultrasound microbubbles-mediated miR-216b affects MALAT1-miRNA axis in non-small cell lung cancer cells. Tissue Cell 2021; 74:101703. [PMID: 34896788 DOI: 10.1016/j.tice.2021.101703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/19/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022]
Abstract
MiR-216b is ectopically expressed in various cancers. Ultrasound microbubbles (UTMBs) are an effective method for miRNA delivery. This article mainly explored the involvement of lncRNA in the effects of UTMBs-mediated miR-216b on non-small cell lung cancer (NSCLC) progression. Expressions and relationship of miR-216b and MALAT1 were examined using quantitative real-time polymerase chain reaction (qRT-PCR), Pearson, TargetScan, and dual-luciferase reporter assay. After the transfection with liposome- or UTMBs-mediated miR-216b mimic (M) or MALAT1 overexpression plasmid alone or together, levels of miR-216b and MALAT1, cell biological behaviors, as well as expressions of apoptosis- and epithelial mesenchymal transition (EMT)-related markers were examined using qRT-PCR, cell functional experiments, and western blot. Besides, we used qRT-PCR to quantify the expressions of multiple downstream miRNAs of MALAT1. MiR-216b expression was weakened yet MALAT1 expression was enhanced in NSCLC tissues, and miR-216b was negatively bound to MALAT1. TargetScan analysis manifested that miR-216b, targeted by MALAT1, was down-regulated in NSCLC cells. UTMBs-mediated miR-216b M further intensified miR-216b level yet weakened cell biological behaviors. The inhibitory effect of UTMBs-mediated miR-216b M on cell biological behaviors and MALAT1 expression was greatly better relative to that of miR-216b M. Moreover, miR-216b restrained the cell biological behaviors by repressing MALAT1 expression. We further manifested that miR-216b facilitated the expressions of apoptosis-related markers, but restrained those of EMT-related markers by repressing MALAT1 expression. Moreover, UTMBs-mediated miR-216b M enhanced the expressions of downstream multiple miRNAs of MALAT1, but this tendency was reversed by co-transfection of overexpressed MALAT1 and miR-216b M. Collectively, UTMBs-mediated miR-216b M restrained NSCLC cell growth by modulating the MALAT1-miRNA axis.
Collapse
Affiliation(s)
- Jian Wang
- Thoracic Surgery Department, Shenzhen People's Hospital, China
| | - Jianming Mo
- Pulmonary and Critical Care Medicine Department, Peking University Shenzhen Hospital, China
| | - Yuancai Xie
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, China.
| | - Chunguang Wang
- Thoracic Surgery Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
10
|
Huang X, Dong H, Liu Y, Yu F, Yang S, Chen Z, Li J. Silencing of let-7b-5p inhibits ovarian cancer cell proliferation and stemness characteristics by Asp-Glu-Ala-Asp-box helicase 19A. Bioengineered 2021; 12:7666-7677. [PMID: 34612147 PMCID: PMC8806929 DOI: 10.1080/21655979.2021.1982276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The emergence and recurrence of ovarian cancer are associated with ovarian cancer stem cells. For cancer treatment, gene delivery of microbubbles (MB)-mediated microRNA (miRNA) is considered as a promising approach. In this study, our aim is to investigate the effects of MB-mediated let-7b-5p inhibitor on the proliferation and stemness characteristics of ovarian cancer (OVCA) cells. Let-7b-5p inhibitor mediated by MB was prepared (termed MB-let-7b-5p inhibitor), and the effects of MB-let-7b-5p inhibitor and let-7b-5p inhibitor on OVCA cell viability, proliferation and stemness characteristics were investigated. We found that MB-let-7b-5p inhibitor presented a higher transfection efficiency than let-7b-5p inhibitor alone. The inhibitory effect of MB-let-7b-5p inhibitor on OVCA cells was more significant than let-7b-5p inhibitor. Let-7b-5p targeted DEAD (Asp-Glu-Ala-Asp)-box helicase 19A (DDX19A), which was downregulated in OVCA cells. The downregulation of DDX19A reversed the inhibitory effects of MB-let-7b-5p inhibitor on OVCA cells. To sum up, we found that MB-let-7b-5p suppressed OVCA cell malignant behaviors by targeting DDX19A.
Collapse
Affiliation(s)
- Xiujuan Huang
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Hongxia Dong
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yang Liu
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Fen Yu
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Shunshi Yang
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Jueying Li
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| |
Collapse
|
11
|
Paknahad AA, Kerr L, Wong DA, Kolios MC, Tsai SSH. Biomedical nanobubbles and opportunities for microfluidics. RSC Adv 2021; 11:32750-32774. [PMID: 35493576 PMCID: PMC9042222 DOI: 10.1039/d1ra04890b] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/19/2021] [Indexed: 12/17/2022] Open
Abstract
The use of bulk nanobubbles in biomedicine is increasing in recent years, which is attributable to the array of therapeutic and diagnostic tools promised by developing bulk nanobubble technologies. From cancer drug delivery and ultrasound contrast enhancement to malaria detection and the diagnosis of acute donor tissue rejection, the potential applications of bulk nanobubbles are broad and diverse. Developing these technologies to the point of clinical use may significantly impact the quality of patient care. This review compiles and summarizes a representative collection of the current applications, fabrication techniques, and characterization methods of bulk nanobubbles in biomedicine. Current state-of-the-art generation methods are not designed to create nanobubbles of high concentration and low polydispersity, both characteristics of which are important for several bulk nanobubble applications. To date, microfluidics has not been widely considered as a tool for generating nanobubbles, even though the small-scale precision and real-time control offered by microfluidics may overcome the challenges mentioned above. We suggest possible uses of microfluidics for improving the quality of bulk nanobubble populations and propose ways of leveraging existing microfluidic technologies, such as organ-on-a-chip platforms, to expand the experimental toolbox of researchers working to develop biomedical nanobubbles. The use of bulk nanobubbles in biomedicine is increasing in recent years. This translates into new opportunities for microfluidics, which may enable the generation of higher quality nanobubbles that lead to advances in diagnostics and therapeutics.![]()
Collapse
Affiliation(s)
- Ali A Paknahad
- Department of Mechanical and Industrial Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada .,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada
| | - Liam Kerr
- Department of Mechanical and Industrial Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada .,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada
| | - Daniel A Wong
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada.,Department of Electrical, Computer, and Biomedical Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada
| | - Michael C Kolios
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada.,Department of Physics, Ryerson University Toronto Ontario M5B 2K3 Canada
| | - Scott S H Tsai
- Department of Mechanical and Industrial Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada .,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada.,Graduate Program in Biomedical Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada
| |
Collapse
|
12
|
Ultrasound-Responsive Smart Drug Delivery System of Lipid Coated Mesoporous Silica Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13091396. [PMID: 34575472 PMCID: PMC8468042 DOI: 10.3390/pharmaceutics13091396] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/23/2022] Open
Abstract
The immediate release of chemotherapeutics at the target site, along with no premature release in circulation is always challenging. The purpose of this study was to develop a stimuli responsive drug delivery system, composed of lipid supported mesoporous silica nanoparticles (MSNPs) for triggered drug release at the target site and simultaneously avoiding the premature release. MSNPs with a higher drug loading capacity and very slow release were designed so as to enhance release by FDA approved US-irradiation. Doxorubicin, as a model drug, and perfluoropentane (PFP) as a US responsive material, were entrapped in the porous structure of MSNPs. Lipid coating enhanced the cellular uptake and in addition provided a gatekeeping effect at the pore opening to reduce premature release. The mechanical and thermal effects of US induced the conversion of liquid PFP to a gaseous form that was able to rupture the lipid layer, resulting in triggered drug release. The prolonged stability profile and non-toxic behavior made them suitable candidate for the delivery of anticancer drugs. This smart system, with the abilities of better cellular uptake and higher cytotoxic effects on US-irradiation, would be a good addition to the applied side of chemotherapeutic advanced drug delivery systems.
Collapse
|
13
|
Kamali M, Dewil R, Appels L, Aminabhavi TM. Nanostructured materials via green sonochemical routes - Sustainability aspects. CHEMOSPHERE 2021; 276:130146. [PMID: 33740648 DOI: 10.1016/j.chemosphere.2021.130146] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 02/01/2021] [Accepted: 02/25/2021] [Indexed: 06/12/2023]
Abstract
The production of environmentally friendly nanostructured materials with well-defined properties is a major challenge. Characteristics of the nanomaterials such as dimensionality, size and morphology strongly affect their performance in various applications. Additionally, sustainability considerations require an acceptable level of efficiency while being economically feasible and environmentally benign. The use of ultrasonic irradiation (UI) is a green and powerful technology, which can be applied for the synthesis of a variety of nanostructured materials. This review critically discusses the progress made in the fabrication of environmentally benign engineered nanomaterials with various dimensionalities (i.e., zero, one, two, or three dimensions) assisted by UI. The evolution and current status in this area are further illustrated using a scientometric approach. Application of UI for the synthesis of nanostructured materials has been also assessed according to the main sustainability pillars including the performance and environmental compatibility, as well as the relevant economic and social considerations. The outlook as well as recommendations for future research has been also provided and discussed towards the promotion of sustainable nanomaterials synthesis and application in various fields.
Collapse
Affiliation(s)
- Mohammadreza Kamali
- KU Leuven, Department of Chemical Engineering, Process and Environmental Technology Lab, J. De Nayerlaan 5, 2860, Sint-Katelijne-Waver, Belgium
| | - Raf Dewil
- KU Leuven, Department of Chemical Engineering, Process and Environmental Technology Lab, J. De Nayerlaan 5, 2860, Sint-Katelijne-Waver, Belgium.
| | - Lise Appels
- KU Leuven, Department of Chemical Engineering, Process and Environmental Technology Lab, J. De Nayerlaan 5, 2860, Sint-Katelijne-Waver, Belgium
| | - Tejraj M Aminabhavi
- Pharmaceutical Engineering, SETs' College of Pharmacy, Dharwad, 580002, India.
| |
Collapse
|
14
|
Li Y, Chen Z, Ge S. Sonoporation: Underlying Mechanisms and Applications in Cellular Regulation. BIO INTEGRATION 2021. [DOI: 10.15212/bioi-2020-0028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ultrasound combined with microbubble-mediated sonoporation has been applied to enhance drug or gene intracellular delivery. Sonoporation leads to the formation of openings in the cell membrane, triggered by ultrasound-mediated oscillations and destruction of microbubbles. Multiple mechanisms
are involved in the occurrence of sonoporation, including ultrasonic parameters, microbubbles size, and the distance of microbubbles to cells. Recent advances are beginning to extend applications through the assistance of contrast agents, which allow ultrasound to connect directly to cellular
functions such as gene expression, cellular apoptosis, differentiation, and even epigenetic reprogramming. In this review, we summarize the current state of the art concerning microbubble‐cell interactions and sonoporation effects leading to cellular functions.
Collapse
Affiliation(s)
- Yue Li
- First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhiyi Chen
- First Affiliated Hospital of University of South China, Hengyang, China
| | - Shuping Ge
- Department of Pediatrics, St Christopher’s Hospital for Children, Tower Health and Drexel University, Philadelphia, PA (S.G.)
| |
Collapse
|
15
|
Omar MM, Hasan OA, Zaki RM, Eleraky NE. Externally Triggered Novel Rapid-Release Sonosensitive Folate-Modified Liposomes for Gemcitabine: Development and Characteristics. Int J Nanomedicine 2021; 16:683-700. [PMID: 33536754 PMCID: PMC7850458 DOI: 10.2147/ijn.s266676] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/10/2020] [Indexed: 01/07/2023] Open
Abstract
PURPOSE To develop an externally triggered rapid-release targeted system for treating ovarian cancer, gemcitabine (GMC) was entrapped into sonosensitive (SoS) folate (Fo)-modified liposomes (LPs). METHODS GMC-loaded LPs (GMC LPs), GMC-loaded Fo-targeted LPs (GMC-Fo LPs), and GMC-loaded Fo-targeted SoS LPs (GMC-SoS Fo LPs) were prepared utilizing a film-hydration technique and evaluated based on particle size, ζ-potential, and percentage entrapped drug. Cellular uptake of the fluorescent delivery systems in Fo-expressing ovarian cancer cells was quantified using flow cytometry. Finally, tumor-targeting ability, in vivo evaluation, and pharmacokinetic studies were performed. RESULTS GMC LPs, GMC-Fo LPs, and GMC-SoS Fo LPs were successfully prepared, with sizes of <120.3±2.4 nm, 39.7 mV ζ-potential, and 86.3%±1.84% entrapped drug. Cellular uptake of GMC-SoS Fo LPs improved 6.51-fold over GMC LPs (under ultrasonic irradiation - p<0.05). However, cellular uptake of GMC-Fo LPs improved just 1.24-fold over GMC LPs (p>0.05). Biodistribution study showed that of GMC concentration in tumors treated with GMC-SoS-Fo LPs (with ultrasound) improved 2.89-fold that of free GMC (p<0.05). In vivo, GMC-SoS Fo LPs showed the highest antiproliferative and antitumor action on ovarian cancer. CONCLUSION These findings showed that externally triggered rapid-release SoS Fo-modified LPs are a promising system for delivering rapid-release drugs into tumors.
Collapse
Affiliation(s)
- Mahmoud M Omar
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Deraya University, Minia, 61768, Egypt
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy,Sohag University, Sohag, 82524, Egypt
| | - Omiya Ali Hasan
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Deraya University, Minia, 61768, Egypt
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy,Sohag University, Sohag, 82524, Egypt
| | - Randa Mohammed Zaki
- Department of Pharmaceutics, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Nermin E Eleraky
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| |
Collapse
|
16
|
Basuony SAHAE, Hamed RS. Anti-Micro RNA-221 a Promising Genetic Therapy of Oral Squamous Cell Carcinoma (SCC-25). Braz Dent J 2020; 31:634-639. [PMID: 33237235 DOI: 10.1590/0103-6440202003350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Micro-RNA-221(miR-221) is one of oncogenic miRNAs that plays a vital role in the development and progression of oral cancers. The aim of this study is to introduce a new gene therapy for oral squamous cell carcinoma by blocking the expression of oncogenic miR-221 by its inhibitor. The present work was performed on squamous cell carcinoma cell line SCC-25 and anti-miR-221 was delivered to the cells using an ultrasound micro bubbles. Assessment of the effect of miR-221 inhibitor on SCC-25 cells was done using MTT assay, cell cycle analysis and apoptosis detection. In addition, reverse transcription-polymerase chain reaction was also used to detect the expression -miR-221 and its target genes. Using ANOVA, statistical analysis of the results showed significant inhibition of cell viability with and induction of cell apoptosis of SCC-25 cell line after transfection. Moreover, the expression of miR-221, Epidermal growth factor receptor (EGFR) and CDKNIB/p27 were downregulated without significant difference. Transfection of SCC-25 by inhibitor of miR-221 resulting in blockage of its expression leading to arresting of tumor growth. These results proved the effective role of micro-RNA inhibitors as novel therapeutic agent for oral cancers.
Collapse
Affiliation(s)
| | - Reham S Hamed
- Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Cairo University, Egypt
| |
Collapse
|
17
|
Yu Y, Zhao Y, Zhou G, Wang X. Therapeutic Efficacy of Delta-Like Ligand 4 Gene Vaccine Overexpression on Liver Cancer in Mice. Technol Cancer Res Treat 2020; 19:1533033820942205. [PMID: 33191858 PMCID: PMC7672725 DOI: 10.1177/1533033820942205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Delta-like ligand 4 is a notch ligand that is predominantly expressed in the endothelial tip cells and plays essential roles in the regulation of angiogenesis. In this study, we explored the therapeutic effects of delta-like ligand 4 gene vaccine overexpression on the syngeneic model mouse model of liver cancer and the underlying mechanisms. Mouse hepatocellular carcinoma cell line H22-H8D8 was used to generate subcutaneous syngeneic model liver cancer in Kunming mice, and the effects of recombinant plasmid pVAX1 containing delta-like ligand 4 vaccine on tumor growth was examined. Compared to controls, delta-like ligand 4 vaccination reduced syngeneic model tumor size by 70.31% (from 17.11 ± 9.30 cm3 to 5.08 ± 2.75 cm3, P = .035) and tumor weight by 34.19% (from 6.26 ± 3.01 g to 4.12 ± 2.52 g, P = .102), while the mouse survival was significantly increased (from 27.7 ± 6.0 days to 33.1 ± 6.1 days, P = .047). High level of delta-like ligand 4 antibody, together with a significantly increased number of CD4+ and decreased CD8+ cells were identified in the mouse peripheral blood serum samples after delta-like ligand 4 immunization. In addition, elevated serum levels of interleukin 2, interleukin 4, and interferon γ were detected in the delta-like ligand 4-vaccinated mice when compared to the controls. Further studies have revealed increased CD31 and decreased Ki67 expression in the syngeneic model tumor tissues of vaccinated mice. Taken together, our studies suggest that delta-like ligand 4 gene vaccine can inhibit the growth of hepatocellular carcinoma in mice through inhibiting tumor angiogenesis and boosting antitumor immune responses. Hence, delta-like ligand 4 gene vaccination may be a promising strategy for the treatment of transplanted liver cancer.
Collapse
Affiliation(s)
- Yi Yu
- Key Laboratory of Digestive Disease, Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yang Zhao
- Key Laboratory of Digestive System Tumors, Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Guangming Zhou
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Xiang Wang
- Key Laboratory of Digestive Disease, Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Xiang Wang, Key Laboratory of Digestive Disease, Department of Gastroenterology, Lanzhou University Second Hospital, 82 Cuiyingmen, Lanzhou 730030, China.
| |
Collapse
|
18
|
Yan W, Cheng L, Zhang D. Ultrasound-Targeted Microbubble Destruction Mediated si-CyclinD1 Inhibits the Development of Hepatocellular Carcinoma via Suppression of PI3K/AKT Signaling Pathway. Cancer Manag Res 2020; 12:10829-10839. [PMID: 33149688 PMCID: PMC7605614 DOI: 10.2147/cmar.s263590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Background and Aim In our study, we aimed to investigate the effect of ultrasound-targeted microbubble destruction (UTMD)mediated si-CyclinD1 (CCND1) on the growth of hepatocellular carcinoma (HCC) cells. Patients and Methods Bioinformatics analysis was performed to detect the difference of CCND1 expression of HCC and normal liver tissues. After treatment with UTMDmediated si-CCND1, the growth and apoptosis of HepG2 cells were detected by flow cytometry, MTT, EdU staining, colony formation assay, Hoechst 33,258 staining and Western blot analysis. The growth of HepG2 cells in vivo was also studied via xenograft tumor in nude mice. Results CCND1 was highly expressed in HCC tissues and HCC cell lines. UTMDmediated si-CCND1 could inhibit the growth of HepG2 cells and promote apoptosis via suppression of the PI3K/AKT signaling pathway. UTMDmediated si-CCND1 could also suppress the growth of HepG2 cells in vivo. Conclusion Our study provided evidence that UTMDmediated si-CCND1 could inhibit the growth of HepG2 cells and promote apoptosis via suppression of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Wei Yan
- Department of Electrical Diagnosis, Changchun University of Traditional Chinese Medicine Affiliated Hospital, Changchun 130021, People's Republic of China
| | - Li Cheng
- Department of Electrical Diagnosis, Changchun University of Traditional Chinese Medicine Affiliated Hospital, Changchun 130021, People's Republic of China
| | - Dongmei Zhang
- Department of Electrical Diagnosis, Changchun University of Traditional Chinese Medicine Affiliated Hospital, Changchun 130021, People's Republic of China
| |
Collapse
|
19
|
Ultrasound Microbubble-Mediated microRNA-505 Regulates Cervical Cancer Cell Growth via AKT2. ACTA ACUST UNITED AC 2020; 2020:3731953. [PMID: 33123457 PMCID: PMC7584975 DOI: 10.1155/2020/3731953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/25/2020] [Indexed: 02/08/2023]
Abstract
The application of ultrasound and microbubbles (USMB-) mediated microRNA (miR) is a promising approach of gene delivery for cancer treatment. We aimed to discuss the effects of USMB-miR-505 on cervical cancer (CC) development. miR-505 mediated by USMB was prepared. The effect of miR-505 on its transfection efficiency and the effect of miR-505 on HeLa cell proliferation, cell cycle, apoptosis, migration, and invasion were studied. The target gene of miR-505 was predicted, and its expression in CC was detected. The effect of the target gene on HeLa cells was further verified. USMB-miR-505 showed a higher transfection efficiency than miR-505 alone. The inhibitory effect of miR-505 mediated by USMB on HeLa cells was better than miR-505. miR-505 targeted AKT2, which was upregulated in CC. Overexpression of AKT2 reversed the inhibitory effect of USMB-miR-505 on HeLa cell malignant behaviors. Overall, we highlighted that USMB-miR-505 inhibited HeLa cell malignant behaviors by targeting AKT2.
Collapse
|
20
|
Karmur BS, Philteos J, Abbasian A, Zacharia BE, Lipsman N, Levin V, Grossman S, Mansouri A. Blood-Brain Barrier Disruption in Neuro-Oncology: Strategies, Failures, and Challenges to Overcome. Front Oncol 2020; 10:563840. [PMID: 33072591 PMCID: PMC7531249 DOI: 10.3389/fonc.2020.563840] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/13/2020] [Indexed: 01/05/2023] Open
Abstract
The blood-brain barrier (BBB) presents a formidable challenge in the development of effective therapeutics in neuro-oncology. This has fueled several decades of efforts to develop strategies for disrupting the BBB, but progress has not been satisfactory. As such, numerous drug- and device-based methods are currently being investigated in humans. Through a focused assessment of completed, active, and pending clinical trials, our first aim in this review is to outline the scientific foundation, successes, and limitations of the BBBD strategies developed to date. Among 35 registered trials relevant to BBBD in neuro-oncology in the ClinicalTrials.gov database, mannitol was the most common drug-based method, followed by RMP-7 and regadenoson. MR-guided focused ultrasound was the most common device-based method, followed by MR-guided laser ablation, ultrasound, and transcranial magnetic stimulation. While most early-phase studies focusing on safety and tolerability have met stated objectives, advanced-phase studies focusing on survival differences and objective tumor response have been limited by heterogeneous populations and tumors, along with a lack of control arms. Based on shared challenges among all methods, our second objective is to discuss strategies for confirmation of BBBD, choice of systemic agent and drug design, alignment of BBBD method with real-world clinical workflow, and consideration of inadvertent toxicity associated with disrupting an evolutionarily-refined barrier. Finally, we conclude with a strategic proposal to approach future studies assessing BBBD.
Collapse
Affiliation(s)
- Brij S Karmur
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Aram Abbasian
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Brad E Zacharia
- Penn State Health Neurosurgery, College of Medicine, Penn State University, Hershey, PA, United States
| | - Nir Lipsman
- Division of Neurosurgery, University of Toronto, Toronto, ON, Canada
| | - Victor Levin
- Department of Neurosurgery, Medical School, University of California, San Francisco, San Francisco, CA, United States
| | - Stuart Grossman
- Department of Oncology, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Alireza Mansouri
- Penn State Health Neurosurgery, College of Medicine, Penn State University, Hershey, PA, United States
| |
Collapse
|
21
|
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
|
22
|
Fisher DG, Price RJ. Recent Advances in the Use of Focused Ultrasound for Magnetic Resonance Image-Guided Therapeutic Nanoparticle Delivery to the Central Nervous System. Front Pharmacol 2019; 10:1348. [PMID: 31798453 PMCID: PMC6864822 DOI: 10.3389/fphar.2019.01348] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Targeting systemically-administered drugs and genes to specific regions of the central nervous system (CNS) remains a challenge. With applications extending into numerous disorders and cancers, there is an obvious need for approaches that facilitate the delivery of therapeutics across the impervious blood-brain barrier (BBB). Focused ultrasound (FUS) is an emerging treatment method that leverages acoustic energy to oscillate simultaneously administered contrast agent microbubbles. This FUS-mediated technique temporarily disrupts the BBB, allowing ordinarily impenetrable agents to diffuse and/or convect into the CNS. Under magnetic resonance image guidance, FUS and microbubbles enable regional targeting-limiting the large, and potentially toxic, dosage that is often characteristic of systemically-administered therapies. Subsequent to delivery across the BBB, therapeutics face yet another challenge: penetrating the electrostatically-charged, mesh-like brain parenchyma. Non-bioadhesive, encapsulated nanoparticles can help overcome this additional barrier to promote widespread treatment in selected target areas. Furthermore, nanoparticles offer significant advantages over conventional systemically-administered therapeutics. Surface modifications of nanoparticles can be engineered to enhance targeted cellular uptake, and nanoparticle formulations can be tailored to control many pharmacokinetic properties such as rate of drug liberation, distribution, and excretion. For instance, nanoparticles loaded with gene plasmids foster relatively stable transfection, thus obviating the need for multiple, successive treatments. As the formulations and applications of these nanoparticles can vary greatly, this review article provides an overview of FUS coupled with polymeric or lipid-based nanoparticles currently utilized for drug delivery, diagnosis, and assessment of function in the CNS.
Collapse
Affiliation(s)
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
23
|
Meng L, Yuan S, Zhu L, ShangGuan Z, Zhao R. Ultrasound-microbubbles-mediated microRNA-449a inhibits lung cancer cell growth via the regulation of Notch1. Onco Targets Ther 2019; 12:7437-7450. [PMID: 31686849 PMCID: PMC6752164 DOI: 10.2147/ott.s217021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Background The application of gene-loaded microbubbles (MBs) combined with ultrasound that results in increased delivery efficiency may be an excellent method of gene delivery. This study aimed to discuss the effects of ultrasound-MB-mediated microRNA (miR)-449a on lung cancer (LC) development by targeting Notch1. Methods Initially, miR-449a expression in LC tissues, paracancerous tissues, LC cell lines, and lung epithelial cells was detected and its association with LC patients’ clinical characteristics was analyzed. The gain-of-function studies were performed to probe the roles of miR-449a and ultrasound-MB-mediated miR-449a in LC progression. Then, RT-qPCR combined with Western blot analysis was applied to verify the levels of miR-449a, Notch1, proliferation- and apoptosis-related proteins. Moreover, xenograft tumors in nude mice were also applied for in vivo experiments. Results Poorly expressed miR-449a was observed in LC, and its expression was associated with clinical staging, differentiation and lymph node metastasis of LC patients. Overexpression of miR-449a suppressed LC cell proliferation and promoted G2/M arrest and apoptosis. Ultrasound-MB-mediated miR-449a strengthened inhibitory effects of miR-449a on cell growth and resistance to apoptosis. miR-449a inhibited H1299 cell activity by targeting Notch1. Conclusion Our data supported that miR-449a overexpression inhibited LC cell growth, and ultrasound-MB-mediated miR-449a reinforced the repressive effects of miR-449a on LC progression. This investigation may offer new insight for LC treatment.
Collapse
Affiliation(s)
- Lingwu Meng
- Department of Minimally Invasive Therapy, First People's Hospital of Shangqiu, Shangqiu 476100, People's Republic of China
| | - Shaofei Yuan
- Department of Thoracic Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang Province 325200, People's Republic of China
| | - Linjia Zhu
- Department of Thoracic Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang Province 325200, People's Republic of China
| | - Zongxiao ShangGuan
- Department of Thoracic Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang Province 325200, People's Republic of China
| | - Renguo Zhao
- Department of Thoracic Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Ruian, Zhejiang Province 325200, People's Republic of China
| |
Collapse
|
24
|
Andrews LE, Chan MH, Liu RS. Nano-lipospheres as acoustically active ultrasound contrast agents: evolving tumor imaging and therapy technique. NANOTECHNOLOGY 2019; 30:182001. [PMID: 30645984 DOI: 10.1088/1361-6528/aafeb9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Applying nanobubbles (NBs) for contrast-enhanced ultrasound imaging has received increased attention. NBs are biocompatible, multifunctional, theranostic agents. Their properties of high echogenicity and stability create an agent suitable for ultrasonography diagnosis. Their favorable properties of size, in vivo stability, and ease of modification are being exploited to implement a theranostic platform for cancer treatment. The considerable development offers the potential to overcome drug resistance and adverse side effects that are associated with traditional chemotherapy. This review outlines the principles of ultrasonography and angiogenesis. Microbubbles and micelles are also discussed to underline the superior capabilities of NBs for the application. NBs could passively accumulate to tumor tissue by enhanced permeability and retention effect. In addition, it can also achieve the active transportation by surface modification. Active targeting modalities and stimuli-responsive drug delivery modifications generate a therapeutic vehicle. The cytotoxicity of NBs formulations, multimodal imaging capability, active targeting mechanisms, and drug delivery methods are highlighted to confirm the NB as a vehicle for targeted treatment and enhanced ultrasound imaging.
Collapse
Affiliation(s)
- Laura Emma Andrews
- Department of Chemistry, National Taiwan University, Taiwan. School of Chemistry, The University of Edinburgh, United Kingdom
| | | | | |
Collapse
|
25
|
Low Electric Treatment activates Rho GTPase via Heat Shock Protein 90 and Protein Kinase C for Intracellular Delivery of siRNA. Sci Rep 2019; 9:4114. [PMID: 30858501 PMCID: PMC6412017 DOI: 10.1038/s41598-019-40904-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 01/28/2019] [Indexed: 11/08/2022] Open
Abstract
Low electric treatment (LET) promotes intracellular delivery of naked siRNA by altering cellular physiology. However, which signaling molecules and cellular events contribute to LET-mediated siRNA uptake are unclear. Here, we used isobaric tags in relative and absolute quantification (iTRAQ) proteomic analysis to identify changes in the levels of phosphorylated proteins that occur during cellular uptake of siRNA promoted by LET. iTRAQ analysis revealed that heat shock protein 90 (Hsp90)α and myristoylated alanine-rich C-kinase substrate (Marcks) were highly phosphorylated following LET of NIH 3T3 cells, but not untreated cells. Furthermore, the levels of phosphorylated Hsp90α and protein kinase C (PKC)γ were increased by LET both with siRNA and liposomes having various physicochemical properties used as model macromolecules, suggesting that PKCγ activated partly by Ca2+ influx as well as Hsp90 chaperone function were involved in LET-mediated cellular siRNA uptake. Furthermore, LET with siRNA induced activation of Rho GTPase via Hsp90 and PKC, which could contribute to cellular siRNA uptake accompanied by actin cytoskeleton remodeling. Collectively, our results suggested that LET-induced Rho GTPase activation via Hsp90 and PKC would participate in actin-dependent cellular uptake of siRNA.
Collapse
|
26
|
Keller S, Bruce M, Averkiou MA. Ultrasound Imaging of Microbubble Activity during Sonoporation Pulse Sequences. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:833-845. [PMID: 30638695 PMCID: PMC6690385 DOI: 10.1016/j.ultrasmedbio.2018.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 05/28/2023]
Abstract
Ultrasound-mediated drug delivery using the mechanical action of oscillating and/or collapsing microbubbles has been studied on many different experimental platforms, both in vitro and in vivo; however, the mechanisms remain to be elucidated. Many groups use sterile, enclosed chambers, such as Opticells and Clinicells, to optimize acoustic parameters in vitro needed for effective drug delivery in vivo, as well as for mechanistic investigation of sonoporation or the use of sound to permeate cell membranes. In these containers, cell monolayers are seeded on one side, and the remainder of the volume is filled with a solution containing microbubbles and a model drug. Ultrasound is then applied to study the effect of different parameters on model drug uptake in cell monolayers. Despite the simplicity of this system, the field has been unable to appropriately address what parameters and microbubble concentrations are most effective at enhancing drug uptake and minimizing cellular toxicity. In this work, a common in vitro sonoporation experimental setup was characterized through quantitative analysis of microbubble-dependent acoustic attenuation in combination with high-frame-rate and high-resolution imaging of bubble activity during sonoporation pulse sequences. The goal was to visualize the effect that ultrasound parameters have on microbubble activity. It was observed that under literature-derived sonoporation conditions (0.1-1 MPa, 20-1000 cycles and 10,000 to 10,000,000 microbubbles/mL), there is strong and non-linear acoustic attenuation, as well as bubble destruction, gas diffusion and bubble motion resulting in spatiotemporal pressure and concentration gradients. Ultimately, it was found that the acoustic conditions in common in vitro sonoporation setups are much more complex and confounding than often assumed.
Collapse
Affiliation(s)
- Sara Keller
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Matthew Bruce
- Applied Physics Laboratory, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
27
|
Hamarat Şanlıer Ş, Ak G, Yılmaz H, Ünal A, Bozkaya ÜF, Tanıyan G, Yıldırım Y, Yıldız Türkyılmaz G. Development of Ultrasound-Triggered and Magnetic-Targeted Nanobubble System for Dual-Drug Delivery. J Pharm Sci 2019; 108:1272-1283. [DOI: 10.1016/j.xphs.2018.10.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/14/2018] [Accepted: 10/16/2018] [Indexed: 01/06/2023]
|
28
|
Abstract
Theranostics is a term used to describe the combination of diagnostic and therapeutic functions in a single agent. Ultrasound, for example, is a good tool for theranostics due to its multi-potency as both a diagnostic tool using sonography, and as a therapeutic, i.e., by high intensity focused ultrasound (HIFU). Likewise, microbubbles and nanobubbles are not only used as contrast imaging agents, but also as enhancers of drug delivery. Recently, the combination of these bubbles with low intensity ultrasound has been utilized as an effective drug delivery system. We have implemented a similar technique by combining bubbles and ultrasound to study cancer gene therapy and chemotherapy. In addition, we have used high intensity ultrasound as a method for directly damaging tumor cells, thus serving as a cancer therapy. For effective cancer treatment, however, the properties of the bubbles are of utmost importance. Currently, we are applying these bubbles to various therapeutic strategies in cancer treatment. In this session, we would like to introduce the feasibility study of our use of these bubbles in cancer treatment.
Collapse
Affiliation(s)
- Ryo Suzuki
- Faculty of Pharma-Sciences, Teikyo University
| | | |
Collapse
|
29
|
Noble-Vranish ML, Song S, Morrison KP, Tran DM, Sun RR, Loeb KR, Keilman GW, Miao CH. Ultrasound-Mediated Gene Therapy in Swine Livers Using Single-Element, Multi-lensed, High-Intensity Ultrasound Transducers. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 10:179-188. [PMID: 30105275 PMCID: PMC6077835 DOI: 10.1016/j.omtm.2018.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/26/2018] [Indexed: 01/21/2023]
Abstract
We have achieved significant enhancement of gene delivery into livers of large animals using ultrasound (US)-targeted microbubble (MB) destruction methods. An infusion of pGL4 (encoding a luciferase reporter gene) plasmid DNA (pDNA) and MBs into a portal-vein segmental branch of a porcine liver was exposed to US for 4 min. Therapeutic US induced cavitation of MBs to temporarily permeabilize the vascular endothelium and cell membranes, allowing entry of pDNA. We obtained a 64-fold enhancement in luciferase expression in pig livers compared to control without US using an unfocused, dual-element transducer (H105, center frequency [fc] = 1.10 MHz) at 2.7 MPa peak negative pressure (PNP). However, input electrical energy was limited, and modified transducers were designed to have spherical (H185A, fc = 1.10 MHz) or cylindrical foci (H185B, fc = 1.10 MHz; H185D, fc = 1.05 MHz) to enhance PNP output. The revised transducers required less electrical input to achieve 2.7 MPa PNP compared to H105, thereby allowing PNP outputs of up to 6.2 MPa without surpassing the piezo-material limitations. Subsequently, luciferase expression significantly improved up to 9,000-fold compared to controls with minor liver damage. These advancements will allow us to modify our current protocols toward minimally invasive US gene therapy.
Collapse
Affiliation(s)
- Misty L Noble-Vranish
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Shuxian Song
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Dominic M Tran
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Ryan R Sun
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Keith R Loeb
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Carol H Miao
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
30
|
Abstract
Although viral vectors comprise the majority of gene delivery vectors, their various safety, production, and other practical concerns have left a research gap to be addressed. The non-viral vector space encompasses a growing variety of physical and chemical methods capable of gene delivery into the nuclei of target cells. Major physical methods described in this chapter are microinjection, electroporation, and ballistic injection, magnetofection, sonoporation, optical transfection, and localized hyperthermia. Major chemical methods described in this chapter are lipofection, polyfection, gold complexation, and carbon-based methods. Combination approaches to improve transfection efficiency or reduce immunological response have shown great promise in expanding the scope of non-viral gene delivery.
Collapse
Affiliation(s)
- Chi Hong Sum
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | | | - Shirley Wong
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | | |
Collapse
|
31
|
Abdalkader R, Kawakami S, Unga J, Higuchi Y, Suzuki R, Maruyama K, Yamashita F, Hashida M. The development of mechanically formed stable nanobubbles intended for sonoporation-mediated gene transfection. Drug Deliv 2017; 24:320-327. [PMID: 28165819 PMCID: PMC8241156 DOI: 10.1080/10717544.2016.1250139] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/11/2016] [Accepted: 10/14/2016] [Indexed: 12/23/2022] Open
Abstract
In this study, stable nano-sized bubbles (nanobubbles [NBs]) were produced using the mechanical agitation method in the presence of perfluorocarbon gases. NBs made with perfluoropropane had a smaller size (around 400 nm) compared to that of those made with perfluorobutane or nitrogen gas. The lipid concentration in NBs affected both their initial size and post-formulation stability. NBs formed with a final lipid concentration of 0.5 mg/ml tended to be more stable, having a uniform size distribution for 24 h at room temperature and 50 h at 4 °C. In vitro gene expression revealed that NBs/pDNA in combination with ultrasound (US) irradiation had significantly higher transfection efficacy in colon C26 cells. Moreover, for in vivo gene transfection in mice left limb muscles, there was notable local transfection activity by NBs/pDNA when combined with US irradiation. In addition, the aged NBs kept at room temperature or 4 °C were still functional at enhancing gene transfection in mice. We succeeded in preparing stable NBs for efficient in vivo gene transfection, using the mechanical agitation method.
Collapse
Affiliation(s)
- Rodi Abdalkader
- Department of Drug Delivery Researches, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Johan Unga
- Department of Drug Delivery System, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan, and
| | - Yuriko Higuchi
- Department of Drug Delivery Researches, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Ryo Suzuki
- Department of Drug Delivery System, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan, and
| | - Kazuo Maruyama
- Department of Drug Delivery System, Faculty of Pharma-Sciences, Teikyo University, Tokyo, Japan, and
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Researches, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Mitsuru Hashida
- Department of Drug Delivery Researches, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Kyoto University Institute for Integrated Cell-Material Science (iCeMS), Kyoto, Japan
| |
Collapse
|
32
|
Du J, Zhao X, Li B, Mou Y, Wang Y. DNA-loaded microbubbles with crosslinked bovine serum albumin shells for ultrasound-promoted gene delivery and transfection. Colloids Surf B Biointerfaces 2017; 161:279-287. [PMID: 29096372 DOI: 10.1016/j.colsurfb.2017.10.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 12/30/2022]
Abstract
The microbubble is a kind of clinically applied ultrasound contrast agent in disease diagnosis that can also rupture under sonication to increase membrane permeability and promote gene entry into targeted cells. However, the development of ultrasound-mediated gene delivery might be restricted because genes and microbubbles were separated and would not reach the targeted cells simultaneously. Herein, a kind of crosslinked positive microbubbles (CPMBs) were prepared to load DNA as gene vectors to promote gene delivery efficiency. The BSA shell of the CPMBs was crosslinked with disulfide bonds, which obviously enhanced the stability of the CPMBs. Furthermore, the CPMBs revealed sonoporation effects comparable to those of clinically applied SonoVue microbubbles. As DNA and CPMBs were electrostatically linked as an entirety, they would reach cells simultaneously. Thus, with the aid of ultrasound, these DNA-loaded microbubbles promoted DNA entry into cytoplasm more effectively and obtained higher cellular uptake efficiency and better transfection efficiency than DNA-mixed microbubbles. Confocal microscopy results showed that rupturing of the CPMBs/DNA entire microbubbles under sonication could carry DNA directly into the cytoplasm or nucleus. All results indicated that the cytocompatible DNA-loaded microbubbles have promising prospects in ultrasound-mediated gene delivery.
Collapse
Affiliation(s)
- Jianwei Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Xiao Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Bangbang Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Yun Mou
- Echocardiography and Vascular Ultrasound Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China.
| |
Collapse
|
33
|
Kono Y, Jinzai H, Kotera Y, Fujita T. Influence of Physicochemical Properties and PEG Modification of Magnetic Liposomes on Their Interaction with Intestinal Epithelial Caco-2 Cells. Biol Pharm Bull 2017; 40:2166-2174. [PMID: 28966298 DOI: 10.1248/bpb.b17-00563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study aimed to investigate the effect of particle size (100, 500 nm), surface charge (cationic, neutral and anionic) and polyethylene glycol (PEG) modification of magnetic liposomes on their interaction with the human intestinal epithelial cell line, Caco-2. The cellular associated amount of all the magnetic liposomes was significantly increased by the presence of a magnetic field. The highest association and internalization into Caco-2 cells was observed with magnetic cationic liposomes. Moreover, small magnetic liposomes were more efficiently associated and taken up into the cells, than large ones. In contrast, PEG modification significantly attenuated the enhancing effect of the magnetic field on the cellular association of magnetic liposomes. We also found that magnetic cationic liposomes had the highest retention properties to Caco-2 cells. Moreover, the retention of large magnetic liposomes to the cells was much longer than that of small ones. In addition, magnetic cationic and neutral liposomes had relatively high stability in Caco-2 cells, whereas magnetic anionic liposomes rapidly degraded. These results indicate that the physicochemical properties and PEG modification of magnetic liposomes greatly influences their intestinal epithelial transport.
Collapse
Affiliation(s)
- Yusuke Kono
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University.,Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University
| | - Hitomi Jinzai
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Yota Kotera
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University.,Ritsumeikan-Global Innovation Research Organization, Ritsumeikan University.,Research Center for Drug Discovery and Development, Ritsumeikan University
| |
Collapse
|
34
|
Shen ZY, Liu C, Wu MF, Shi HF, Zhou YF, Zhuang W, Xia GL. Spiral computed tomography evaluation of rabbit VX2 hepatic tumors treated with 20 kHz ultrasound and microbubbles. Oncol Lett 2017; 14:3124-3130. [PMID: 28928850 DOI: 10.3892/ol.2017.6557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 05/11/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the therapeutic effect of 20 kHz ultrasound (US) and microbubbles (MBs) on rabbit VX2 liver tumors by spiral computed tomography (CT) scanning. A total of 16 New Zealand rabbits with hepatic VX2 tumors were divided into four groups: Control, MB, low-frequency US and US + MB. The treatment effect was evaluated by spiral CT scanning prior to, during and following treatment (at 0 weeks and the end of 1 and 2 weeks). The tumor growth rate was recorded. The specimens of VX2 tumors were collected for histological examination and transmission electron microscopy (TEM). No significant differences were observed between tumor areas measured by CT and pathology after 2-week treatment (P>0.05). The mean tumor growth rates in the control, MB, US and US + MB groups after 2 weeks of treatment were 385±21, 353±12, 302±14 and 154±9%, respectively (P<0.05, US + MB vs. the other three groups). Hematoxylin and eosin staining in the US + MB group revealed coagulation necrosis, interstitial hemorrhage and intravascular thrombosis. In the control, MB and US groups, tumor cells exhibited clear nuclear hyperchromatism. TEM of US + MB revealed vascular endothelial cell wall rupture, widened endothelial cell gaps, interstitial erythrocyte leakage and microvascular thrombosis, while intact vascular endothelial cells and normal erythrocytes in the tumor vessels were observed in the control, MB and US groups. A combination of 20 kHz US and MBs may effectively inhibit rabbit VX2 tumors. Spiral CT scanning is an ideal method to evaluate the US treatment on rabbit tumors.
Collapse
Affiliation(s)
- Zhi-Yong Shen
- Department of Radiology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Chun Liu
- Laboratory Animal Center of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ming-Feng Wu
- Department of Radiology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Hai-Feng Shi
- Department of Radiology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Yu-Feng Zhou
- Department of Radiology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Wei Zhuang
- Department of Radiology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| | - Gan-Lin Xia
- Department of Radiology, Nantong Tumor Hospital, Nantong, Jiangsu 226361, P.R. China
| |
Collapse
|
35
|
Cao S, Zhou Q, Chen JL, Jiang N, Wang YJ, Deng Q, Hu B, Guo RQ. Enhanced effect of nuclear localization signal peptide during ultrasound‑targeted microbubble destruction‑mediated gene transfection. Mol Med Rep 2017; 16:565-572. [PMID: 28586046 PMCID: PMC5482142 DOI: 10.3892/mmr.2017.6661] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/03/2017] [Indexed: 11/06/2022] Open
Abstract
Ultrasound‑targeted microbubble destruction (UTMD) can promote the entry of plasmid DNA (pDNA) into the cell cytoplasm, by increasing the permeability of the cell membrane. But the transfection efficiency remains low due to inability of the pDNA to enter the nucleus. Various methods have been explored to improve the UTMD transfection efficiency, but with little success. In cells, the classic nuclear localization signal (cNLS) peptide is an amino acid sequence that signals proteins that are due for nuclear transport. The present study aimed to investigate whether binding of a cNLS peptide to the pDNA may improve the transfection efficiency of UTMD. Four experimental groups were analyzed: Control group (UTMD + pDNA), group with cNLS (UTMD + pDNA + cNLS), group with mutated NLS (mNLS; UTMD + pDNA + mNLS), and group with cNLS and the nuclear import blocker, wheat germ agglutinin (WGA; UTMD + pDNA + cNLS + WGA). The NLS was labeled by fluorescein isothiocyanate, whereas pDNA was labeled with Cy3. Different molar ratios were tested for the NLS and pDNA combination in order to achieve optimal binding of the two molecules. Human umbilical vein endothelial cells were then transfected using the optimum ultrasonic irradiation parameters and NLS/pDNA molar ratio. At 6 h post‑transfection, the rates of Cy3‑labeled pDNA inside the cells and their nuclei were detected by flow cytometry and laser confocal microscopy, and the cellular vs. nuclear uptake of pDNA was calculated. In order to further evaluate the effect of NLS on UTMD‑mediated gene transfection, the transfection efficiency and relative expression levels of mRNA and protein were detected at 48 h post‑transfection. The results demonstrated that the optimal molar ratio of NLS with pDNA was 104:1. The rates of pDNA successful entry into the cell and nucleus were significantly higher in the cNLS group compared with the control group. The transfection efficiency, and relative expression levels of mRNA and protein from the plasmid were significantly increased in the cNLS group compared with the control group. The mNLS group displayed no significant difference compared with the control group, while the WGA group exhibited significant inhibition in most indicators of transfection efficiency compared to the cNLS group. These results suggest that combining a cNLS peptide with pDNA during UTMD‑mediated transfection significantly improved transfection efficiency. Thus, a cNLS peptide may be an important mediator and a new strategy in enhancing the efficiency of UTMD‑mediated gene transfection.
Collapse
Affiliation(s)
- Sheng Cao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jin-Ling Chen
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Nan Jiang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yi-Jia Wang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rui-Qiang Guo
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
36
|
Lee JY, Crake C, Teo B, Carugo D, de Saint Victor M, Seth A, Stride E. Ultrasound-Enhanced siRNA Delivery Using Magnetic Nanoparticle-Loaded Chitosan-Deoxycholic Acid Nanodroplets. Adv Healthc Mater 2017; 6. [PMID: 28195673 DOI: 10.1002/adhm.201601246] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/07/2017] [Indexed: 12/14/2022]
Abstract
Small interfering RNA (siRNA) has significant therapeutic potential but its clinical translation has been severely inhibited by a lack of effective delivery strategies. Previous work has demonstrated that perfluorocarbon nanodroplets loaded with magnetic nanoparticles can facilitate the intracellular delivery of a conventional chemotherapeutic drug. The aim of this study is to determine whether a similar agent can provide a means of delivering siRNA, enabling efficient transfection without degradation of the molecule. Chitosan-deoxycholic acid nanoparticles containing perfluoropentane and iron oxide (d 0 = 7.5 ± 0.35 nm) with a mean hydrodynamic diameter of 257.6 ± 10.9 nm are produced. siRNA (AllStars Hs cell death siRNA) is electrostatically bound to the particle surface and delivery to lung cancer cells and breast cancer cells is investigated with and without ultrasound exposure (500 kHz, 1 MPa peak-to-peak focal pressure, 40 cycles per burst, 1 kHz pulse repetition frequency, 10 s duration). The results show that siRNA functionality is not impaired by the treatment protocol and that the nanodroplets are able to successfully promote siRNA uptake, leading to significant apoptosis (52.4%) 72 h after ultrasound treatment.
Collapse
Affiliation(s)
- Jeong Yu Lee
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Calum Crake
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Boon Teo
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Dario Carugo
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
- Faculty of Engineering and the Environment; Southampton University; Southampton SO17 1BJ UK
| | - Marie de Saint Victor
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Anjali Seth
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| | - Eleanor Stride
- Institute of Biomedical Engineering; Department of Engineering Science; University of Oxford; Oxford, Old Road Campus OX3 7DQ UK
| |
Collapse
|
37
|
Hayashi K, Iwai H, Kamei T, Iwamoto K, Shimanouchi T, Fujita S, Nakamura H, Umakoshi H. Tailor-made drug carrier: Comparison of formation-dependent physicochemical properties within self-assembled aggregates for an optimal drug carrier. Colloids Surf B Biointerfaces 2017; 152:269-276. [DOI: 10.1016/j.colsurfb.2017.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/30/2016] [Accepted: 01/07/2017] [Indexed: 10/20/2022]
|
38
|
Anti-microRNA-21/221 and microRNA-199a transfected by ultrasound microbubbles induces the apoptosis of human hepatoma HepG2 cells. Oncol Lett 2017; 13:3669-3675. [PMID: 28529584 PMCID: PMC5431703 DOI: 10.3892/ol.2017.5910] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/07/2017] [Indexed: 12/23/2022] Open
Abstract
Gene therapy, particularly microRNA (miRNA), is a promising candidate in the treatment of cancer; however, it is challenging to develop gene delivery systems. Ultrasound microbubbles have been used for gene delivery with excellent results. The present study aimed to investigate the transfection efficiency of HepG2 cells using ultrasound microbubbles. The effects of three miRNAs (miR-21, miR-221 and miR-199a) on HepG2 cells were also determined by performing ultrasound microbubble-mediated gene transfection. Three recombinant plasmids containing anti-miR-21, anti-miR-221 and miR-199a were fused with enhanced green fluorescent protein. For the transfection of genes, the type of contrast agent, the concentration of microbubble contrast agent and the exposure intensity of ultrasound were optimized. The expression of miRNAs was detected using reverse transcription-polymerase chain reaction. To determine the effect of anti-miR-21, anti-miR-221 and miR-199a on HepG2 cells, MTT, cell cycle analysis and Annexin V-PE/7-ADD apoptosis assays were performed. The optimal condition was 10% sulfur hexafluoride microbubbles at an ultrasound frequency of 2.0 MHz and mechanical index of 0.28. When cells were transfected with three recombinant plasmids using ultrasound microbubbles, there was significant downregulation of miR-21 and miR-221 and upregulation of miR-199a (P<0.05). All three treatments inhibited cell proliferation and promoted the apoptosis of cells. The present data indicated that the delivery of anti-miR-21, anti-miR-221 and miR-199a may be mediated by ultrasound microbubble contrast agents. With this approach, cell proliferation may be effectively inhibited and cell apoptosis may be induced. These are novel cancer therapy targets.
Collapse
|
39
|
Li Y, Gao J, Zhang C, Cao Z, Cheng D, Liu J, Shuai X. Stimuli-Responsive Polymeric Nanocarriers for Efficient Gene Delivery. Top Curr Chem (Cham) 2017; 375:27. [DOI: 10.1007/s41061-017-0119-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/31/2017] [Indexed: 11/25/2022]
|
40
|
Tosi U, Marnell CS, Chang R, Cho WC, Ting R, Maachani UB, Souweidane MM. Advances in Molecular Imaging of Locally Delivered Targeted Therapeutics for Central Nervous System Tumors. Int J Mol Sci 2017; 18:ijms18020351. [PMID: 28208698 PMCID: PMC5343886 DOI: 10.3390/ijms18020351] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/19/2016] [Accepted: 01/26/2017] [Indexed: 12/24/2022] Open
Abstract
Thanks to the recent advances in the development of chemotherapeutics, the morbidity and mortality of many cancers has decreased significantly. However, compared to oncology in general, the field of neuro-oncology has lagged behind. While new molecularly targeted chemotherapeutics have emerged, the impermeability of the blood–brain barrier (BBB) renders systemic delivery of these clinical agents suboptimal. To circumvent the BBB, novel routes of administration are being applied in the clinic, ranging from intra-arterial infusion and direct infusion into the target tissue (convection enhanced delivery (CED)) to the use of focused ultrasound to temporarily disrupt the BBB. However, the current system depends on a “wait-and-see” approach, whereby drug delivery is deemed successful only when a specific clinical outcome is observed. The shortcomings of this approach are evident, as a failed delivery that needs immediate refinement cannot be observed and corrected. In response to this problem, new theranostic agents, compounds with both imaging and therapeutic potential, are being developed, paving the way for improved and monitored delivery to central nervous system (CNS) malignancies. In this review, we focus on the advances and the challenges to improve early cancer detection, selection of targeted therapy, and evaluation of therapeutic efficacy, brought forth by the development of these new agents.
Collapse
Affiliation(s)
- Umberto Tosi
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| | - Christopher S Marnell
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| | - Raymond Chang
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China.
| | - Richard Ting
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Uday B Maachani
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
41
|
Bui L, Aleid A, Alassaf A, Wilson OC, Raub CB, Frenkel V. Development of a custom biological scaffold for investigating ultrasound-mediated intracellular delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:461-470. [DOI: 10.1016/j.msec.2016.09.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/08/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023]
|
42
|
Xu T, Nie Y, Bai J, Li L, Yang B, Zheng G, Zhang J, Yu J, Cheng X, Jiao J, Jing H. Suppression of human 8-oxoguanine DNA glycosylase (OGG1) augments ultrasound-induced apoptosis in cervical cancer cells. ULTRASONICS 2016; 72:1-14. [PMID: 27447800 DOI: 10.1016/j.ultras.2016.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 07/07/2016] [Accepted: 07/10/2016] [Indexed: 06/06/2023]
Abstract
PURPOSE Human 8-oxoguanine DNA glycosylase (OGG1) is a major base excision repair enzyme, and it was reported to suppress the activation of intrinsic apoptotic signaling pathway in response to oxidative stress. In this study, our aim was to investigate the effects of OGG1 downregulation on ultrasound-induced apoptosis in cervical cancer cells. METHODS OGG1 expression was silenced by shRNA in the cervical cancer SW756 and CaSki cells. Cell viability was evaluated by MTT assay after OGG1 knockdown following ultrasound treatment. Ultrasound-induced apoptosis was measured by Annexin V-FITC/propidium iodide. Intracellular reactive oxygen species (ROS) production and Ca(2+) concentration were detected using a fluorescent probe, 2',7'-dichlorofluorescin diacetate (DCFH-DA) and a green fluorescent dye fluo-4AM, respectively. Western blotting was used to analyze the expression of Bcl-2, Bax, cleaved caspase-3, and nuclear factor-κB p65 (NF-κB p65). RESULTS The results indicated that OGG1 knockdown did not suppress cell proliferation, but significantly augmented ultrasound-induced inhibitory effects on the cell viability, and increased ultrasound-induced early apoptosis and late apoptosis and necrosis in the SW756 and CaSki cells when exposure to ultrasound (1MHz) at 1.5W/cm(2) for 30 and 60s. OGG1 knockdown significantly increased intracellular ROS production and Ca(2+) concentration after incubation of 6, 24, and 48h post-ultrasound treatment. The downregulation of Bcl-2 protein and the upregulation of Bax, cleaved caspase-3, and NF-κB p65 protein levels were observed in the shRNA-OGG1 cells and mock-shRNA cells, but no significant change of these protein levels was found between of them. CONCLUSIONS These results indicate that downregulation of OGG1 expression can augment ultrasound-induced apoptosis in cervical cancer cells, which suggests that OGG1 suppression might provide a new insight for ultrasound-induced therapeutic effects on cervical cancer treatment.
Collapse
Affiliation(s)
- Tao Xu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Yongli Nie
- Department of Oncology, Han Jiang Group Co. Ltd-Han Jiang Hospital, DanJiangKou 442700, Hubei Province, China
| | - Jiao Bai
- Department of Ultrasound, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Linjun Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Bo Yang
- Department of Ultrasound, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Guangmei Zheng
- Department of Ultrasound, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Jun Zhang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Jianyun Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Xiongfei Cheng
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Jiao Jiao
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Hongxia Jing
- Department of Ultrasound, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China.
| |
Collapse
|
43
|
Yu MH, Lee JY, Kim HR, Kim BR, Park EJ, Kim HS, Han JK, Choi BI. Therapeutic Effects of Microbubbles Added to Combined High-Intensity Focused Ultrasound and Chemotherapy in a Pancreatic Cancer Xenograft Model. Korean J Radiol 2016; 17:779-88. [PMID: 27587968 PMCID: PMC5007406 DOI: 10.3348/kjr.2016.17.5.779] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/15/2016] [Indexed: 12/21/2022] Open
Abstract
Objective To investigate whether high-intensity focused ultrasound (HIFU) combined with microbubbles enhances the therapeutic effects of chemotherapy. Materials and Methods A pancreatic cancer xenograft model was established using BALB/c nude mice and luciferase-expressing human pancreatic cancer cells. Mice were randomly assigned to five groups according to treatment: control (n = 10), gemcitabine alone (GEM; n = 12), HIFU with microbubbles (HIFU + MB, n = 11), combined HIFU and gemcitabine (HIGEM; n = 12), and HIGEM + MB (n = 13). After three weekly treatments, apoptosis rates were evaluated using the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay in two mice per group. Tumor volume and bioluminescence were monitored using high-resolution 3D ultrasound imaging and in vivo bioluminescence imaging for eight weeks in the remaining mice. Results The HIGEM + MB group showed significantly higher apoptosis rates than the other groups (p < 0.05) and exhibited the slowest tumor growth. From week 5, the tumor-volume-ratio relative to the baseline tumor volume was significantly lower in the HIGEM + MB group than in the control, GEM, and HIFU + MB groups (p < 0.05). Despite visible distinction, the HIGEM and HIGEM + MB groups showed no significant differences. Conclusion High-intensity focused ultrasound combined with microbubbles enhances the therapeutic effects of gemcitabine chemotherapy in a pancreatic cancer xenograft model.
Collapse
Affiliation(s)
- Mi Hye Yu
- Department of Radiology, Konkuk University Medical Center, Seoul 05030, Korea
| | - Jae Young Lee
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea
| | - Hae Ri Kim
- Department of Pre-Dentistry, Gangneung-Wonju National University College of Dentistry, Gangneung 25457, Korea
| | - Bo Ram Kim
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea
| | - Eun-Joo Park
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea
| | - Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea
| | - Joon Koo Han
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Korea
| | - Byung Ihn Choi
- Department of Radiology, Chung-Ang University Hospital, Seoul 06973, Korea
| |
Collapse
|
44
|
Xie X, Lin W, Li M, Yang Y, Deng J, Liu H, Chen Y, Fu X, Liu H, Yang Y. Efficient siRNA Delivery Using Novel Cell-Penetrating Peptide-siRNA Conjugate-Loaded Nanobubbles and Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1362-1374. [PMID: 27012462 DOI: 10.1016/j.ultrasmedbio.2016.01.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/13/2016] [Accepted: 01/27/2016] [Indexed: 06/05/2023]
Abstract
Because of the absence of tolerable and effective carriers for in vivo delivery, the applications of small interfering RNA (siRNA) in the clinic for therapeutic purposes have been limited. In this study, development of a novel siRNA delivery system based on ultrasound-sensitive nanobubbles (NBs, nano-sized echogenic liposomes) and cell-permeable peptides (CPPs) is described. A CPP-siRNA conjugate was entrapped in an NB, (CPP-siRNA)-NB, and the penetration of CPP-siRNA was temporally masked; local ultrasound stimulation triggered the release of CPP-siRNA from the NBs and activated its penetration. Subsequent research revealed that the (CPP-siRNA)-NBs had a mean particle size of 201 ± 2.05 nm and a siRNA entrapment efficiency >85%. In vitro release results indicated that >90% of the encapsulated CPP-siRNA was released from NBs in the presence of ultrasound, whereas <1.5% (30 min) was released in the absence of ultrasound. Cell experiments indicated higher cellular CPP-siRNA uptake of (CPP-siRNA)-NBs with ultrasound among the various formulations in human breast adenocarcinoma cells (HT-1080). Additionally, after systemic administration in mice, (CPP-siRNA)-NBs accumulated in the tumor, augmented c-myc silencing and delayed tumor progression. In conclusion, the application of (CPP-siRNA)-NBs with ultrasound may constitute an approach to selective targeted delivery of siRNA.
Collapse
Affiliation(s)
- Xiangyang Xie
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Wen Lin
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, China
| | - Mingyuan Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.
| | - Jianping Deng
- Department of Clinical Laboratory, Huangshi Love & Health Hospital of Hubei Province, Huangshi, China
| | - Hui Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Ying Chen
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Xudong Fu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Hong Liu
- Department of Pharmacy, Wuhan General Hospital of Guangzhou Military Command, Wuhan, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
45
|
Liu Y, Yan J, Santangelo PJ, Prausnitz MR. DNA uptake, intracellular trafficking and gene transfection after ultrasound exposure. J Control Release 2016; 234:1-9. [PMID: 27165808 DOI: 10.1016/j.jconrel.2016.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/26/2016] [Accepted: 05/06/2016] [Indexed: 11/17/2022]
Abstract
Ultrasound has been studied as a promising tool for intracellular gene delivery. In this work, we studied gene transfection of a human prostate cancer cell line exposed to megahertz pulsed ultrasound in the presence of contrast agent and assessed the efficiency of fluorescently labelled DNA delivery into cell nuclei, which is necessary for gene transfection. At the sonication conditions studied, ~30% of cells showed DNA uptake 30min after sonication, but that fraction decreased over time to ~10% of cells after 24h. Most cells containing DNA had DNA in their nuclei, but the amount varied significantly. Transfection efficiency peaked at ~10% at 8h post sonication. Among those cells containing DNA, ~30% of DNA was localized in the cell nuclei, ~30% was in autophagosomes/autophagolysosomes and the remainder was "free" in the cytoplasm 30min after sonication. At later times up to 24h, ~30% of DNA continued to be found in the nuclei and most or all of the rest of the DNA was in autophagosomes/autophagolysosomes. These results demonstrate that ultrasound can deliver DNA into cell nuclei shortly after sonication and that the rest of the DNA can be cleared by autophagosomes/autophagolysosomes.
Collapse
Affiliation(s)
- Ying Liu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | - Jing Yan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA; Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
46
|
Chen F, Li Y, Feng Y, He X, Wang L. Evaluation of Antimetastatic Effect of lncRNA-ATB siRNA Delivered Using Ultrasound-Targeted Microbubble Destruction. DNA Cell Biol 2016; 35:393-7. [PMID: 27027475 DOI: 10.1089/dna.2016.3254] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common human malignancies around the world. The poor prognosis and high recurrence rate of HCC are largely the result of the high frequencies of intrahepatic and extrahepatic metastases. However, the treatment of metastasis is very limited. Ultrasound-targeted microbubble destruction (UTMD) technology has been recognized as a promising technology for drug and gene delivery in vivo and in vitro. Long noncoding RNA activated by transforming growth factor-β (TGF-β; lncRNA-ATB) was recently identified, which was upregulated in HCC metastases and associated with poor prognosis of HCC patients. In this study, we used microbubbles for UTMD-mediated siRNA transfection to silence lncRNA-ATB expression. We found that UTMD-mediated siRNA transfection significantly inhibited lncRNA-ATB expression and ZEB1 and ZEB2 expression and suppressed cell migration and invasion. We also demonstrated that transfecting siRNA against lncRNA-ATB by using UTMD was more efficient than that by using lipidosome. UTMD-mediated siRNA transfection against lncRNA-ATB may be a promising therapy for HCC metastasis.
Collapse
Affiliation(s)
- Fei Chen
- 1 Department of Ultrasound, The First Affiliated Hospital of Liaoning Medical University , Jinzhou, China
| | - Yuhong Li
- 1 Department of Ultrasound, The First Affiliated Hospital of Liaoning Medical University , Jinzhou, China
| | - Yanhong Feng
- 1 Department of Ultrasound, The First Affiliated Hospital of Liaoning Medical University , Jinzhou, China
| | - Xiuli He
- 1 Department of Ultrasound, The First Affiliated Hospital of Liaoning Medical University , Jinzhou, China
| | - Liang Wang
- 2 Department of Hepatobiliary Surgery, The First Affiliated Hospital of Liaoning Medical University , Jinzhou, China
| |
Collapse
|
47
|
Zimmerman KA, Xing D, Pallero MA, Lu A, Ikawa M, Black L, Hoyt KL, Kabarowski JH, Michalak M, Murphy-Ullrich JE. Calreticulin Regulates Neointima Formation and Collagen Deposition following Carotid Artery Ligation. J Vasc Res 2016; 52:306-20. [PMID: 26910059 DOI: 10.1159/000443884] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 01/07/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The endoplasmic reticulum (ER) stress protein, calreticulin (CRT), is required for the production of TGF-β-stimulated extracellular matrix (ECM) by fibroblasts. Since TGF-β regulates vascular fibroproliferative responses and collagen deposition, we investigated the effects of CRT knockdown on vascular smooth-muscle cell (VSMC) fibroproliferative responses and collagen deposition. METHODS Using a carotid artery ligation model of vascular injury, Cre-recombinase-IRES-GFP plasmid was delivered with microbubbles (MB) to CRT-floxed mice using ultrasound (US) to specifically reduce CRT expression in the carotid artery. RESULTS In vitro, Cre-recombinase-mediated CRT knockdown in isolated, floxed VSMCs decreased the CRT transcript and protein, and attenuated the induction of collagen I protein in response to TGF-β. TGF-β stimulation of collagen I was partly blocked by the NFAT inhibitor 11R-VIVIT. Following carotid artery ligation, CRT staining was upregulated with enhanced expression in the neointima 14-21 days after injury. Furthermore, Cre-recombinase-IRES-GFP plasmid delivered by targeted US reduced CRT expression in the neointima of CRT-floxed mice and led to a significant reduction in neointima formation and collagen deposition. The neointimal cell number was also reduced in mice, with a local, tissue-specific knockdown of CRT. CONCLUSIONS This work establishes a novel role for CRT in mediating VSMC responses to injury through the regulation of collagen deposition and neointima formation.
Collapse
Affiliation(s)
- Kurt A Zimmerman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Ala., USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
In recent decades ultrasound-guided delivery of drugs loaded on nanocarriers has been the focus of increasing attention to improve therapeutic treatments. Ultrasound has often been used in combination with microbubbles, micron-sized spherical gas-filled structures stabilized by a shell, to amplify the biophysical effects of the ultrasonic field. Nanometer size bubbles are defined nanobubbles. They were designed to obtain more efficient drug delivery systems. Indeed, their small sizes allow extravasation from blood vessels into surrounding tissues and ultrasound-targeted site-specific release with minimal invasiveness. Additionally, nanobubbles might be endowed with improved stability and longer residence time in systemic circulation. This review will describe the physico-chemical properties of nanobubbles, the formulation parameters and the drug loading approaches, besides potential applications as a therapeutic tool.
Collapse
|
49
|
Development of tissue-selective gene delivery system with ultrasound. Asian J Pharm Sci 2016. [DOI: 10.1016/j.ajps.2015.11.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
50
|
Parameters affecting intracellular delivery of molecules using laser-activated carbon nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1003-1011. [PMID: 26772422 DOI: 10.1016/j.nano.2015.12.380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/02/2015] [Accepted: 12/24/2015] [Indexed: 11/20/2022]
Abstract
UNLABELLED Previous studies showed that carbon nanoparticles exposed to nanosecond laser pulses cause intracellular uptake of molecules. In this study, prostate cancer cells incubated with carbon-black (CB) nanoparticles and fluorescent marker compounds were exposed to 10ns laser pulses at 1064nm wavelength, after which intracellular uptake was measured by flow cytometry. Calcein and dextran (150kDa) were delivered into >50% of cells, whereas larger dextrans (≤2000kDa) were taken up by ~10% of cells. Under all conditions studied, cell viability loss was minimal. Uptake also increased with increasing laser power, increasing CB nanoparticle concentration, increasing CB nanoparticle size and decreasing laser wavelength. CB nanoparticles enabled uptake better than gold nanoparticles or multi-walled carbon nanotubes under the conditions studied. Proof-of-principle experiments showed intracellular uptake by cells in vivo. We conclude that intracellular uptake of molecules using laser-activated CB nanoparticles provides a promising approach to deliver molecules into cells. FROM THE CLINICAL EDITOR Delivery of drugs using nanoparticles as carriers is promising. The authors in this study investigated the use of laser-activated carbon nanoparticles to increase the cellular uptake of payloads in various parameters. The positive data generated should provide further platform for a new approach for intracellular delivery of molecules.
Collapse
|