1
|
Eş I, Thakur A, Mousavi Khaneghah A, Foged C, de la Torre LG. Engineering aspects of lipid-based delivery systems: In vivo gene delivery, safety criteria, and translation strategies. Biotechnol Adv 2024; 72:108342. [PMID: 38518964 DOI: 10.1016/j.biotechadv.2024.108342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024]
Abstract
Defects in the genome cause genetic diseases and can be treated with gene therapy. Due to the limitations encountered in gene delivery, lipid-based supramolecular colloidal materials have emerged as promising gene carrier systems. In their non-functionalized form, lipid nanoparticles often demonstrate lower transgene expression efficiency, leading to suboptimal therapeutic outcomes, specifically through reduced percentages of cells expressing the transgene. Due to chemically active substituents, the engineering of delivery systems for genetic drugs with specific chemical ligands steps forward as an innovative strategy to tackle the drawbacks and enhance their therapeutic efficacy. Despite intense investigations into functionalization strategies, the clinical outcome of such therapies still needs to be improved. Here, we highlight and comprehensively review engineering aspects for functionalizing lipid-based delivery systems and their therapeutic efficacy for developing novel genetic cargoes to provide a full snapshot of the translation from the bench to the clinics. We outline existing challenges in the delivery and internalization processes and narrate recent advances in the functionalization of lipid-based delivery systems for nucleic acids to enhance their therapeutic efficacy and safety. Moreover, we address clinical trials using these vectors to expand their clinical use and principal safety concerns.
Collapse
Affiliation(s)
- Ismail Eş
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Headington, Oxford OX3 7DQ, UK.
| | - Aneesh Thakur
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Amin Mousavi Khaneghah
- Faculty of Biotechnologies (BioTech), ITMO University 191002, 9 Lomonosova Street, Saint Petersburg, Russia.
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Lucimara Gaziola de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
2
|
Kagami S, Kanagawa T. Weakly nonlinear focused ultrasound in viscoelastic media containing multiple bubbles. ULTRASONICS SONOCHEMISTRY 2023; 97:106455. [PMID: 37271029 PMCID: PMC10248557 DOI: 10.1016/j.ultsonch.2023.106455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/12/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023]
Abstract
To facilitate practical medical applications such as cancer treatment utilizing focused ultrasound and bubbles, a mathematical model that can describe the soft viscoelasticity of human body, the nonlinear propagation of focused ultrasound, and the nonlinear oscillations of multiple bubbles is theoretically derived and numerically solved. The Zener viscoelastic model and Keller-Miksis bubble equation, which have been used for analyses of single or few bubbles in viscoelastic liquid, are used to model the liquid containing multiple bubbles. From the theoretical analysis based on the perturbation expansion with the multiple-scales method, the Khokhlov-Zabolotskaya-Kuznetsov (KZK) equation, which has been used as a mathematical model of weakly nonlinear propagation in single phase liquid, is extended to viscoelastic liquid containing multiple bubbles. The results show that liquid elasticity decreases the magnitudes of the nonlinearity, dissipation, and dispersion of ultrasound and increases the phase velocity of the ultrasound and linear natural frequency of the bubble oscillation. From the numerical calculation of resultant KZK equation, the spatial distribution of the liquid pressure fluctuation for the focused ultrasound is obtained for cases in which the liquid is water or liver tissue. In addition, frequency analysis is carried out using the fast Fourier transform, and the generation of higher harmonic components is compared for water and liver tissue. The elasticity suppresses the generation of higher harmonic components and promotes the remnant of the fundamental frequency components. This indicates that the elasticity of liquid suppresses shock wave formation in practical applications.
Collapse
Affiliation(s)
- Shunsuke Kagami
- Department of Engineering Mechanics and Energy, Degree Program of Systems and Information Engineering, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Japan
| | - Tetsuya Kanagawa
- Department of Engineering Mechanics and Energy, Degree Program of Systems and Information Engineering, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Japan
| |
Collapse
|
3
|
Goncin U, Curiel L, Geyer CR, Machtaler S. Aptamer-Functionalized Microbubbles Targeted to P-selectin for Ultrasound Molecular Imaging of Murine Bowel Inflammation. Mol Imaging Biol 2023; 25:283-293. [PMID: 35851673 DOI: 10.1007/s11307-022-01755-9] [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: 03/16/2022] [Revised: 06/01/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Our objectives were to develop a targeted microbubble with an anti-P-selectin aptamer and assess its ability to detect bowel inflammation in two murine models of acute colitis. PROCEDURES Lipid-shelled microbubbles were prepared using mechanical agitation. A rapid copper-free click chemistry approach (azide-DBCO) was used to conjugate the fluorescent anti-P-selectin aptamer (Fluor-P-Ap) to the microbubble surface. Bowel inflammation was chemically induced using 2,4,6-trinitrobenzenesulfonic acid (TNBS) in both Balb/C and interleukin-10-deficient (IL-10 KO) mice. Mouse bowels were imaged using non-linear contrast mode following an i.v. bolus of 1 × 108 microbubbles. Each mouse received a bolus of aptamer-functionalized and non-targeted microbubbles. Mouse phenotypes and the presence of P-selectin were validated using histology and immunostaining, respectively. RESULTS Microbubble labelling of Fluor-P-Ap was complete after 20 min at 37 ̊C. We estimate approximately 300,000 Fluor-P-Ap per microbubble and confirmed fluorescence using confocal microscopy. There was a significant increase in ultrasound molecular imaging signal from both Balb/C (p = 0.003) and IL-10 KO (p = 0.02) mice with inflamed bowels using aptamer-functionalized microbubbles in comparison to non-targeted microbubbles. There was no signal in healthy mice (p = 0.4051) using either microbubble. CONCLUSIONS We constructed an aptamer-functionalized microbubble specific for P-selectin using a clinically relevant azide-DBCO click reaction, which could detect bowel inflammation in vivo. Aptamers have potential as a next generation targeting agent for developing cost-efficient and clinically translatable targeted microbubbles.
Collapse
Affiliation(s)
- Una Goncin
- Department of Medical Imaging, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Laura Curiel
- Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, T2N 4V8, Canada
| | - C Ronald Geyer
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Steven Machtaler
- Department of Medical Imaging, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
4
|
Batchelor DB, Armistead FJ, Ingram N, Peyman SA, McLaughlan JR, Coletta PL, Evans SD. The Influence of Nanobubble Size and Stability on Ultrasound Enhanced Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:13943-13954. [PMID: 36322191 PMCID: PMC9671049 DOI: 10.1021/acs.langmuir.2c02303] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/17/2022] [Indexed: 06/16/2023]
Abstract
Lipid-shelled nanobubbles (NBs) are emerging as potential dual diagnostic and therapeutic agents. Similar to their micron-scale counterparts, microbubbles (1-10 μm), they can act as ultrasound contrast agents as well as locally enhance therapeutic uptake. Recently, it has been shown that the reduced size of NBs (<1 μm) promotes increased uptake and accumulation in tumor interstitial space, which can enhance their diagnostic and therapeutic performance. However, accurate characterization of NB size and concentration is challenging and may limit their translation into clinical use. Their submicron nature limits accuracy of conventional microscopy techniques, while common light scattering techniques fail to distinguish between subpopulations present in NB samples (i.e., bubbles and liposomes). Due to the difficulty in the characterization of NBs, relatively little is known about the influence of size on their therapeutic performance. In this study, we describe a novel method of using a commercially available nanoparticle tracking analysis system, to distinguish between NBs and liposomes based on their differing optical properties. We used this technique to characterize three NB populations of varying size, isolated via centrifugation, and subsequently used this to assess their potential for enhancing localized delivery. Confocal fluorescence microscopy and image analysis were used to quantify the ultrasound enhanced uptake of fluorescent dextran into live colorectal cancer cells. Our results showed that the amount of localized uptake did not follow the expected trends, in which larger NB populations out-perform smaller NBs, at matched concentration. To understand this observed behavior, the stability of each NB population was assessed. It was found that dilution of the NB samples from their stock concentration influences their stability, and it is hypothesized that both the total free lipid and interbubble distance play a role in NB lifetime, in agreement with previously proposed theories and models.
Collapse
Affiliation(s)
- Damien
V. B. Batchelor
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - Fern J. Armistead
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - Nicola Ingram
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, LeedsLS9 7TF, United Kingdom
- Faculty
of Electronic and Electrical Engineering, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - Sally A. Peyman
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - James R. McLaughlan
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, LeedsLS9 7TF, United Kingdom
- Faculty
of Electronic and Electrical Engineering, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, LeedsLS9 7TF, United Kingdom
| | - Stephen D. Evans
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| |
Collapse
|
5
|
Lin Y, Huang J, Chen Y, Wen Z, Cao Y, Zhang L, Cai T, Yu C, He X. Evaluation of perfluoropropane (C 3F 8)-filled chitosan polyacrylic acid nanobubbles for ultrasound imaging of sentinel lymph nodes and tumors. Biomater Sci 2022; 10:6447-6459. [PMID: 36018299 DOI: 10.1039/d2bm01140a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Accurate sentinel lymph node (SLN) identification is an important prerequisite for sentinel lymph node biopsy (SLNB). However, existing SLN mapping techniques, mainly imaging-guided methods, are severely restricted by the high cost of the instruments, harmful radiation or unsatisfactory imaging depths. Herein, we prepared a new ultrasound contrast agent by filling perfluoropropane (C3F8) into chitosan polyacrylic acid nanobubbles for precise SLN identification. The obtained ultrasound contrast agent, coined C3F8-CS-PAA nanobubbles, presents a nanometer size with a diameter of approximately 120 nm. The C3F8-CS-PAA nanobubbles of desirable size are able to enter lymphatic vessels and accumulate in the sentinel lymph node to enhance ultrasound imaging. As a result, the injection of C3F8-CS-PAA nanobubbles can remarkably enhance the ultrasound imaging lymph system, providing image guidance for sentinel lymph node biopsy. Furthermore, it was shown that such C3F8-CS-PAA nanobubbles can effectively permeate into the tumor region via the tumor-enhanced permeability and retention (EPR) effect to enhance tumor ultrasound imaging for monitoring tumorigenesis. This work highlights a novel nanoscale ultrasound contrast agent for the lymphatic system and tumor imaging, with great promise for subsequent studies and clinical applications.
Collapse
Affiliation(s)
- Yi Lin
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Ju Huang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yinyin Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering & Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400010, China
| | - Ziwei Wen
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Tao Cai
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chaoqun Yu
- College of Pharmacy, Chongqing Medical University, Chongqing 400010, China.
| | - Xuemei He
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
6
|
Kagami S, Kanagawa T. Weakly nonlinear propagation of focused ultrasound in bubbly liquids with a thermal effect: Derivation of two cases of Khokolov-Zabolotskaya-Kuznetsoz equations. ULTRASONICS SONOCHEMISTRY 2022; 88:105911. [PMID: 35810619 PMCID: PMC9696949 DOI: 10.1016/j.ultsonch.2022.105911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 05/06/2023]
Abstract
A physico-mathematical model composed of a single equation that consistently describes nonlinear focused ultrasound, bubble oscillations, and temperature fluctuations is theoretically proposed for microbubble-enhanced medical applications. The Khokhlov-Zabolotskaya-Kuznetsov (KZK) equation that has been widely used as a simplified model for nonlinear propagation of focused ultrasound in pure liquid is extended to that in liquid containing many spherical microbubbles, by applying the method of multiple scales to the volumetric averaged basic equations for bubbly liquids. As a result, for two-dimensional and three-dimensional cases, KZK equations composed of the linear combination of nonlinear, dissipation, dispersion, and focusing terms are derived. Especially, the dissipation term depends on three factors, i.e., interfacial liquid viscosity, liquid compressibility, and thermal conductivity of gas inside bubbles; the thermal conduction is evaluated by using four types of temperature gradient models. Finally, we numerically solve the derived KZK equation and show a moderate temperature rise appropriate to medical applications.
Collapse
Affiliation(s)
- Shunsuke Kagami
- Department of Engineering Mechanics and Energy, Graduate School of Systems and Information Engineering, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Japan
| | - Tetsuya Kanagawa
- Department of Engineering Mechanics and Energy, Faculty of Engineering, Information and Systems, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Japan.
| |
Collapse
|
7
|
|
8
|
Abstract
In recent years, minuscule gas bubbles called bulk nanobubbles (BNBs) have drawn increasing attention due to their unique properties and broad applicability in various technological fields, such as biomedical engineering, water treatment, and nanomaterials. However, questions remain regarding the stability and behavior of BNBs. In the present work, BNBs were generated in water using a gas-liquid mixing method. NB analysis was performed using a nanoparticle tracking analysis (NTA) method to investigate the coarsening behavior of BNBs in water over time. The diameters of the BNBs increased, and their cubic radii increased linearly (r3 ~ t) over time. While the concentration of BNBs decreased, the total volume of BNBs remained the same. The size distribution of the BNBs broadened, and the concentration of larger BNBs increased over time. These results indicate that relatively small BNBs disappeared due to dissolution and larger BNBs grew through mass transfer between BNBs instead of coalescence. In other words, BNBs underwent Ostwald ripening: gas molecules from smaller BNBs diffused through the continuous phase to be absorbed into larger BNBs.
Collapse
|
9
|
Yuan C, Li Y, Liu L, Tayier B, Yang L, Guan L, Mu Y. Experimental Study on the Compatibility and Characteristics of a Dual-Target Microbubble Loaded with Anti-miR-33. Int J Nanomedicine 2021; 16:6265-6280. [PMID: 34539179 PMCID: PMC8445104 DOI: 10.2147/ijn.s324514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/22/2021] [Indexed: 12/27/2022] Open
Abstract
Objective To prepare a new type of dual-target microbubble loaded with anti-miR-33 (ANM33). Methods Carrier core nanobubbles (NBs) were prepared by thin film hydration, and microbubbles loaded with PM1 (PCNBs) were prepared by grafting DSPE-PEG2000-maleimide-PM1 onto the NB surface. ANM33 was connected via electrostatic adsorption and covalent bonding, and hyaluronic acid (HA) was covalently connected. PM1 and HA were the targets, and ANM33 was the intervention drug. To evaluate the general physical and chemical properties of the prepared dual-target microbubbles loaded with ANM33 (HA-PANBs), we observed their morphology, particle size and surface potential while monitoring their stability and in vitro imaging ability, evaluated their toxic effect on cells and verified their ability to target cells. Results HA-PANBs had a regular morphology and good stability. The average particle size measured by a Malvern potentiometer was 1421.75±163.23 nm, and the average surface potential was −5.51±1.87 mV. PM1 and ANM33 were effectively connected to the NBs. The PM1, ANM33, and HA binding reached 89.0±1.1%, 65.02±5.0%, and 61.4±3.5%, respectively, and the maximum binding reached 2 µg, 5 µg, and 7 µg/108 microbubbles, respectively. HA-PANBs had no obvious toxic effects on cells, and their ability to continuously enhance imaging in vitro persisted for more than 15 minutes, obviously targeting foam cells in the early stage of AS. Conclusion HA-PANBs are ideal ultrasound contrast agents. The successful, firm connection of PM1 and HA to the NBs significantly increased the amount of carried ANM33. When microbubbles prepared with 2:4:7 PM1:ANM33:HA were used as a contrast agent, they had a high ANM33 carrying capacity, stable physical properties, and significantly enhanced imaging and targeting of foam cells in the early stage of AS.
Collapse
Affiliation(s)
- Chen Yuan
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Yanhong Li
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Liyun Liu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Baihetiya Tayier
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Lingjie Yang
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Lina Guan
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Yuming Mu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| |
Collapse
|
10
|
Exner AA, Kolios MC. Bursting Microbubbles: How Nanobubble Contrast Agents Can Enable the Future of Medical Ultrasound Molecular Imaging and Image-Guided Therapy. Curr Opin Colloid Interface Sci 2021; 54:101463. [PMID: 34393610 PMCID: PMC8356903 DOI: 10.1016/j.cocis.2021.101463] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of medical ultrasound has undergone a significant evolution since the development of microbubbles as contrast agents. However, due to their size, microbubbles remain in the vasculature, and therefore have limited clinical applications. Building a better - and smaller - bubble can expand the applications of contrast-enhanced ultrasound by allowing bubbles to extravasate from blood vessels - creating new opportunities. In this review, we summarize recent research on the formulation and use of NBs as imaging agents and as therapeutic vehicles. We discuss the ongoing debates in the field and reluctance to accepting NBs as an acoustically active construct and a potentially impactful clinical tool that can help shape the future of medical ultrasound. We hope that the overview of key experimental and theoretical findings in the NB field presented in this paper provides a fundamental framework that will help clarify NB-ultrasound interactions and inspire engagement in the field.
Collapse
Affiliation(s)
- Agata A. Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
11
|
Batchelor DV, Armistead FJ, Ingram N, Peyman SA, Mclaughlan JR, Coletta PL, Evans SD. Nanobubbles for therapeutic delivery: Production, stability and current prospects. Curr Opin Colloid Interface Sci 2021. [DOI: 10.1016/j.cocis.2021.101456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Xu J, Salari A, Wang Y, He X, Kerr L, Darbandi A, de Leon AC, Exner AA, Kolios MC, Yuen D, Tsai SSH. Microfluidic Generation of Monodisperse Nanobubbles by Selective Gas Dissolution. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100345. [PMID: 33811441 DOI: 10.1002/smll.202100345] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/26/2021] [Indexed: 06/12/2023]
Abstract
Nanotechnology currently enables the fabrication of uniform solid nanoparticles and liquid nano-emulsions, but not uniform gaseous nanobubbles (NBs). In this article, for the first time, a method based on microfluidics that directly produces monodisperse NBs is reported. Specifically, a two-component gas mixture of water-soluble nitrogen and water-insoluble octafluoropropane as the gas phase are used in a microfluidic bubble generator. First, monodisperse microbubbles (MBs) with a classical microfluidic flow-focusing junction is generated, then the MBs shrink down to ≈100 nm diameter, due to the dissolution of the water-soluble components in the gas mixture. The degree of shrinkage is controlled by tuning the ratio of water-soluble to water-insoluble gas components. This technique maintains the monodispersity of the NBs, and enables precise control of the final NB size. It is found that the monodisperse NBs show better homogeneity than polydisperse NBs in in vitro ultrasound imaging experiments. Proof-of-concept in vivo kidney imaging is performed in live mice, demonstrating enhanced contrast using the monodisperse NBs. The NB monodispersity and imaging results make microfluidically generated NBs promising candidates as ultrasound contrast and molecular imaging agents.
Collapse
Affiliation(s)
- Jiang Xu
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Alinaghi Salari
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, Ontario, M5B 2K2, Canada
| | - Yanjie Wang
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Department of Physics, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Xiaolin He
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Division of Nephrology, Department of Medicine, Unity Health Toronto and University of Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Liam Kerr
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Ali Darbandi
- Nanoimaging Centre, The Hospital for Sick Children, Peter Gilgan Centre for Research & Learning, Toronto, Ontario, M5G 0A4, Canada
| | - Al C de Leon
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Michael C Kolios
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Department of Physics, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Darren Yuen
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Division of Nephrology, Department of Medicine, Unity Health Toronto and University of Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Scott S H Tsai
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| |
Collapse
|
13
|
Jafari Sojahrood A, de Leon AC, Lee R, Cooley M, Abenojar EC, Kolios MC, Exner AA. Toward Precisely Controllable Acoustic Response of Shell-Stabilized Nanobubbles: High Yield and Narrow Dispersity. ACS NANO 2021; 15:4901-4915. [PMID: 33683878 PMCID: PMC7992193 DOI: 10.1021/acsnano.0c09701] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Understanding the pressure dependence of the nonlinear behavior of ultrasonically excited phospholipid-stabilized nanobubbles (NBs) is important for optimizing ultrasound exposure parameters for implementations of contrast enhanced ultrasound, critical to molecular imaging. The viscoelastic properties of the shell can be controlled by the introduction of membrane additives, such as propylene glycol as a membrane softener or glycerol as a membrane stiffener. We report on the production of high-yield NBs with narrow dispersity and different shell properties. Through precise control over size and shell structure, we show how these shell components interact with the phospholipid membrane, change their structure, affect their viscoelastic properties, and consequently change their acoustic response. A two-photon microscopy technique through a polarity-sensitive fluorescent dye, C-laurdan, was utilized to gain insights on the effect of membrane additives to the membrane structure. We report how the shell stiffness of NBs affects the pressure threshold (Pt) for the sudden amplification in the scattered acoustic signal from NBs. For narrow size NBs with 200 nm mean size, we find Pt to be between 123 and 245 kPa for the NBs with the most flexible membrane as assessed using C-Laurdan, 465-588 kPa for the NBs with intermediate stiffness, and 588-710 kPa for the NBs with stiff membranes. Numerical simulations of the NB dynamics are in good agreement with the experimental observations, confirming the dependence of acoustic response to shell properties, thereby substantiating further the development in engineering the shell of ultrasound contrast agents. The viscoelastic-dependent threshold behavior can be utilized for significantly and selectively enhancing the diagnostic and therapeutic ultrasound applications of potent narrow size NBs.
Collapse
Affiliation(s)
- Amin Jafari Sojahrood
- Department
of Physics, Ryerson University, Toronto, Ontario M5B 2K3, Canada
- Institute
for Biomedical Engineering and Science Technology, A Partnership between Ryerson University and St. Michael’s
Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Al C. de Leon
- Department
of Radiology Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Richard Lee
- Light
Microscopy Imaging Core, Case Western Reserve
University, Cleveland, Ohio 44106, United
States
| | - Michaela Cooley
- Department
of Radiology Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Eric C. Abenojar
- Department
of Radiology Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Michael C. Kolios
- Department
of Physics, Ryerson University, Toronto, Ontario M5B 2K3, Canada
- Institute
for Biomedical Engineering and Science Technology, A Partnership between Ryerson University and St. Michael’s
Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Agata A. Exner
- Department
of Radiology Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
14
|
Dong W, Huang A, Huang J, Wu P, Guo S, Liu H, Qin M, Yang X, Zhang B, Wan M, Zong Y. Plasmid-loadable magnetic/ultrasound-responsive nanodroplets with a SPIO-NP dispersed perfluoropentane core and lipid shell for tumor-targeted intracellular plasmid delivery. Biomater Sci 2020; 8:5329-5345. [PMID: 32793943 DOI: 10.1039/d0bm00699h] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Using ultrasound activating contrast agents to induce sonoporation is a potential strategy for effective lesion-targeted gene delivery. Previous reports have proven that submicron nanodroplets have a better advantage than microbubbles in that they can pass through tumor vasculature endothelial gaps by passive targeting; however, they cannot achieve an adequate dose in tumors to facilitate ultrasound-enhanced gene delivery. Additionally, a few studies focused on delivering macromolecular genetic materials (i.e. overexpression plasmid and CRISPR plasmid) have presented more unique advantages than small-molecular genetic materials (i.e. miRNA mimics, siRNA and shRNA etc.), such as enhancing the expression of target genes with long-term effectiveness. Thereby, we constructed novel plasmid-loadable magnetic/ultrasound-responsive nanodroplets, where superparamagnetic iron oxide nanoparticle dispersed perfluoropentane was encapsulated with lipids to which plasmids could be adhered, and branched polyethylenimine was used to protect the plasmids from enzymolysis. Furthermore, in vitro and in vivo studies were performed to verify the magnetic tumor-targeting ability of the plasmid-loadable magnetic/ultrasound-responsive nanodroplets and focused ultrasound enhanced intracellular plasmid delivery. The plasmid-loadable magnetic/ultrasound-responsive nanodroplets, carrying 16-19 plasmids per droplet, had desirable diameters less than 300 nm, and integrated the merits of excellent magnetic targeting capabilities and phase transition sensitivity to focused ultrasound. Under programmable focused ultrasound exposure, the plasmid-loadable magnetic/ultrasound-responsive nanodroplets underwent a phase-transition into echogenic microbubbles and the subsequent inertial cavitation of the microbubbles achieved an ∼40% in vitro plasmid delivery efficiency. Following intravenous administration, T2-weighted magnet resonance imaging, scanning electron microscopy and inductively coupled plasma optical emission spectrometry of the tumors showed significantly enhanced intratumoral accumulation of the plasmid-loadable magnetic/ultrasound-responsive nanodroplets under an external magnetic field. And a GFP ELISA assay and immunofluorescence staining indicated that focused ultrasound-induced inertial cavitation of the plasmid-loadable magnetic/ultrasound-responsive nanodroplets significantly enhanced the intracellular delivery of plasmids within the tumor after magnet-assisted accumulation, while only lower GFP levels were observed in the tumors on applying focused ultrasound or an external magnet alone. Taken together, utilizing the excellent plasmid-loadable magnetic/ultrasound-responsive nanodroplets combined with magnetism and ultrasound could efficiently deliver plasmids to cancer cells, which could be a potential strategy for macromolecular genetic material delivery in the clinic to treat cancer.
Collapse
Affiliation(s)
- Wei Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Calzada V. Aptamers in Diagnostic and Molecular Imaging Applications. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2020; 174:141-160. [PMID: 31848635 DOI: 10.1007/10_2019_115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The origin of the term diagnostic comes from the Greek word gnosis, meaning "to know." In medicine, a diagnostic can predict the pathology risk, disease status, treatment, and prognosis, even following therapy. An early and correct diagnosis is necessary for an efficient treatment. Moreover, it is possible to predict if and why a therapy will be successful or fail, enabling the timely application of alternative therapeutic strategies. Available diagnostics are due to the advances in biotechnology; however, more sensitive, low-cost, and noninvasive methodologies are still a challenge. Knowledge about molecular characteristics provide personalized information, which is the goal of future medicine. Today, multiple diagnostic techniques have emerged, with which it is possible to distinguish molecular patterns.In this way, aptamers are the perfect tools to recognize molecular targets and can be easily modified to confer additional functions. Their versatile characteristics and low cost make aptamers ideal for diagnostic applications.This chapter is a review of aptamer-based diagnostics in biomedicine, with a special focus on probe design and molecular imaging. Graphical Abstract.
Collapse
Affiliation(s)
- Victoria Calzada
- Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
16
|
Zhang X, Wu M, Zhang Y, Zhang J, Su J, Yang C. Molecular imaging of atherosclerotic plaque with lipid nanobubbles as targeted ultrasound contrast agents. Colloids Surf B Biointerfaces 2020; 189:110861. [DOI: 10.1016/j.colsurfb.2020.110861] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/01/2020] [Accepted: 02/10/2020] [Indexed: 01/17/2023]
|
17
|
Dong W, Wu P, Zhou D, Huang J, Qin M, Yang X, Wan M, Zong Y. Ultrasound-Mediated Gene Therapy of Hepatocellular Carcinoma Using Pre-microRNA Plasmid-Loaded Nanodroplets. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:90-107. [PMID: 31668943 DOI: 10.1016/j.ultrasmedbio.2019.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 05/19/2023]
Abstract
The PIK3 CA gene encodes the p110α protein subunit and is one of the most efficient cancer genes in solid and hematological tumors including hepatocellular carcinoma (HCC). There are currently ongoing therapies against tumors based on PIK3 CA inhibition. Because microRNAs (miRNAs) play an important role in post-transcriptional regulation and are also involved in the inhibition of PIK3 CA expression to suppress cancer cell proliferation, overexpression of tumor-suppressive miRNA is a promising therapeutic approach for HCC therapy. The successful and localized delivery of miRNA overexpression vectors (pre-miRNA plasmids) is very important in improving the therapeutic efficacy of this miRNA therapy strategy. In the study described here, submicron acoustic phase-shifted nanodroplets were used to efficiently deliver pre-miRNA plasmid in vitro and in vivo for HCC therapy under focused ultrasound (US) activation. Briefly, six miRNAs, inhibiting PIK3 CA and downregulated in HCC, were selected through summary and analysis of the currently existing literature data. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blot and cell apoptosis assay revealed that pre-miR-139, -203a, -378a and -422a plasmids among the six miRNA overexpression vectors could suppress growth of the hepatoma cell line SMMC-7721. These four pre-miRNA plasmids were then electrostatically adhered to positively charged lipid-shelled nanodroplets to obtain plasmid-loaded nanodroplets (PLNDs). The PLND-generated microbubbles oscillated and even collapsed under US exposure to release the loaded pre-miRNA plasmids and enhance their cellular uptake through consequent sonoporation, that is, formation of small pores on the cell membrane induced by the mechanical effects of PLND cavitation. Fluorescence microscopy results revealed that PLNDs could effectively deliver the aforementioned four pre-miRNA plasmids into SMMC-7721 cells in vitro under 1.2-MHz 60-cycle sinusoid US exposure with a peak negative pressure >5.5 MPa at a 40-Hz pulse repetition frequency. Plasmid delivery efficiency and cell viability positively correlated with the inertial cavitation dose that was determined mainly by peak negative pressure. Furthermore, PLNDs combined with US were evaluated in vivo to deliver these four pre-miRNAs plasmids and verify their therapeutic efficacy in subcutaneous tumor of the mouse xenograft HCC model. The results revealed that the PLNDs loaded with pre-miR-139 and -378a plasmids could effectively suppress tumor growth after US treatment. Thus, combination of pre-miRNA PLNDs with US activation seems to constitute a potential strategy for HCC therapy.
Collapse
Affiliation(s)
- Wei Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China
| | - Pengying Wu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China
| | - Di Zhou
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China
| | - Jixiu Huang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China
| | - Mengfan Qin
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China
| | - Xinxing Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China; Department of Ultrasound, First Affiliated Hospital of AFMU (Xijing Hospital), Air Force Medical University, Xi' an, China
| | - Mingxi Wan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China
| | - Yujin Zong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi' an Jiaotong University, Xi' an, China.
| |
Collapse
|
18
|
van Ballegooie C, Man A, Win M, Yapp DT. Spatially Specific Liposomal Cancer Therapy Triggered by Clinical External Sources of Energy. Pharmaceutics 2019; 11:E125. [PMID: 30884786 PMCID: PMC6470770 DOI: 10.3390/pharmaceutics11030125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 11/25/2022] Open
Abstract
This review explores the use of energy sources, including ultrasound, magnetic fields, and external beam radiation, to trigger the delivery of drugs from liposomes in a tumor in a spatially-specific manner. Each section explores the mechanism(s) of drug release that can be achieved using liposomes in conjunction with the external trigger. Subsequently, the treatment's formulation factors are discussed, highlighting the parameters of both the therapy and the medical device. Additionally, the pre-clinical and clinical trials of each triggered release method are explored. Lastly, the advantages and disadvantages, as well as the feasibility and future outlook of each triggered release method, are discussed.
Collapse
Affiliation(s)
- Courtney van Ballegooie
- Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Alice Man
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Mi Win
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
| | - Donald T Yapp
- Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
19
|
Zhang J, Lan T, Lu Y. Molecular Engineering of Functional Nucleic Acid Nanomaterials toward In Vivo Applications. Adv Healthc Mater 2019; 8:e1801158. [PMID: 30725526 PMCID: PMC6426685 DOI: 10.1002/adhm.201801158] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/14/2019] [Indexed: 12/25/2022]
Abstract
Recent advances in nanotechnology and engineering have generated many nanomaterials with unique physical and chemical properties. Over the past decade, numerous nanomaterials are introduced into many research areas, such as sensors for environmental monitoring, food safety, point-of-care diagnostics, and as transducers for solar energy transfer. Meanwhile, functional nucleic acids (FNAs), including nucleic acid enzymes, aptamers, and aptazymes, have attracted major attention from the biomedical community due to their unique target recognition and catalytic properties. Benefiting from the recent progress of molecular engineering strategies, the physicochemical properties of nanomaterials are endowed by the target recognition and catalytic activity of FNAs in the presence of a target analyte, resulting in numerous smart nanoprobes for diverse applications including intracellular imaging, drug delivery, in vivo imaging, and tumor therapy. This progress report focuses on the recent advances in designing and engineering FNA-based nanomaterials, highlighting the functional outcomes toward in vivo applications. The challenges and opportunities for the future translation of FNA-based nanomaterials into clinical applications are also discussed.
Collapse
Affiliation(s)
- JingJing Zhang
- Department of Chemistry, University of Illinois at Urbana-Champaign, 601 S. Mathews Ave., Urbana, IL, 61801, USA
| | - Tian Lan
- GlucoSentient, Inc., 2100 S. Oak Street Suite 101, Champaign, IL, 61820, USA
| | - Yi Lu
- Department of Chemistry, University of Illinois at Urbana-Champaign, 601 S. Mathews Ave., Urbana, IL, 61801, USA
| |
Collapse
|
20
|
Khan MS, Hwang J, Lee K, Choi Y, Jang J, Kwon Y, Hong JW, Choi J. Surface Composition and Preparation Method for Oxygen Nanobubbles for Drug Delivery and Ultrasound Imaging Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E48. [PMID: 30609703 PMCID: PMC6358755 DOI: 10.3390/nano9010048] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/22/2018] [Accepted: 12/23/2018] [Indexed: 02/06/2023]
Abstract
Phospholipids have been widely investigated for the preparation of liposomes, and micro and nanobubbles. They comprise biocompatible and biodegradable molecules and offer simple preparation with a variety of functions in diagnostic and therapeutic applications. Phospholipids require emulsifiers and surfactants to assemble in the form of bubbles. These surfactants determine the size, zeta potential, and other characteristics of particles. Polyethylene glycol (PEG) and its various derivatives have been employed by researchers to synthesize micro and nanobubbles. The stability of phospholipid-shelled nanobubbles has been reported by various researchers owing to the reduction of surface tension by surfactants in the shell. Nanobubbles have been employed to deliver oxygen to tissues and hypoxic cells. In this study, we investigated the effects of different ratios of phospholipids to PEG on the size, distribution, and characterization of oxygen nanobubbles (ONBs). ONBs were synthesized using a sonication technique. We analyzed and compared the sizes, numbers of generated particles, and zeta potentials of different compositions of ONBs using dynamic light scattering and nanoparticle tracking analysis. Then, we employed these oxygen nanobubbles to enhance the cellular microenvironment and cell viability. ONBs were also investigated for ultrasound imaging.
Collapse
Affiliation(s)
- Muhammad Saad Khan
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Jangsun Hwang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Kyungwoo Lee
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Jaehee Jang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Yejin Kwon
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Jong Wook Hong
- Department of Bionano Technology, Hanyang University, Seoul, Korea.
- Department of Bionano Engineering, Hanyang University, Ansan, Korea.
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| |
Collapse
|
21
|
Lafond M, Watanabe A, Yoshizawa S, Umemura SI, Tachibana K. Cavitation-threshold Determination and Rheological-parameters Estimation of Albumin-stabilized Nanobubbles. Sci Rep 2018; 8:7472. [PMID: 29748624 PMCID: PMC5945894 DOI: 10.1038/s41598-018-25913-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/25/2018] [Indexed: 12/24/2022] Open
Abstract
Nanobubbles (NBs) are of high interest for ultrasound (US) imaging as contrast agents and therapy as cavitation nuclei. Because of their instability (Laplace pressure bubble catastrophe) and low sensitivity to US, reducing the size of commonly used microbubbles to submicron-size is not trivial. We introduce stabilized NBs in the 100-250-nm size range, manufactured by agitating human serum albumin and perfluoro-propane. These NBs were exposed to 3.34- and 5.39-MHz US, and their sensitivity to US was proven by detecting inertial cavitation. The cavitation-threshold information was used to run a numerical parametric study based on a modified Rayleigh-Plesset equation (with a Newtonian rheology model). The determined values of surface tension ranged from 0 N/m to 0.06 N/m. The corresponding values of dilatational viscosity ranged from 5.10-10 Ns/m to 1.10-9 Ns/m. These parameters were reported to be 0.6 N/m and 1.10-8 Ns/m for the reference microbubble contrast agent. This result suggests the possibility of using albumin as a stabilizer for the nanobubbles that could be maintained in circulation and presenting satisfying US sensitivity, even in the 3-5-MHz range.
Collapse
Affiliation(s)
- Maxime Lafond
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, 980-8579, Japan.
| | - Akiko Watanabe
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Shin Yoshizawa
- Graduate School of Engineering, Tohoku University, Sendai, Miyagi, 980-8579, Japan
| | - Shin-Ichiro Umemura
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, 980-8579, Japan
| | - Katsuro Tachibana
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|
22
|
Abstract
The phenomenal advances in pharmaceutical sciences over the last few decades have led to the development of new therapeutics like peptides, proteins, RNAs, DNAs and highly potent small molecules. Fruitful applications of these therapeutics have been challenged by several anatomical and physiological barriers that limit adequate drug disposition at the site-of-action and by off-target drug distribution to undesired tissues, which together result in the reduced effectiveness and increased side effects of therapeutic agents. As such, the development of drug delivery and targeting systems has been recognised as a cornerstone for future drug development. Research in pharmaceutical sciences is now devoted to tackling delivery challenges through engineering delivery systems that move beyond conventional dosage forms and regimens into state-of-the-art targeted drug delivery tailored toward specific therapeutic needs. Modern drug delivery systems comprise passive and active targeting approaches. While passive targeting relies on the natural course of distribution of drugs or drug carriers in the body, as governed by their physicochemical properties, active targeting often exploits targeting moieties that home preferentially into target tissues. Here, we provide an overview of theories of and approaches to passive and active drug delivery. As the design of drug delivery is dependent on the unique structure of target tissues and organs, we present our discussion in an organ-specific manner with the aim to inspire the development of new strategies for curing disease with high accuracy and efficiency.
Collapse
Affiliation(s)
- Mohammad Alsaggar
- a Department of Pharmaceutical Technology, College of Pharmacy , Jordon University of Science and Technology , Irbid , Jordan
| | - Dexi Liu
- b Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy , University of Georgia , Athens , GA , USA
| |
Collapse
|
23
|
Chandola C, Kalme S, Casteleijn MG, Urtti A, Neerathilingam M. Application of aptamers in diagnostics, drug-delivery and imaging. J Biosci 2017; 41:535-61. [PMID: 27581942 DOI: 10.1007/s12038-016-9632-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aptamers are small, single-stranded oligonucleotides (DNA or RNA) that bind to their target with high specificity and affinity. Although aptamers are analogous to antibodies for a wide range of target recognition and variety of applications, they have significant advantages over antibodies. Since aptamers have recently emerged as a class of biomolecules with an application in a wide array of fields, we need to summarize the latest developments herein. In this review we will discuss about the latest developments in using aptamers in diagnostics, drug delivery and imaging. We begin with diagnostics, discussing the application of aptamers for the detection of infective agents itself, antigens/ toxins (bacteria), biomarkers (cancer), or a combination. The ease of conjugation and labelling of aptamers makes them a potential tool for diagnostics. Also, due to the reduced off-target effects of aptamers, their use as a potential drug delivery tool is emerging rapidly. Hence, we discuss their use in targeted delivery in conjugation with siRNAs, nanoparticles, liposomes, drugs and antibodies. Finally, we discuss about the conjugation strategies applicable for RNA and DNA aptamers for imaging. Their stability and self-assembly after heating makes them superior over protein-based binding molecules in terms of labelling and conjugation strategies.
Collapse
Affiliation(s)
- Chetan Chandola
- 1Center for Cellular and Molecular Platforms, NCBS-TIFR, Bangalore 560 065, India
| | | | | | | | | |
Collapse
|
24
|
Tang H, Zheng Y, Chen Y. Materials Chemistry of Nanoultrasonic Biomedicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1604105. [PMID: 27991697 DOI: 10.1002/adma.201604105] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/12/2016] [Indexed: 06/06/2023]
Abstract
As a special cross-disciplinary research frontier, nanoultrasonic biomedicine refers to the design and synthesis of nanomaterials to solve some critical issues of ultrasound (US)-based biomedicine. The concept of nanoultrasonic biomedicine can also overcome the drawbacks of traditional microbubbles and promote the generation of novel US-based contrast agents or synergistic agents for US theranostics. Here, we discuss the recent developments of material chemistry in advancing the nanoultrasonic biomedicine for diverse US-based bio-applications. We initially introduce the design principles of novel nanoplatforms for serving the nanoultrasonic biomedicine, from the viewpoint of synthetic material chemistry. Based on these principles and diverse US-based bio-application backgrounds, the representative proof-of-concept paradigms on this topic are clarified in detail, including nanodroplet vaporization for intelligent/responsive US imaging, multifunctional nano-contrast agents for US-based multi-modality imaging, activatable synergistic agents for US-based therapy, US-triggered on-demand drug releasing, US-enhanced gene transfection, US-based synergistic therapy on combating the cancer and potential toxicity issue of screening various nanosystems suitable for nanoultrasonic biomedicine. It is highly expected that this novel nanoultrasonic biomedicine and corresponding high performance in US imaging and therapy can significantly promote the generation of new sub-discipline of US-based biomedicine by rationally integrating material chemistry and theranostic nanomedicine with clinical US-based biomedicine.
Collapse
Affiliation(s)
- Hailin Tang
- Department of Diagnostic Ultrasound, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, P. R. China
| | - Yuanyi Zheng
- Shanghai Institute of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated, Shanghai Sixth People's Hospital, Shanghai, 200233, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
25
|
Wang J, Mi P, Lin G, Wáng YXJ, Liu G, Chen X. Imaging-guided delivery of RNAi for anticancer treatment. Adv Drug Deliv Rev 2016; 104:44-60. [PMID: 26805788 DOI: 10.1016/j.addr.2016.01.008] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/27/2015] [Accepted: 01/13/2016] [Indexed: 12/12/2022]
Abstract
The RNA interference (RNAi) technique is a new modality for cancer therapy, and several candidates are being tested clinically. In the development of RNAi-based therapeutics, imaging methods can provide a visible and quantitative way to investigate the therapeutic effect at anatomical, cellular, and molecular level; to noninvasively trace the distribution; to and study the biological processes in preclinical and clinical stages. Their abilities are important not only for therapeutic optimization and evaluation but also for shortening of the time of drug development to market. Typically, imaging-functionalized RNAi therapeutics delivery that combines nanovehicles and imaging techniques to study and improve their biodistribution and accumulation in tumor site has been progressively integrated into anticancer drug discovery and development processes. This review presents an overview of the current status of translating the RNAi cancer therapeutics in the clinic, a brief description of the biological barriers in drug delivery, and the roles of imaging in aspects of administration route, systemic circulation, and cellular barriers for the clinical translation of RNAi cancer therapeutics, and with partial content for discussing the safety concerns. Finally, we focus on imaging-guided delivery of RNAi therapeutics in preclinical development, including the basic principles of different imaging modalities, and their advantages and limitations for biological imaging. With growing number of RNAi therapeutics entering the clinic, various imaging methods will play an important role in facilitating the translation of RNAi cancer therapeutics from bench to bedside.
Collapse
|
26
|
Yildirim A, Chattaraj R, Blum NT, Goodwin AP. Understanding Acoustic Cavitation Initiation by Porous Nanoparticles: Toward Nanoscale Agents for Ultrasound Imaging and Therapy. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2016; 28:5962-5972. [PMID: 28484307 PMCID: PMC5419691 DOI: 10.1021/acs.chemmater.6b02634] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Ultrasound is widely applied in medical diagnosis and therapy due to its safety, high penetration depth, and low cost. In order to improve the contrast of sonographs and efficiency of the ultrasound therapy, echogenic gas bodies or droplets (with diameters from 200 nm to 10 µm) are often used, which are not very stable in the bloodstream and unable to penetrate into target tissues. Recently, it was demonstrated that nanobubbles stabilized by nanoparticles can nucleate ultrasound responsive microbubbles under reduced acoustic pressures, which is very promising for the development of nanoscale (<100 nm) ultrasound agents. However, there is still very little understanding about the effects of nanoparticle properties on the stabilization of nanobubbles and nucleation of acoustic cavitation by these nanobubbles. Here, a series of mesoporous silica nanoparticles with sizes around 100 nm but with different morphologies were synthesized to understand the effects of nanoparticle porosity, surface roughness, hydrophobicity, and hydrophilic surface modification on acoustic cavitation inception by porous nanoparticles. The chemical analyses of the nanoparticles showed that, while the nanoparticles were prepared using the same silica precursor (TEOS) and surfactant (CTAB), they revealed varying amounts of carbon impurities, hydroxyl content, and degrees of silica crosslinking. Carbon impurities or hydrophobic modification with methyl groups is found to be essential for nanobubble stabilization by mesoporous silica nanoparticles. The acoustic cavitation experiments in the presence of ethanol and/or bovine serum albumin (BSA) demonstrated that acoustic cavitation is predominantly nucleated by the nanobubbles stabilized at the nanoparticle surface not inside the mesopores. Finally, acoustic cavitation experiments with rough and smooth nanoparticles were suggested that a rough nanoparticle surface is needed to largely preserve surface nanobubbles after coating the surface with hydrophilic macromolecules, which is required for in vivo applications of nanoparticles.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| | - Rajarshi Chattaraj
- Department of Mechanical Engineering, University of Colorado Boulder. Boulder, Colorado 80309, United States
| | - Nicholas T. Blum
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder. Boulder, Colorado 80303, United States
| |
Collapse
|
27
|
Improving performance of nanoscale ultrasound contrast agents using N,N-diethylacrylamide stabilization. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:59-67. [PMID: 27565686 DOI: 10.1016/j.nano.2016.08.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/25/2016] [Accepted: 08/09/2016] [Indexed: 12/21/2022]
Abstract
The design of nanoscale yet highly echogenic agents for imaging outside of the vasculature and for ultrasound-mediated drug delivery remains a formidable challenge. We have previously reported on formulation of echogenic perfluoropropane gas nanobubbles stabilized by a lipid-pluronic surfactant shell. In the current work we describe the development of a new generation of these nanoparticles which consist of perfluoropropane gas stabilized by a surfactant and lipid membrane and a crosslinked network of N,N-diethylacrylamide. The resulting crosslinked nanobubbles (CL-PEG-NB) were 95.2±25.2nm in diameter and showed significant improvement in stability and retention of echogenic signal over 24h. In vivo analysis via ultrasound and fluorescence mediated tomography showed greater tumor extravasation and accumulation with CL-PEG-NB compared to microbubbles. Together these results demonstrate the capabilities and advantages of a new, more stable, nanometer-scale ultrasound contrast agent that can be utilized in future work for diagnostic scans and molecular imaging.
Collapse
|
28
|
Yildirim A, Chattaraj R, Blum NT, Goldscheitter GM, Goodwin AP. Stable Encapsulation of Air in Mesoporous Silica Nanoparticles: Fluorocarbon-Free Nanoscale Ultrasound Contrast Agents. Adv Healthc Mater 2016; 5:1290-8. [PMID: 26990167 PMCID: PMC5058514 DOI: 10.1002/adhm.201600030] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/17/2016] [Indexed: 01/09/2023]
Abstract
While gas-filled micrometer-sized ultrasound contrast agents vastly improve signal-to-noise ratios, microbubbles have short circulation lifetimes and poor extravasation from the blood. Previously reported fluorocarbon-based nanoscale contrast agents are more stable but their contrast is generally lower owing to their size and dispersity. The contrast agents reported here are composed of silica nanoparticles of ≈100 nm diameter that are filled with ≈3 nm columnar mesopores. Functionalization of the silica surface with octyl groups and resuspension with Pluronic F127 create particles with pores that remain filled with air but are stable in buffer and serum. Administration of high intensity focused ultrasound (HIFU) allows sensitive imaging of the silica nanoparticles down to 10(10) particles mL(-1) , with continuous imaging for at least 20 min. Control experiments with different silica particles supported the hypothesis that entrapped air could be pulled into bubble nuclei, which can then in turn act as acoustic scatterers. This process results in very little hemolysis in whole blood, indicating potential for nontoxic blood pool imaging. Finally, the particles are lyophilized and reconstituted or stored in PBS (phosphate-buffered saline, at least for four months) with no loss in contrast, indicating stability to storage and reformulation.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Rajarshi Chattaraj
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Nicholas T. Blum
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Galen M. Goldscheitter
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
29
|
Park JY, Lee TS, Song IH, Cho YL, Chae JR, Yun M, Kang H, Lee JH, Lim JH, Cho WG, Kang WJ. Hybridization-based aptamer labeling using complementary oligonucleotide platform for PET and optical imaging. Biomaterials 2016; 100:143-51. [PMID: 27258484 DOI: 10.1016/j.biomaterials.2016.05.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 11/29/2022]
Abstract
Aptamers are promising next-generation ligands used in molecular imaging and theragnosis. Aptamers are synthetic nucleic acids that can be held together with complementary sequences by base-pair hybridization. In this study, the complementary oligonucleotide (cODN) hybridization-based aptamer conjugation platform was developed to use aptamers as the molecular imaging agent. The cODN was pre-labeled with fluorescent dye or radioisotope and hybridized with a matched sequence containing aptamers in aqueous conditions. The cODN platform-hybridized aptamers exhibited good serum stability and specific binding affinity towards target cancer cells both in vitro and in vivo. These results suggest that the newly designed aptamer conjugation platform offers great potential for the versatile application of aptamers as molecular imaging agents.
Collapse
Affiliation(s)
- Jun Young Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Republic of Korea
| | - Tae Sup Lee
- Molecular Imaging Research Center, Research Institute of Radiological and Medical Sciences, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - In Ho Song
- Molecular Imaging Research Center, Research Institute of Radiological and Medical Sciences, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Ye Lim Cho
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ju Ri Chae
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyungu Kang
- R&D Strategic Planning, Bundang CHA Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13496, Republic of Korea
| | - Jung Hwan Lee
- APGEN Inc, 220, Yeongsin-ro, Yeongdeungpo-gu, Seoul 07228, Republic of Korea
| | - Jong Hoon Lim
- APGEN Inc, 220, Yeongsin-ro, Yeongdeungpo-gu, Seoul 07228, Republic of Korea
| | - Won Gil Cho
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Republic of Korea.
| | - Won Jun Kang
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
30
|
Peyman SA, McLaughlan JR, Abou-Saleh RH, Marston G, Johnson BRG, Freear S, Coletta PL, Markham AF, Evans SD. On-chip preparation of nanoscale contrast agents towards high-resolution ultrasound imaging. LAB ON A CHIP 2016; 16:679-87. [PMID: 26689151 DOI: 10.1039/c5lc01394a] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Micron-sized lipid-stabilised bubbles of heavy gas have been utilised as contrast agents for diagnostic ultrasound (US) imaging for many years. Typically bubbles between 1 and 8 μm in diameter are produced to enhance imaging in US by scattering sound waves more efficiently than surrounding tissue. A potential area of interest for Contrast Enhanced Ultrasound (CEUS) are bubbles with diameters <1 μm or 'nanobubbles.' As bubble diameter decreases, ultrasonic resonant frequency increases, which could lead to an improvement in resolution for high-frequency imaging applications when using nanobubbles. In addition, current US contrast agents are limited by their size to the vasculature in vivo. However, molecular-targeted nanobubbles could penetrate into the extra-vascular space of cancerous tissue providing contrast in regions inaccessible to traditional microbubbles. This paper reports a new microfluidic method for the generation of sub-micron sized lipid stabilised particles containing perfluorocarbon (PFC). The nanoparticles are produced in a unique atomisation-like flow regime at high production rates, in excess of 10(6) particles per s and at high concentration, typically >10(11) particles per mL. The average particle diameter appears to be around 100-200 nm. These particles, suspected of being a mix of liquid and gaseous C4F10 due to Laplace pressure, then phase convert into nanometer sized bubbles on the application of US. In vitro ultrasound characterisation from these nanoparticle populations showed strong backscattering compared to aqueous filled liposomes of a similar size. The nanoparticles were stable upon injection and gave excellent contrast enhancement when used for in vivo imaging, compared to microbubbles with an equivalent shell composition.
Collapse
Affiliation(s)
- Sally A Peyman
- School of Physics and Astronomy, University of Leeds, LS2 9JT, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
Dougherty CA, Cai W, Hong H. Applications of aptamers in targeted imaging: state of the art. Curr Top Med Chem 2016; 15:1138-52. [PMID: 25866268 DOI: 10.2174/1568026615666150413153400] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/13/2015] [Accepted: 02/14/2015] [Indexed: 01/23/2023]
Abstract
Aptamers are single-stranded oligonucleotides with high affinity and specificity to the target molecules or cells, thus they can serve as an important category of molecular targeting ligand. Since their discovery, aptamers have been rapidly translated into clinical practice. The strong target affinity/selectivity, cost-effectivity, chemical versatility and safety of aptamers are superior to traditional peptides- or proteins-based ligands which make them unique choices for molecular imaging. Therefore, aptamers are considered to be extremely useful to guide various imaging contrast agents to the target tissues or cells for optical, magnetic resonance, nuclear, computed tomography, ultrasound and multimodality imaging. This review aims to provide an overview of aptamers' advantages as targeting ligands and their application in targeted imaging. Further research in synthesis of new types of aptamers and their conjugation with new categories of contrast agents is required to develop clinically translatable aptamer-based imaging agents which will eventually result in improved patient care.
Collapse
Affiliation(s)
| | - Weibo Cai
- Department of Radiology, University of Wisconsin - Madison, Wisconsin 53705-2275, United States.
| | | |
Collapse
|
33
|
Zhang X, Zhao K, Wang J, Bai S, Jiao S, Zhang J, Yu L. Design of simvastatin-loaded polymeric microbubbles as targeted ultrasound contrast agents for vascular imaging and drug delivery in the identification of atherosclerotic plaque. NEW J CHEM 2016. [DOI: 10.1039/c5nj02292d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Simvastatin-loaded polymeric microbubbles were synthesized as targeted ultrasound contrast agents and ultrasound-triggered drug carriers.
Collapse
Affiliation(s)
- Xiangyu Zhang
- College of Pharmacy
- Jiamusi University
- Jiamusi 154007
- China
| | - Kaiyue Zhao
- College of Pharmacy
- Jiamusi University
- Jiamusi 154007
- China
| | - Jun Wang
- College of Material Science and Chemical Engineering
- Harbin Engineering University
- Harbin 150001
- China
| | - Shujie Bai
- College of Pharmacy
- Jiamusi University
- Jiamusi 154007
- China
| | - Shuqing Jiao
- College of Pharmacy
- Jiamusi University
- Jiamusi 154007
- China
| | - Jie Zhang
- College of Pharmacy
- Jiamusi University
- Jiamusi 154007
- China
| | - Lian Yu
- College of Pharmacy
- Jiamusi University
- Jiamusi 154007
- China
| |
Collapse
|
34
|
Zhu Y, Hu XC, Shi S, Gao RR, Huang HL, Zhu YY, Lv XY, Yao TM. Ultrasensitive and universal fluorescent aptasensor for the detection of biomolecules (ATP, adenosine and thrombin) based on DNA/Ag nanoclusters fluorescence light-up system. Biosens Bioelectron 2015; 79:205-12. [PMID: 26706942 DOI: 10.1016/j.bios.2015.12.015] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 10/22/2022]
Abstract
We report here an ultrasensitive strategy based on the recognition-induced conformational alteration of aptamer and fluorescence turn-on abilities of guanine-rich (G-rich) DNA sequence in proximity to silver nanoclusters for adenosine triphosphate (ATP), adenosine (A) and thrombin (TB) detection. Herein, we designed two tailored DNA sequences noted as complementary DNA (abbreviated as c-DNA) and signal probe DNA (abbreviated as s-DNA), respectively. c-DNA is designed as a special structure consisting of a sequence complementary to aptamer at the 3'-end and a guanine-rich DNA sequence at the 5'-end; s-DNA contains a cytosine-rich sequence responsible for Ag NCs templated synthesis at the 3'-end and a link sequence (part of aptamer) complementary to partial of the c-DNA at the 5'-end. In the presence of target, the aptamer associated with the target, resulting in the formation of duplex DNA (dsDNA), the DNA-Ag NCs thereafter could close to the guanine-rich sequence, leading to enhanced fluorescence signal readout. The widespread application of the sensing system is achieved success in the detection of three biomolecules. ATP, adenosine and thrombin in the range of 0.5-8.0 μM, 0.5-7.0 μM and 50-900 nM could be linearly detected with the detection limits of 91.6 nM, 103.4 nM and 8.4 nM, respectively. This label-free and turn-on fluorescent sensing system employing the mechanism proposed here turns out to be sensitive, selective, and convenient for the detection of biomolecules without washing and separation steps.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Xiao-Chun Hu
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Shuo Shi
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China.
| | - Ru-Ru Gao
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Hai-Liang Huang
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Yan-Yan Zhu
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Xiao-Yan Lv
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Tian-Ming Yao
- Department of Chemistry, and Shanghai Key Lab of Chemical Assessment and Sustainability, Tongji University, Shanghai 200092, China.
| |
Collapse
|
35
|
Prakash JS, Rajamanickam K. Aptamers and Their Significant Role in Cancer Therapy and Diagnosis. Biomedicines 2015; 3:248-269. [PMID: 28536411 PMCID: PMC5344239 DOI: 10.3390/biomedicines3030248] [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: 06/03/2015] [Accepted: 08/07/2015] [Indexed: 02/07/2023] Open
Abstract
Aptamers are nucleic acid/peptide molecules that can be generated by a sophisticated, well-established technique known as Systematic Evolution of Ligands by EXponential enrichment (SELEX). Aptamers can interact with their targets through structural recognition, as in antibodies, though with higher specificity. With this added advantage, they can be made useful for clinical applications such as targeted therapy and diagnosis. In this review, we have discussed the steps involved in SELEX process and modifications executed to attain high affinity nucleic acid aptamers. Moreover, our review also highlights the therapeutic applications of aptamer functionalized nanoparticles and nucleic acids as chemo-therapeutic agents. In addition, we have described the development of "aptasensor" in clinical diagnostic application for detecting cancer cells and the use of aptamers in different routine imaging techniques, such as Positron Emission Tomography/Computed Tomography, Ultrasound, and Magnetic Resonance Imaging.
Collapse
Affiliation(s)
- Joy Sebastian Prakash
- Faculty of Allied Health Sciences (FAHS), Chettinad Academy of Research and Education, Kelambakkam, Chennai 603103, Tamil Nadu, India.
| | - Karunanithi Rajamanickam
- Faculty of Allied Health Sciences (FAHS), Chettinad Academy of Research and Education, Kelambakkam, Chennai 603103, Tamil Nadu, India.
| |
Collapse
|
36
|
Moyer LC, Timbie KF, Sheeran PS, Price RJ, Miller GW, Dayton PA. High-intensity focused ultrasound ablation enhancement in vivo via phase-shift nanodroplets compared to microbubbles. J Ther Ultrasound 2015; 3:7. [PMID: 26045964 PMCID: PMC4455327 DOI: 10.1186/s40349-015-0029-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/15/2015] [Indexed: 12/28/2022] Open
Abstract
Background During high-intensity focused ultrasound (HIFU) surgical procedures, there is a need to rapidly ablate pathological tissue while minimizing damage to healthy tissue. Current techniques are limited by relatively long procedure times and risks of off-target heating of healthy tissue. One possible solution is the use of microbubbles, which can improve the efficiency of thermal energy delivery during HIFU procedures. However, microbubbles also suffer from limitations such as low spatial selectivity and short circulation time in vivo. In this study, the use of a dual-perfluorocarbon nanodroplet that can enhance thermal ablation, yet retains high spatial selectivity and circulation half-life, was evaluated in vivo and compared to traditional microbubble agents during HIFU ablations of rat liver. Methods High-intensity focused ultrasound (1.1 MHz, 4.1 MPa, 15-s continuous wave) was applied to rat liver in vivo, and heating was monitored during sonication by magnetic resonance thermometry. Thermometry data were analyzed to quantify temperature rise and ablated area, both at the target and prefocally, for HIFU applied 5, 15, or 95 min after intravenous injection of either nanodroplet or microbubble agents. Sham control experiments (no injected agents) were also performed. Results At all three time points, nanodroplets significantly enhanced thermal delivery to the target, achieving temperatures 130 % higher and ablated areas 30 times larger than no-agent control sonications. Nanodroplets did not significantly enhance off-target surface heating. Microbubbles also resulted in significantly greater thermal delivery, but heating was concentrated at the proximal surface of the animal, causing skin burns. Furthermore, microbubbles resulted in lower thermal delivery to the desired target than even the control case, with the notable exception of the 95-min time point. Conclusions Results indicate that the nanodroplet formulation studied here can substantially increase thermal delivery at the acoustic focus while avoiding prefocal heating. In contrast, microbubbles resulted in greater prefocal heating and less heating at the target. Furthermore, nanodroplets are sufficiently stable to enhance HIFU ablation in vivo for at least 1.5 h after injection. The use of a dual-perfluorocarbon nanodroplet formulation as described herein could substantially reduce HIFU procedure times without increasing the risk of skin burns.
Collapse
Affiliation(s)
- Linsey C Moyer
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and NC State University Campus, Box 7575, Chapel Hill, NC 27599 USA
| | - Kelsie F Timbie
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, Virginia 22908 USA
| | - Paul S Sheeran
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and NC State University Campus, Box 7575, Chapel Hill, NC 27599 USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, Virginia 22908 USA ; Department of Radiology and Medical Imaging, University of Virginia, Box 801339, Charlottesville, Virginia 22908 USA
| | - G Wilson Miller
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, Virginia 22908 USA ; Department of Radiology and Medical Imaging, University of Virginia, Box 801339, Charlottesville, Virginia 22908 USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and NC State University Campus, Box 7575, Chapel Hill, NC 27599 USA
| |
Collapse
|
37
|
Chen Q, Luo L, White HS. Electrochemical Generation of a Hydrogen Bubble at a Recessed Platinum Nanopore Electrode. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:4573-81. [PMID: 25811080 DOI: 10.1021/acs.langmuir.5b00234] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We report the electrochemical generation of a single hydrogen bubble within the cavity of a recessed Pt nanopore electrode. The recessed Pt electrode is a conical pore in glass that contains a micrometer-scale Pt disk (1-10 μm radius) at the nanopore base and a nanometer-scale orifice (10-100 nm radius) that restricts diffusion of electroactive molecules and dissolved gas between the nanopore cavity and bulk solution. The formation of a H2 bubble at the Pt disk electrode in voltammetric experiments results from the reduction of H(+) in a 0.25 M H2SO4 solution; the liquid-to-gas phase transformation is indicated in the voltammetric response by a precipitous decrease in the cathodic current due to rapid bubble nucleation and growth within the nanopore cavity. Finite element simulations of the concentration distribution of dissolved H2 within the nanopore cavity, as a function of the H(+) reduction current, indicate that H2 bubble nucleation at the recessed Pt electrode surface occurs at a critical supersaturation concentration of ∼0.22 M, in agreement with the value previously obtained at (nonrecessed) Pt disk electrodes (∼0.25 M). Because the nanopore orifice limits the diffusion of H2 out of the nanopore cavity, an anodic peak corresponding to the oxidation of gaseous and dissolved H2 trapped in the recessed cavity is readily observed on the reverse voltammetric scan. Integration of the charge associated with the H2 oxidation peak is found to approach that of the H(+) reduction peak at high scan rates, confirming the assignment of the anodic peak to H2 oxidation. Preliminary results for the electrochemical generation of O2 bubbles from water oxidation at a recessed nanopore electrode are consistent with the electrogeneration of H2 bubbles.
Collapse
Affiliation(s)
- Qianjin Chen
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | - Long Luo
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| | - Henry S White
- Department of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
| |
Collapse
|
38
|
Huang HY, Liu HL, Hsu PH, Chiang CS, Tsai CH, Chi HS, Chen SY, Chen YY. A multitheragnostic nanobubble system to induce blood-brain barrier disruption with magnetically guided focused ultrasound. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:655-61. [PMID: 25472627 DOI: 10.1002/adma.201403889] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Indexed: 05/11/2023]
Abstract
A novel magnetically guidable nanobubble is designed for disrupting the blood-brain barrier (BBB) by combining magnetic guidance with focused ultrasound in vivo. The magnetic-nanobubble platform also demonstrates the potential to serve as a unique theranostic tool via performing focused ultrasound (FUS)-induced BBB disruption and magnetic resonance imaging (MRI)/ultrasound dual-modality contrast-agent imaging to improve the drug delivery of therapeutic substances or gene therapy into the central nervous system.
Collapse
Affiliation(s)
- Hsin-Yang Huang
- Department of Materials Science and Engineering, National Chiao Tung University, No. 1001 Ta-Hsueh Rd., Taiwan, Hsinchu, 300, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Wu X, Chen J, Wu M, Zhao JX. Aptamers: active targeting ligands for cancer diagnosis and therapy. Theranostics 2015; 5:322-44. [PMID: 25699094 PMCID: PMC4329498 DOI: 10.7150/thno.10257] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023] Open
Abstract
Aptamers, including DNA, RNA and peptide aptamers, are a group of promising recognition units that can specifically bind to target molecules and cells. Due to their excellent specificity and high affinity to targets, aptamers have attracted great attention in various fields in which selective recognition units are required. They have been used in biosensing, drug delivery, disease diagnosis and therapy (especially for cancer treatment). In this review, we summarized recent applications of DNA and RNA aptamers in cancer theranostics. The specific binding ability of aptamers to cancer-related markers and cancer cells ensured their high performance for early diagnosis of cancer. Meanwhile, the efficient targeting ability of aptamers to cancer cells and tissues provided a promising way to deliver imaging agents and drugs for cancer imaging and therapy. Furthermore, with the development of nanoscience and nanotechnology, the conjugation of aptamers with functional nanomaterials paved an exciting way for the fabrication of theranostic agents for different types of cancers, which might be a powerful tool for cancer treatment.
Collapse
Affiliation(s)
- Xu Wu
- 1. Department of Chemistry, School of Arts and Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Jiao Chen
- 1. Department of Chemistry, School of Arts and Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Min Wu
- 2. Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Julia Xiaojun Zhao
- 1. Department of Chemistry, School of Arts and Sciences, University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|
40
|
Perera RH, Hernandez C, Zhou H, Kota P, Burke A, Exner AA. Ultrasound imaging beyond the vasculature with new generation contrast agents. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 7:593-608. [PMID: 25580914 DOI: 10.1002/wnan.1326] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/17/2014] [Accepted: 11/08/2014] [Indexed: 12/21/2022]
Abstract
Current commercially available ultrasound contrast agents are gas-filled, lipid- or protein-stabilized microbubbles larger than 1 µm in diameter. Because the signal generated by these agents is highly dependent on their size, small yet highly echogenic particles have been historically difficult to produce. This has limited the molecular imaging applications of ultrasound to the blood pool. In the area of cancer imaging, microbubble applications have been constrained to imaging molecular signatures of tumor vasculature and drug delivery enabled by ultrasound-modulated bubble destruction. Recently, with the rise of sophisticated advancements in nanomedicine, ultrasound contrast agents, which are an order of magnitude smaller (100-500 nm) than their currently utilized counterparts, have been undergoing rapid development. These agents are poised to greatly expand the capabilities of ultrasound in the field of targeted cancer detection and therapy by taking advantage of the enhanced permeability and retention phenomenon of many tumors and can extravasate beyond the leaky tumor vasculature. Agent extravasation facilitates highly sensitive detection of cell surface or microenvironment biomarkers, which could advance early cancer detection. Likewise, when combined with appropriate therapeutic agents and ultrasound-mediated deployment on demand, directly at the tumor site, these nanoparticles have been shown to contribute to improved therapeutic outcomes. Ultrasound's safety profile, broad accessibility and relatively low cost make it an ideal modality for the changing face of healthcare today. Aided by the multifaceted nano-sized contrast agents and targeted theranostic moieties described herein, ultrasound can considerably broaden its reach in future applications focused on the diagnosis and staging of cancer.
Collapse
Affiliation(s)
- Reshani H Perera
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher Hernandez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Haoyan Zhou
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pavan Kota
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Alan Burke
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
41
|
Chen Y, Chen H, Shi J. Nanobiotechnology promotes noninvasive high-intensity focused ultrasound cancer surgery. Adv Healthc Mater 2015; 4:158-65. [PMID: 24898413 DOI: 10.1002/adhm.201400127] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/12/2014] [Indexed: 01/20/2023]
Abstract
The successful cancer eradication in a noninvasive manner is the ultimate objective in the fight against cancer. As a "bloodless scalpel," high-intensity focused ultrasound (HIFU) is regarded as one of the most promising and representative noninvasive therapeutic modalities for cancer surgery. However, large-scale clinical applications of HIFU are still in their infancy because of critical efficiency and safety issues which remain to be solved. Fortunately, recently developed nanobiotechnology provides an alternative efficient approach to improve such important issues in HIFU, especially for cancer therapy. This Research News presents the very recent exciting progresses on the elaborate design and fabrication of organic, inorganic, and organic/inorganic hybrid nanoparticles for enhancing the HIFU ablation efficiency against tumor tissues. It is highly expected that this Research News can arouse more extensive research enthusiasm on the development of functional nanomaterials for highly efficient HIFU-based synergistic therapy, which will give a promising noninvasive therapeutic modality for the successful cancer therapy with minimal damage to surrounding normal tissues, due to the noninvasive and site-specific therapeutic features of HIFU.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures; Shanghai Institute of Ceramics, Chinese Academy of Sciences; 1295 Ding-Xi Road Shanghai 200050 P. R. China
| | - Hangrong Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures; Shanghai Institute of Ceramics, Chinese Academy of Sciences; 1295 Ding-Xi Road Shanghai 200050 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures; Shanghai Institute of Ceramics, Chinese Academy of Sciences; 1295 Ding-Xi Road Shanghai 200050 P. R. China
| |
Collapse
|
42
|
Yuksel Durmaz Y, Vlaisavljevich E, Xu Z, ElSayed M. Development of Nanodroplets for Histotripsy-Mediated Cell Ablation. Mol Pharm 2014; 11:3684-95. [DOI: 10.1021/mp500419w] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Yasemin Yuksel Durmaz
- Department
of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Eli Vlaisavljevich
- Department
of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhen Xu
- Department
of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mohamed ElSayed
- Department
of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Macromolecular
Science and Engineering Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
43
|
Preparation and in vivo evaluation of ligand-conjugated polymeric microbubbles as targeted ultrasound contrast agents. Colloids Surf A Physicochem Eng Asp 2014. [DOI: 10.1016/j.colsurfa.2014.03.082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
44
|
Abstract
This review highlights recent progress in developing DNA aptamers for personalized medicine, with more focus on in vivo studies for potential clinical applications. Examples include design of aptamers in combination with DNA nanostructures, nanomaterials, or microfluidic devices as diagnostic probes or therapeutic agents for cancers and other diseases. The use of aptamers as targeting agents in drug delivery is also covered. The advantages and future directions of such DNA aptamer-based technology for the continued development of personalized medicine are discussed.
Collapse
Affiliation(s)
- Hang Xing
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801 ; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Kevin Hwang
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Ji Li
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801 ; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Seyed-Fakhreddin Torabi
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Yi Lu
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801 ; Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801 ; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
45
|
Kong HY, Byun J. Nucleic Acid aptamers: new methods for selection, stabilization, and application in biomedical science. Biomol Ther (Seoul) 2014; 21:423-34. [PMID: 24404332 PMCID: PMC3879913 DOI: 10.4062/biomolther.2013.085] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/05/2013] [Accepted: 11/05/2013] [Indexed: 12/19/2022] Open
Abstract
The adoption of oligonucleotide aptamer is well on the rise, serving an ever increasing demand for versatility in biomedical field. Through the SELEX (Systematic Evolution of Ligands by EXponential enrichment), aptamer that can bind to specific target with high affinity and specificity can be obtained. Aptamers are single-stranded nucleic acid molecules that can fold into complex threedimensional structures, forming binding pockets and clefts for the specific recognition and tight binding of any given molecular target. Recently, aptamers have attracted much attention because they not only have all of the advantages of antibodies, but also have unique merits such as thermal stability, ease of synthesis, reversibility, and little immunogenicity. The advent of novel technologies is revolutionizing aptamer applications. Aptamers can be easily modified by various chemical reactions to introduce functional groups and/or nucleotide extensions. They can also be conjugated to therapeutic molecules such as drugs, drug containing carriers, toxins, or photosensitizers. Here, we discuss new SELEX strategies and stabilization methods as well as applications in drug delivery and molecular imaging.
Collapse
Affiliation(s)
- Hoon Young Kong
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Yongin 448-701, Republic of Korea
| | - Jonghoe Byun
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, Yongin 448-701, Republic of Korea
| |
Collapse
|
46
|
Abstract
Aptamers, single-stranded oligonucleotides, are an important class of molecular targeting ligand. Since their discovery, aptamers have been rapidly translated into clinical practice. They have been approved as therapeutics and molecular diagnostics. Aptamers also possess several properties that make them uniquely suited to molecular imaging. This review aims to provide an overview of aptamers' advantages as targeting ligands and their application in molecular imaging.
Collapse
Affiliation(s)
- Andrew Z. Wang
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, University of North Carolina–Chapel Hill, Chapel Hill, North Carolina
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
47
|
Peng SL, Wang FN, Wang CH, Peng HH, Lu CT, Yeh CK. Using microbubbles as an MRI contrast agent for the measurement of cerebral blood volume. NMR IN BIOMEDICINE 2013; 26:1540-1546. [PMID: 23794141 DOI: 10.1002/nbm.2988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 05/17/2013] [Accepted: 05/17/2013] [Indexed: 06/02/2023]
Abstract
The susceptibility differences at the gas-liquid interface of microbubbles (MBs) allow their use as an intravascular susceptibility contrast agent for in vivo MRI. However, the characteristics of MBs are very different from those of the standard gadolinium-diethylenetriaminepentaacetic acid (Gd-DPTA) contrast agent, including the size distribution and hemodynamic properties, which could influence MRI outcomes. Here, we investigate quantitatively the correlation between the relative cerebral blood volume (rCBV) derived from Gd-DTPA (rCBV(Gd)) and the MB-induced susceptibility effect (ΔR(2*MB)) by conventional dynamic susceptibility contrast MRI (DSC-MRI). Custom-made MBs had a mean diameter of 0.92 µm and were capable of inducing 4.68 ± 3.02% of the maximum signal change (MSC). The MB-associated ΔR(2*MB) was compared with rCBV(Gd) in 16 rats on 4.7-T MRI. We observed a significant effect of the time to peak (TTP) on the correlation between ΔR(2*MB) and rCBV(Gd), and also found a noticeable dependence between TTP and MSC. Our findings suggest that MBs with longer TTPs can be used for the estimation of rCBV by DSC-MRI, and emphasize the critical effect of TTP on MB-based contrast MRI.
Collapse
Affiliation(s)
- Shin-Lei Peng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | | | |
Collapse
|
48
|
Hwang TL, Sung CT, Aljuffali IA, Chang YT, Fang JY. Cationic surfactants in the form of nanoparticles and micelles elicit different human neutrophil responses: a toxicological study. Colloids Surf B Biointerfaces 2013; 114:334-41. [PMID: 24246197 DOI: 10.1016/j.colsurfb.2013.10.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 10/03/2013] [Accepted: 10/16/2013] [Indexed: 01/28/2023]
Abstract
Cationic surfactants are an ingredient commonly incorporated into nanoparticles for clinical practicability; however, the toxicity of cationic surfactants in nanoparticles is not fully elucidated. We aimed to evaluate the inflammatory responses of cationic nanobubbles and micelles in human neutrophils. Soyaethyl morpholinium ethosulfate (SME) and hexadecyltrimethyl-ammonium bromide (CTAB) are the two cationic surfactants employed in this study. The zeta potential of CTAB nanobubbles was 80 mV, which was the highest among all formulations. Nanobubbles, without cationic surfactants, showed no cytotoxic effects on neutrophils in terms of inflammatory responses. Cationic nanobubbles caused a concentration-dependent cytotoxicity of degranulation (elastase release) and membrane damage (release of lactate dehydrogenase, LDH). Among all nanoparticles and micelles, CTAB-containing nanosystems showed the greatest inflammatory responses. A CTAB nanobubble diluent (1/150) increased the LDH release 80-fold. Propidium iodide staining and scanning electron microscopy (SEM) verified cell death and morphological change of neutrophils treated by CTAB nanobubbles. SME, in a micelle form, strengthened the inflammatory response more than SME-loaded nanobubbles. Membrane interaction and subsequent Ca(2+) influx were the mechanisms that triggered inflammation. The information obtained from this work is beneficial in designing nanoparticulate formulations for balancing clinical activity and toxicity.
Collapse
Affiliation(s)
- Tsong-Long Hwang
- Cell Pharmacology Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Calvin T Sung
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA USA
| | - Ibrahim A Aljuffali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Yuan-Ting Chang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan; Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
49
|
Zhu L, Torchilin VP. Stimulus-responsive nanopreparations for tumor targeting. Integr Biol (Camb) 2013; 5:96-107. [PMID: 22869005 DOI: 10.1039/c2ib20135f] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nanopreparations such as liposomes, micelles, polymeric and inorganic nanoparticles, and small molecule/nucleic acid/protein conjugates have demonstrated various advantages over "naked" therapeutic molecules. These nanopreparations can be further engineered with functional moieties to improve their performance in terms of circulation longevity, targetability, enhanced intracellular penetration, carrier-mediated enhanced visualization, and stimuli-sensitivity. The idea of application of a stimulus-sensitive drug or imaging agent delivery system for tumor targeting is based on the significant abnormalities in the tumor microenvironment and its cells, such as an acidic pH, altered redox potential, up-regulated proteins and hyperthermia. These internal conditions as well as external stimuli, such as magnetic field, ultrasound and light, can be used to modify the behavior of the nanopreparations that control drug release, improve drug internalization, control the intracellular drug fate and even allow for certain physical interactions, resulting in an enhanced tumor targeting and antitumor effect. This article provides a critical view of current stimulus-sensitive drug delivery strategies and possible future directions in tumor targeting with primary focus on the combined use of stimulus-sensitivity with other strategies in the same nanopreparation, including multifunctional nanopreparations and theranostics.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Pharmaceutical Sciences and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 140 The Fenway, Boston, MA 02115, USA
| | | |
Collapse
|
50
|
Abstract
Time and space controlled drug delivery still remains a huge challenge in medicine. A novel approach that could offer a solution is ultrasound guided drug delivery. “Ultrasonic drug delivery” is often based on the use of small gas bubbles (so-called microbubbles) that oscillate and cavitate upon exposure to ultrasound waves. Some microbubbles are FDA approved contrast agents for ultrasound imaging and are nowadays widely investigated as promising drug carriers. Indeed, it has been observed that upon exposure to ultrasound waves, microbubbles may (a) release the encapsulated drugs and (b) simultaneously change the structure of the cell membranes in contact with the microbubbles which may facilitate drug entrance into cells. This review aims to highlight (a) major factors known so far which affect ultrasonic drug delivery (like the structure of the microbubbles, acoustic settings, etc.) and (b) summarizes the recent preclinical progress in this field together with a number of promising new concepts and applications.
Collapse
|