1
|
He M, Chen X, Yu F, Qin B, Wang H, Lavery L, Villanueva FS. Enhanced Antitumor Efficacy and Reduced Cardiotoxicity of Ultrasound-Mediated Doxorubicin Delivery by Microbubble-Liposome Complexes. ULTRASOUND IN MEDICINE & BIOLOGY 2025:S0301-5629(25)00127-9. [PMID: 40368702 DOI: 10.1016/j.ultrasmedbio.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
OBJECTIVE Doxorubicin (Dox) is standard of care for treatment of sarcomas, but cumulative dosing is often limited by cardiotoxicity. We hypothesized that ultrasound targeted microbubble (MB) cavitation (UTMC) of a liposomal doxorubicin (LDox) conjugated polymer microbubble complex (DoxLPX) would enhance tumor inhibition and limit Dox cardiotoxicity. METHODS DoxLPX was intravenously injected in MCA205 sarcoma-bearing mice and concurrent ultrasound was delivered to the tumor site (DoxLPX + UTMC). Other mice received equivalent dosages of free Dox, LDox, or LDox + MB co-administration with UTMC (LDox + MB + UTMC). Tumor size and cardiac function were serially imaged with ultrasound. Postmortem cardiac tissue was analyzed for apoptosis. Biodistribution of Dox was performed with bioluminescence imaging postmortem where Cy5.5 was used as a fluorescent Dox analog. RESULTS DoxLPX + UTMC showed increased drug concentration in the tumor, a significant slowdown in tumor growth and prolonged median survival time. LDox and DoxLPX formulations had reduced drug extravasation into the myocardium. LDox + MB + UTMC also demonstrated superior tumor growth inhibition compared to free Dox and LDox. Three weeks after treatment commenced, DoxLPX + UTMC group showed significantly better left ventricular function indices than the free Dox group, consistent with biodistribution findings. Concordantly, heart tissue showed normal architecture of cardiac myocytes and significantly less interstitial/perivascular fibrosis in the DoxLPX + UTMC group. CONCLUSIONS DoxLPX formulation in conjunction with ultrasound provides a targeted drug delivery platform with superior anti-tumor efficacy and reduced cardiac toxicity compared with systemic administration of free Dox.
Collapse
Affiliation(s)
- Mingyu He
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francois Yu
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bin Qin
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huizhu Wang
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Linda Lavery
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Fernandes DA. Comprehensive Review on Bubbles: Synthesis, Modification, Characterization and Biomedical Applications. Bioconjug Chem 2024; 35:1639-1686. [PMID: 39377727 DOI: 10.1021/acs.bioconjchem.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Accurate detection, treatment, and imaging of diseases are important for effective treatment outcomes in patients. In this regard, bubbles have gained much attention, due to their versatility. Bubbles usually 1 nm to 10 μm in size can be produced and loaded with a variety of lipids, polymers, proteins, and therapeutic and imaging agents. This review details the different production and loading methods for bubbles, for imaging and treatment of diseases/conditions such as cancer, tumor angiogenesis, thrombosis, and inflammation. Bubbles can also be used for perfusion measurements, important for diagnostic and therapeutic decision making in cardiac disease. The different factors important in the stability of bubbles and the different techniques for characterizing their physical and chemical properties are explained, for developing bubbles with advanced therapeutic and imaging features. Hence, the review provides important insights for researchers studying bubbles for biomedical applications.
Collapse
|
3
|
Al Refaai KA, AlSawaftah NA, Abuwatfa W, Husseini GA. Drug Release via Ultrasound-Activated Nanocarriers for Cancer Treatment: A Review. Pharmaceutics 2024; 16:1383. [PMID: 39598507 PMCID: PMC11597164 DOI: 10.3390/pharmaceutics16111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Conventional cancer chemotherapy often struggles with safely and effectively delivering anticancer therapeutics to target tissues, frequently leading to dose-limiting toxicity and suboptimal therapeutic outcomes. This has created a need for novel therapies that offer greater efficacy, enhanced safety, and improved toxicological profiles. Nanocarriers are nanosized particles specifically designed to enhance the selectivity and effectiveness of chemotherapy drugs while reducing their toxicity. A subset of drug delivery systems utilizes stimuli-responsive nanocarriers, which enable on-demand drug release, prevent premature release, and offer spatial and temporal control over drug delivery. These stimuli can be internal (such as pH and enzymes) or external (such as ultrasound, magnetic fields, and light). This review focuses on the mechanics of ultrasound-induced drug delivery and the various nanocarriers used in conjunction with ultrasound. It will also provide a comprehensive overview of key aspects related to ultrasound-induced drug delivery, including ultrasound parameters and the biological effects of ultrasound waves.
Collapse
Affiliation(s)
- Khaled Armouch Al Refaai
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - Nour A. AlSawaftah
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Biomedical Engineering Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates; (N.A.A.); (W.A.)
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Biosciences and Bioengineering Ph.D. Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
4
|
Owen J, Negussie AH, Burks SR, Delgado J, Mikhail AS, Rivera J, Pritchard WF, Karanian JW, Stride E, Frank JA, Wood BJ. Microbubbles bound to drug-eluting beads enable ultrasound imaging and enhanced delivery of therapeutics. Sci Rep 2024; 14:20929. [PMID: 39251665 PMCID: PMC11383944 DOI: 10.1038/s41598-024-71831-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Transarterial chemoembolization (TACE) is an image-guided minimally invasive treatment for liver cancer which involves delivery of chemotherapy and embolic material into tumor-supplying arteries to block blood flow to a liver tumor and to deliver chemotherapy directly to the tumor. However, the released drug diffuses only less than a millimeter away from the beads. To enhance the efficacy of TACE, the development of microbubbles electrostatically bound to the surface of drug-eluting beads loaded with different amounts of doxorubicin (0-37.5 mg of Dox/mL of beads) is reported. Up to 400 microbubbles were bound to Dox-loaded beads (70-150 microns). This facilitated ultrasound imaging of the beads and increased the release rate of Dox upon exposure to high intensity focused ultrasound (HIFU). Furthermore, ultrasound exposure (1 MPa peak negative pressure) increased the distance at which Dox could be detected from beads embedded in a tissue-mimicking phantom, compared with a no ultrasound control.
Collapse
Affiliation(s)
- Joshua Owen
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Ayele H Negussie
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jose Delgado
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jocelyne Rivera
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - William F Pritchard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - John W Karanian
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Na L, Fan F. Advances in nanobubbles for cancer theranostics: Delivery, imaging and therapy. Biochem Pharmacol 2024; 226:116341. [PMID: 38848778 DOI: 10.1016/j.bcp.2024.116341] [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: 03/07/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Maximizing treatment efficacy and forecasting patient prognosis in cancer necessitates the strategic use of targeted therapy, coupled with the prompt precise detection of malignant tumors. Theutilizationof gaseous systems as an adaptable platform for creating nanobubbles (NBs) has garnered significant attention as theranostics, which involve combining contrast chemicals typically used for imaging with pharmaceuticals to diagnose and treattumorssynergistically in apersonalizedmanner for each patient. This review specifically examines the utilization of oxygen NBsplatforms as a theranostic weapon in the field of oncology. We thoroughly examine the key factors that impact the effectiveness of NBs preparations and the consequences of these treatment methods. This review extensively examines recent advancements in composition schemes, advanced developments in pre-clinical phases, and other groundbreaking inventions in the area of NBs. Moreover, this review offers a thorough examination of the optimistic future possibilities, addressing prospective methods for improvement and incorporation into widely accepted therapeutic practices. As we explore the ever-changing field of cancer theranostics, the incorporation of oxygen NBs appears as a promising development, providing new opportunities for precision medicine and marking a revolutionary age in cancer research and therapy.
Collapse
Affiliation(s)
- Liu Na
- Ultrasound Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Fan Fan
- School of Automation, Xi'an University of Posts and Telecommunications, Xi'an 710121, China.
| |
Collapse
|
6
|
Del Campo Fonseca A, Ahmed D. Ultrasound robotics for precision therapy. Adv Drug Deliv Rev 2024; 205:115164. [PMID: 38145721 DOI: 10.1016/j.addr.2023.115164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
In recent years, the application of microrobots in precision therapy has gained significant attention. The small size and maneuverability of these micromachines enable them to potentially access regions that are difficult to reach using traditional methods; thus, reducing off-target toxicities and maximizing treatment effectiveness. Specifically, acoustic actuation has emerged as a promising method to exert control. By harnessing the power of acoustic energy, these small machines potentially navigate the body, assemble at the desired sites, and deliver therapies with enhanced precision and effectiveness. Amidst the enthusiasm surrounding these miniature agents, their translation to clinical environments has proven difficult. The primary objectives of this review are threefold: firstly, to offer an overview of the fundamental acoustic principles employed in the field of microrobots; secondly, to assess their current applications in medical therapies, encompassing tissue targeting, drug delivery or even cell infiltration; and lastly, to delve into the continuous efforts aimed at integrating acoustic microrobots into in vivo applications.
Collapse
Affiliation(s)
- Alexia Del Campo Fonseca
- Department of Mechanical and Process Engineering, Acoustic Robotics Systems Lab, ETH Zurich, Säumerstrasse 4, 8803 Rüschlikon, Switzerland.
| | - Daniel Ahmed
- Department of Mechanical and Process Engineering, Acoustic Robotics Systems Lab, ETH Zurich, Säumerstrasse 4, 8803 Rüschlikon, Switzerland.
| |
Collapse
|
7
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
8
|
Bourn MD, Mohajerani SZ, Mavria G, Ingram N, Coletta PL, Evans SD, Peyman SA. Tumour associated vasculature-on-a-chip for the evaluation of microbubble-mediated delivery of targeted liposomes. LAB ON A CHIP 2023; 23:1674-1693. [PMID: 36779251 PMCID: PMC10013341 DOI: 10.1039/d2lc00963c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
The vascular system is the primary route for the delivery of therapeutic drugs throughout the body and is an important barrier at the region of disease interest, such as a solid tumour. The development of complex 3D tumour cultures has progressed significantly in recent years however, the generation of perfusable vascularised tumour models still presents many challenges. This study presents a microfluidic-based vasculature system that can be induced to display properties of tumour-associated blood vessels without direct incorporation of tumour cells. Conditioning healthy endothelial-fibroblast cell vasculature co-cultures with media taken from tumour cell cultures was found to result in the formation of disorganised, tortuous networks which display characteristics consistent with those of tumour-associated vasculature. Integrin αvβ3, a cell adhesion receptor associated with angiogenesis, was found to be upregulated in vasculature co-cultures conditioned with tumour cell media (TCM) - consistent with the reported αvβ3 expression pattern in angiogenic tumour vasculature in vivo. Increased accumulation of liposomes (LSs) conjugated to antibodies against αvβ3 was observed in TCM networks compared to non-conditioned networks, indicating αvβ3 may be a potential target for the delivery of drugs specifically to tumour vasculature. Furthermore, the use of microbubbles (MBs) and ultrasound (US) to further enhance the delivery of LSs to TCM-conditioned vasculature was investigated. Quantification of fluorescent LS accumulation post-perfusion of the vascular network showed 3-fold increased accumulation with the use of MBs and US, suggesting that targeted LS delivery could be further improved with the use of locally administered MBs and US.
Collapse
Affiliation(s)
- Matthew D Bourn
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Safoura Zahed Mohajerani
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Georgia Mavria
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Nicola Ingram
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - P Louise Coletta
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| | - Stephen D Evans
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
| | - Sally A Peyman
- School of Physics and Astronomy, University of Leeds, Leeds, LS2 9JT, UK.
- Leeds Institute for Medical Research, Wellcome Trust Brenner Building, St James' University Hospital, Leeds, LS9 7TF, UK
| |
Collapse
|
9
|
Moradi Kashkooli F, Jakhmola A, Hornsby TK, Tavakkoli JJ, Kolios MC. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions. J Control Release 2023; 355:552-578. [PMID: 36773959 DOI: 10.1016/j.jconrel.2023.02.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The application of biocompatible nanocarriers in medicine has provided several benefits over conventional treatment methods. However, achieving high treatment efficacy and deep penetration of nanocarriers in tumor tissue is still challenging. To address this, stimuli-responsive nano-sized drug delivery systems (DDSs) are an active area of investigation in delivering anticancer drugs. While ultrasound is mainly used for diagnostic purposes, it can also be applied to affect cellular function and the delivery/release of anticancer drugs. Therapeutic ultrasound (TUS) has shown potential as both a stand-alone anticancer treatment and a method to induce targeted drug release from nanocarrier systems. TUS approaches have been used to overcome various physiological obstacles, including endothelial barriers, the tumor microenvironment (TME), and immunological hurdles. Combining nanomedicine and ultrasound as a smart DDS can increase in situ drug delivery and improve access to impermeable tissues. Furthermore, smart DDSs can perform targeted drug release in response to distinctive TMEs, external triggers, or dual/multi-stimulus. This results in enhanced treatment efficacy and reduced damage to surrounding healthy tissue or organs at risk. Integrating DDSs and ultrasound is still in its early stages. More research and clinical trials are required to fully understand ultrasound's underlying physical mechanisms and interactions with various types of nanocarriers and different types of cells and tissues. In the present review, ultrasound-mediated nano-sized DDS, specifically focused on cancer treatment, is presented and discussed. Ultrasound interaction with nanoparticles (NPs), drug release mechanisms, and various types of ultrasound-sensitive NPs are examined. Additionally, in vitro, in vivo, and clinical applications of TUS are reviewed in light of the critical challenges that need to be considered to advance TUS toward an efficient, secure, straightforward, and accessible cancer treatment. This study also presents effective TUS parameters and safety considerations for this treatment modality and gives recommendations about system design and operation. Finally, future perspectives are considered, and different TUS approaches are examined and discussed in detail. This review investigates drug release and delivery through ultrasound-mediated nano-sized cancer treatment, both pre-clinically and clinically.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
10
|
Chapla R, Huynh KT, Schutt CE. Microbubble–Nanoparticle Complexes for Ultrasound-Enhanced Cargo Delivery. Pharmaceutics 2022; 14:pharmaceutics14112396. [PMID: 36365214 PMCID: PMC9698658 DOI: 10.3390/pharmaceutics14112396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble–nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid–polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin–avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble–nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.
Collapse
Affiliation(s)
- Rachel Chapla
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katherine T. Huynh
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
11
|
Avry F, Mousset C, Oujagir E, Bouakaz A, Gouilleux-Gruart V, Thépault RA, Renault S, Marouillat S, Machet L, Escoffre JM. Microbubble-Assisted Ultrasound for Imaging and Therapy of Melanoma Skin Cancer: A Systematic Review. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:2174-2198. [PMID: 36050232 DOI: 10.1016/j.ultrasmedbio.2022.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Recent technological developments in ultrasound (US) imaging and ultrasound contrast agents (UCAs) have improved diagnostic confidence in echography. In the clinical management of melanoma, contrast-enhanced ultrasound (CEUS) imaging complements conventional US imaging (i.e., high-resolution US and Doppler imaging) for clinical examination and therapeutic follow-up. These developments have set into motion the combined use of ultrasound and UCAs as a new modality for drug delivery. This modality, called sonoporation, has emerged as a non-invasive, targeted and safe method for the delivery of therapeutic drugs into melanoma. This review focuses on the results and prospects of using US and UCAs as dual modalities for CEUS imaging and melanoma treatment.
Collapse
Affiliation(s)
- François Avry
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Coralie Mousset
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; GICC EA 7501, Université de Tours, Tours, France
| | - Edward Oujagir
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | | | | | | | | | - Laurent Machet
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; Department of Dermatology, Tours University Hospital, Tours, France
| | | |
Collapse
|
12
|
Munir MU. Nanomedicine Penetration to Tumor: Challenges, and Advanced Strategies to Tackle This Issue. Cancers (Basel) 2022; 14:cancers14122904. [PMID: 35740570 PMCID: PMC9221319 DOI: 10.3390/cancers14122904] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Nanomedicine has been under investigation for several years to improve the efficiency of chemotherapeutics, having minimal pharmacological effects clinically. Ineffective tumor penetration is mediated by tumor environments, including limited vascular system, rising cancer cells, higher interstitial pressure, and extra-cellular matrix, among other things. Thus far, numerous methods to increase nanomedicine access to tumors have been described, including the manipulation of tumor micro-environments and the improvement of nanomedicine characteristics; however, such outdated approaches still have shortcomings. Multi-functional convertible nanocarriers have recently been developed as an innovative nanomedicine generation with excellent tumor infiltration abilities, such as tumor-penetrating peptide-mediated transcellular transport. The developments and limitations of nanomedicines, as well as expectations for better outcomes of tumor penetration, are discussed in this review.
Collapse
Affiliation(s)
- Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Aljouf, Saudi Arabia
| |
Collapse
|
13
|
Maciulevičius M, Tamošiūnas M, Navickaitė D, Šatkauskas S, Venslauskas MS. Free- and liposomal- doxorubicin delivery via microbubble inertial cavitation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
14
|
Maciulevičius M, Tamošiūnas M, Jurkonis R, Šatkauskas S. Dosimetric Assessment of Antitumor Treatment by enhanced Bleomycin Delivery via Electroporation and Sonoporation. Bioelectrochemistry 2022; 146:108153. [DOI: 10.1016/j.bioelechem.2022.108153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/15/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
|
15
|
Evaluation of Liposome-Loaded Microbubbles as a Theranostic Tool in a Murine Collagen-Induced Arthritis Model. Sci Pharm 2022. [DOI: 10.3390/scipharm90010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe inflammation of the synovial tissue. Here, we assess the feasibility of liposome-loaded microbubbles as theranostic agents in a murine arthritis model. First, contrast-enhanced ultrasound (CEUS) was used to quantify neovascularization in this model since CEUS is well-established for RA diagnosis in humans. Next, the potential of liposome-loaded microbubbles and ultrasound (US) to selectively enhance liposome delivery to the synovium was evaluated with in vivo fluorescence imaging. This procedure is made very challenging by the presence of hard joints and by the limited lifetime of the microbubbles. The inflamed knee joints were exposed to therapeutic US after intravenous injection of liposome-loaded microbubbles. Loaded microbubbles were found to be quickly captured by the liver. This resulted in fast clearance of attached liposomes while free and long-circulating liposomes were able to accumulate over time in the inflamed joints. Our observations show that murine arthritis models are not well-suited for evaluating the potential of microbubble-mediated drug delivery in joints given: (i) restricted microbubble passage in murine synovial vasculature and (ii) limited control over the exact ultrasound conditions in situ given the much shorter length scale of the murine joints as compared to the therapeutic wavelength.
Collapse
|
16
|
Mukhopadhyay D, Sano C, AlSawaftah N, El-Awady R, Husseini GA, Paul V. Ultrasound-Mediated Cancer Therapeutics Delivery using Micelles and Liposomes: A Review. Recent Pat Anticancer Drug Discov 2021; 16:498-520. [PMID: 34911412 DOI: 10.2174/1574892816666210706155110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/02/2021] [Accepted: 03/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Existing cancer treatment methods have many undesirable side effects that greatly reduce the quality of life of cancer patients. OBJECTIVE This review will focus on the use of ultrasound-responsive liposomes and polymeric micelles in cancer therapy. METHODS This review presents a survey of the literature regarding ultrasound-triggered micelles and liposomes using articles recently published in various journals, as well as some new patents in this field. RESULTS Nanoparticles have proven promising as cancer theranostic tools. Nanoparticles are selective in nature, have reduced toxicity, and controllable drug release patterns making them ideal carriers for anticancer drugs. Numerous nanocarriers have been designed to combat malignancies, including liposomes, micelles, dendrimers, solid nanoparticles, quantum dots, gold nanoparticles, and, more recently, metal-organic frameworks. The temporal and spatial release of therapeutic agents from these nanostructures can be controlled using internal and external triggers, including pH, enzymes, redox, temperature, magnetic and electromagnetic waves, and ultrasound. Ultrasound is an attractive modality because it is non-invasive, can be focused on the diseased site, and has a synergistic effect with anticancer drugs. CONCLUSION The functionalization of micellar and liposomal surfaces with targeting moieties and the use of ultrasound as a triggering mechanism can help improve the selectivity and enable the spatiotemporal control of drug release from nanocarriers.
Collapse
Affiliation(s)
- Debasmita Mukhopadhyay
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Catherine Sano
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Nour AlSawaftah
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
17
|
Browning R, Thomas N, Marsh LK, Tear LR, Owen J, Stride E, Farrer NJ. Ultrasound-Triggered Delivery of Iproplatin from Microbubble-Conjugated Liposomes. ChemistryOpen 2021; 10:1170-1176. [PMID: 34708552 PMCID: PMC8634767 DOI: 10.1002/open.202100222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/06/2021] [Indexed: 12/17/2022] Open
Abstract
The PtIV prodrug iproplatin has been actively loaded into liposomes using a calcium acetate gradient, achieving a 3-fold enhancement in drug concentration compared to passive loading strategies. A strain-promoted cycloaddition reaction (azide- dibenzocyclooctyne) was used to attach iproplatin-loaded liposomes L(Pt) to gas-filled microbubbles (M), forming an ultrasound-responsive drug delivery vehicle [M-L(Pt)]. Ultrasound-triggered release of iproplatin from the microbubble-liposome construct was evaluated in cellulo. Breast cancer (MCF-7) cells treated with both free iproplatin and iproplatin-loaded liposome-microbubbles [M-L(Pt)] demonstrated an increase in platinum concentration when exposed to ultrasound. No appreciable platinum uptake was observed in MCF-7 cells following treatment with L(Pt) only or L(Pt)+ultrasound, suggesting that microbubble-mediated ultrasonic release of platinum-based drugs from liposomal carriers enables greater control over drug delivery.
Collapse
Affiliation(s)
- Richard Browning
- Institute of Biomedical EngineeringUniversity of OxfordOxfordOX3 7DQUK
| | - Nia Thomas
- Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| | - Laura K. Marsh
- Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| | - Louise R. Tear
- Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| | - Joshua Owen
- Institute of Biomedical EngineeringUniversity of OxfordOxfordOX3 7DQUK
| | - Eleanor Stride
- Institute of Biomedical EngineeringUniversity of OxfordOxfordOX3 7DQUK
| | - Nicola J. Farrer
- Chemistry Research LaboratoryUniversity of Oxford12 Mansfield RoadOxfordOX1 3TAUK
| |
Collapse
|
18
|
Lea-Banks H, Hynynen K. Sub-millimetre precision of drug delivery in the brain from ultrasound-triggered nanodroplets. J Control Release 2021; 338:731-741. [PMID: 34530050 DOI: 10.1016/j.jconrel.2021.09.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/17/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022]
Abstract
Drug-loaded nanoscale cavitation agents, called nanodroplets, are an attractive solution to enhance and localize drug delivery, offering increased stability and prolonged half-life in circulation compared to microbubbles. However, the spatial precision with which drug can be released and delivered into brain tissue from such agents has not been directly mapped. Decafluorobutane lipid-shell droplets (206 +/- 6 nm) were loaded with a fluorescent blood-brain barrier (BBB)-penetrating dye (Nile Blue) and vaporized with ultrasound (1.66 MHz, 10 ms pulse length, 1 Hz pulse repetition frequency), generating transient echogenic microbubbles and delivering the encapsulated dye. The distribution and intensity of released fluorophore was mapped in a tissue-mimicking phantom, and in the brain of rats (Sprague Dawley, N = 4, n = 16). The release and distribution of dye was found to be pressure-dependent (0.2-3.5 MPa) and to occur only above the vaporization threshold of the nanodroplets (1.5 +/- 0.25 MPa in vitro, 2.4 +/- 0.05 MPa in vivo). Dye delivery was achieved with sub-millimetre spatial precision, covering an area of 0.4 to 1.5 mm in diameter, determined by the sonication pressure. The distribution and intensity of dye released at depth in the brain followed the axial pressure profile of the ultrasound beam. Nile Blue (354 Da, LogP 2.7) was compared to Nile Red (318 Da, LogP 3.8) and Quantum Dots (CdSe/ZnS, 5 nm diameter) to visualize the role of molecule size and lipophilicity in crossing the intact BBB following triggered release. Acoustic emissions were shown to predict the successful delivery of the BBB-penetrating dye and the extent of the distribution, demonstrating the theranostic capabilities of nanoscale droplets to precisely localize drug delivery in the brain.
Collapse
Affiliation(s)
- Harriet Lea-Banks
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada.
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
19
|
Ho YJ, Huang CC, Fan CH, Liu HL, Yeh CK. Ultrasonic technologies in imaging and drug delivery. Cell Mol Life Sci 2021; 78:6119-6141. [PMID: 34297166 PMCID: PMC11072106 DOI: 10.1007/s00018-021-03904-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Ultrasonic technologies show great promise for diagnostic imaging and drug delivery in theranostic applications. The development of functional and molecular ultrasound imaging is based on the technical breakthrough of high frame-rate ultrasound. The evolution of shear wave elastography, high-frequency ultrasound imaging, ultrasound contrast imaging, and super-resolution blood flow imaging are described in this review. Recently, the therapeutic potential of the interaction of ultrasound with microbubble cavitation or droplet vaporization has become recognized. Microbubbles and phase-change droplets not only provide effective contrast media, but also show great therapeutic potential. Interaction with ultrasound induces unique and distinguishable biophysical features in microbubbles and droplets that promote drug loading and delivery. In particular, this approach demonstrates potential for central nervous system applications. Here, we systemically review the technological developments of theranostic ultrasound including novel ultrasound imaging techniques, the synergetic use of ultrasound with microbubbles and droplets, and microbubble/droplet drug-loading strategies for anticancer applications and disease modulation. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theranostic tool.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
20
|
Tandon A, Singh SJ, Chaturvedi RK. Nanomedicine against Alzheimer's and Parkinson's Disease. Curr Pharm Des 2021; 27:1507-1545. [PMID: 33087025 DOI: 10.2174/1381612826666201021140904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's and Parkinson's are the two most rampant neurodegenerative disorders worldwide. Existing treatments have a limited effect on the pathophysiology but are unable to fully arrest the progression of the disease. This is due to the inability of these therapeutic molecules to efficiently cross the blood-brain barrier. We discuss how nanotechnology has enabled researchers to develop novel and efficient nano-therapeutics against these diseases. The development of nanotized drug delivery systems has permitted an efficient, site-targeted, and controlled release of drugs in the brain, thereby presenting a revolutionary therapeutic approach. Nanoparticles are also being thoroughly studied and exploited for their role in the efficient and precise diagnosis of neurodegenerative conditions. We summarize the role of different nano-carriers and RNAi-conjugated nanoparticle-based therapeutics for their efficacy in pre-clinical studies. We also discuss the challenges underlying the use of nanomedicine with a focus on their route of administration, concentration, metabolism, and any toxic effects for successful therapeutics in these diseases.
Collapse
Affiliation(s)
- Ankit Tandon
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Sangh J Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Rajnish K Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| |
Collapse
|
21
|
Jangjou A, Meisami AH, Jamali K, Niakan MH, Abbasi M, Shafiee M, Salehi M, Hosseinzadeh A, Amani AM, Vaez A. The promising shadow of microbubble over medical sciences: from fighting wide scope of prevalence disease to cancer eradication. J Biomed Sci 2021; 28:49. [PMID: 34154581 PMCID: PMC8215828 DOI: 10.1186/s12929-021-00744-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Microbubbles are typically 0.5-10 μm in size. Their size tends to make it easier for medication delivery mechanisms to navigate the body by allowing them to be swallowed more easily. The gas included in the microbubble is surrounded by a membrane that may consist of biocompatible biopolymers, polymers, surfactants, proteins, lipids, or a combination thereof. One of the most effective implementation techniques for tiny bubbles is to apply them as a drug carrier that has the potential to activate ultrasound (US); this allows the drug to be released by US. Microbubbles are often designed to preserve and secure medicines or substances before they have reached a certain area of concern and, finally, US is used to disintegrate microbubbles, triggering site-specific leakage/release of biologically active drugs. They have excellent therapeutic potential in a wide range of common diseases. In this article, we discussed microbubbles and their advantageous medicinal uses in the treatment of certain prevalent disorders, including Parkinson's disease, Alzheimer's disease, cardiovascular disease, diabetic condition, renal defects, and finally, their use in the treatment of various forms of cancer as well as their incorporation with nanoparticles. Using microbubble technology as a novel carrier, the ability to prevent and eradicate prevalent diseases has strengthened the promise of effective care to improve patient well-being and life expectancy.
Collapse
Affiliation(s)
- Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Hossein Meisami
- Department of Emergency Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kazem Jamali
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Niakan
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Shafiee
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Hosseinzadeh
- Thoracic and Vascular Surgery Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
Ruan JL, Browning RJ, Yildiz YO, Bau L, Kamila S, Gray MD, Folkes L, Hampson A, McHale AP, Callan JF, Vojnovic B, Kiltie AE, Stride E. Evaluation of Loading Strategies to Improve Tumor Uptake of Gemcitabine in a Murine Orthotopic Bladder Cancer Model Using Ultrasound and Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1596-1615. [PMID: 33707089 DOI: 10.1016/j.ultrasmedbio.2021.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
In this study we compared three different microbubble-based approaches to the delivery of a widely used chemotherapy drug, gemcitabine: (i) co-administration of gemcitabine and microbubbles (Gem+MB); (ii) conjugates of microbubbles and gemcitabine-loaded liposomes (GemlipoMB); and (iii) microbubbles with gemcitabine directly bound to their surfaces (GembioMB). Both in vitro and in vivo investigations were carried out, respectively, in the RT112 bladder cancer cell line and in a murine orthotopic muscle-invasive bladder cancer model. The in vitro (in vivo) ultrasound exposure conditions were a 1 (1.1) MHz centre frequency, 0.07 (1.0) MPa peak negative pressure, 3000 (20,000) cycles and 100 (0.5) Hz pulse repetition frequency. Ultrasound exposure produced no significant increase in drug uptake either in vitro or in vivo compared with the drug-only control for co-administered gemcitabine and microbubbles. In vivo, GemlipoMB prolonged the plasma circulation time of gemcitabine, but only GembioMB produced a statistically significant increase in cleaved caspase 3 expression in the tumor, indicative of gemcitabine-induced apoptosis.
Collapse
Affiliation(s)
- Jia-Ling Ruan
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Richard J Browning
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Yesna O Yildiz
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Luca Bau
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Sukanta Kamila
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Michael D Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Lisa Folkes
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Alix Hampson
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anthony P McHale
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - John F Callan
- Biomedical Sciences Research Institute, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Borivoj Vojnovic
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Anne E Kiltie
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
23
|
Awad N, Paul V, AlSawaftah NM, ter Haar G, Allen TM, Pitt WG, Husseini GA. Ultrasound-Responsive Nanocarriers in Cancer Treatment: A Review. ACS Pharmacol Transl Sci 2021; 4:589-612. [PMID: 33860189 PMCID: PMC8033618 DOI: 10.1021/acsptsci.0c00212] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Indexed: 12/13/2022]
Abstract
The safe and effective delivery of anticancer agents to diseased tissues is one of the significant challenges in cancer therapy. Conventional anticancer agents are generally cytotoxins with poor pharmacokinetics and bioavailability. Nanocarriers are nanosized particles designed for the selectivity of anticancer drugs and gene transport to tumors. They are small enough to extravasate into solid tumors, where they slowly release their therapeutic load by passive leakage or biodegradation. Using smart nanocarriers, the rate of release of the entrapped therapeutic(s) can be increased, and greater exposure of the tumor cells to the therapeutics can be achieved when the nanocarriers are exposed to certain internally (enzymes, pH, and temperature) or externally (light, magnetic field, and ultrasound) applied stimuli that trigger the release of their load in a safe and controlled manner, spatially and temporally. This review gives a comprehensive overview of recent research findings on the different types of stimuli-responsive nanocarriers and their application in cancer treatment with a particular focus on ultrasound.
Collapse
Affiliation(s)
- Nahid
S. Awad
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Nour M. AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Gail ter Haar
- Joint
Department of Physics, The Institute of
Cancer Research and The Royal Marsden NHS Foundation Trust, London SM2 5NG, U.K.
| | - Theresa M. Allen
- Department
of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - William G. Pitt
- Department
of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
24
|
Jia C, Shi J, Han T, Yu ACH, Qin P. Plasma Membrane Blebbing Dynamics Involved in the Reversibly Perforated Cell by Ultrasound-Driven Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:733-750. [PMID: 33358511 DOI: 10.1016/j.ultrasmedbio.2020.11.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
The perforation of plasma membrane by ultrasound-driven microbubbles (i.e., sonoporation) provides a temporary window for transporting macromolecules into the cytoplasm that is promising with respect to drug delivery and gene therapy. To improve the efficacy of delivery while ensuring biosafety, membrane resealing and cell recovery are required to help sonoporated cells defy membrane injury and regain their normal function. Blebs are found to accompany the recovery of sonoporated cells. However, the spatiotemporal characteristics of blebs and the underlying mechanisms remain unclear. With a customized platform for ultrasound exposure and 2-D/3-D live single-cell imaging, localized membrane perforation was induced with ultrasound-driven microbubbles, and the cellular responses were monitored using multiple fluorescent probes. The results indicated that localized blebs undergoing four phases (nucleation, expansion, pausing and retraction) on a time scale of tens of seconds to minutes were specifically involved in the reversibly sonoporated cells. The blebs spatially correlated with the membrane perforation site and temporally lagged (about tens of seconds to minutes) the resealing of perforated membrane. Their diameter (about several microns) and lifetime (about tens of seconds to minutes) positively correlated with the degree of sonoporation. Further studies revealed that intracellular calcium transients might be an upstream signal for triggering blebbing nucleation; exocytotic lysosomes not only contributed to resealing of the perforated membrane, but also to the increasing bleb volume during expansion; and actin components accumulation facilitated bleb retraction. These results provide new insight into the short-term strategies that the sonoporated cell employs to recover on membrane perforation and to remodel membrane structure and a biophysical foundation for sonoporation-based therapy.
Collapse
Affiliation(s)
- Caixia Jia
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianmin Shi
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Han
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Alfred C H Yu
- Schlegel Research Institute for Aging, University of Waterloo, Waterloo, ON, Canada
| | - Peng Qin
- Department of Instrument Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
25
|
Abou-Saleh RH, Delaney A, Ingram N, Batchelor DVB, Johnson BRG, Charalambous A, Bushby RJ, Peyman SA, Coletta PL, Markham AF, Evans SD. Freeze-Dried Therapeutic Microbubbles: Stability and Gas Exchange. ACS APPLIED BIO MATERIALS 2020; 3:7840-7848. [DOI: 10.1021/acsabm.0c00982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Radwa H. Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
- Biophysics Group, Department of Physics, Faculty of Science, Mansoura University, Mansoura 35511, Egypt
| | - Aileen Delaney
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
| | - Nicola Ingram
- Leeds Institute of Medical Research, Wellcome Trust Brenner
Building, St. James’s University Hospital, Leeds LS9 7TF, U.K
| | - Damien V. B. Batchelor
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
| | - Benjamin R. G. Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
| | - Antonia Charalambous
- Leeds Institute of Medical Research, Wellcome Trust Brenner
Building, St. James’s University Hospital, Leeds LS9 7TF, U.K
| | - Richard J. Bushby
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
- School of Chemistry, University of Leeds, Leeds LS2 9JT, U.K
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
- Leeds Institute of Medical Research, Wellcome Trust Brenner
Building, St. James’s University Hospital, Leeds LS9 7TF, U.K
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Wellcome Trust Brenner
Building, St. James’s University Hospital, Leeds LS9 7TF, U.K
| | - Alexander F. Markham
- Leeds Institute of Medical Research, Wellcome Trust Brenner
Building, St. James’s University Hospital, Leeds LS9 7TF, U.K
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, U.K
| |
Collapse
|
26
|
Recent advances in ultrasound-triggered drug delivery through lipid-based nanomaterials. Drug Discov Today 2020; 25:2182-2200. [PMID: 33010479 DOI: 10.1016/j.drudis.2020.09.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/24/2020] [Accepted: 09/22/2020] [Indexed: 12/28/2022]
Abstract
The high prescribed dose of anticancer drugs and their resulting adverse effects on healthy tissue are significant drawbacks to conventional chemotherapy (CTP). Ideally, drugs should have the lowest possible degree of interaction with healthy cells, which would diminish any adverse effects. Therefore, an ideal scenario to bring about improvements in CTP is the use of technological strategies to ensure the efficient, specific, and selective transport and/or release of drugs to the target site. One practical and feasible solution to promote the efficiency of conventional CTP is the use of ultrasound (US). In this review, we highlight the potential role of US in combination with lipid-based carriers to achieve a targeted CTP strategy in engineered smart drug delivery systems.
Collapse
|
27
|
Ozdas MS, Shah AS, Johnson PM, Patel N, Marks M, Yasar TB, Stalder U, Bigler L, von der Behrens W, Sirsi SR, Yanik MF. Non-invasive molecularly-specific millimeter-resolution manipulation of brain circuits by ultrasound-mediated aggregation and uncaging of drug carriers. Nat Commun 2020; 11:4929. [PMID: 33004789 PMCID: PMC7529901 DOI: 10.1038/s41467-020-18059-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
Non-invasive, molecularly-specific, focal modulation of brain circuits with low off-target effects can lead to breakthroughs in treatments of brain disorders. We systemically inject engineered ultrasound-controllable drug carriers and subsequently apply a novel two-component Aggregation and Uncaging Focused Ultrasound Sequence (AU-FUS) at the desired targets inside the brain. The first sequence aggregates drug carriers with millimeter-precision by orders of magnitude. The second sequence uncages the carrier's cargo locally to achieve high target specificity without compromising the blood-brain barrier (BBB). Upon release from the carriers, drugs locally cross the intact BBB. We show circuit-specific manipulation of sensory signaling in motor cortex in rats by locally concentrating and releasing a GABAA receptor agonist from ultrasound-controlled carriers. Our approach uses orders of magnitude (1300x) less drug than is otherwise required by systemic injection and requires very low ultrasound pressures (20-fold below FDA safety limits for diagnostic imaging). We show that the BBB remains intact using passive cavitation detection (PCD), MRI-contrast agents and, importantly, also by sensitive fluorescent dye extravasation and immunohistochemistry.
Collapse
Affiliation(s)
- Mehmet S Ozdas
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Aagam S Shah
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland. .,Neuroscience Center, Zurich, Switzerland.
| | - Paul M Johnson
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Nisheet Patel
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland
| | - Markus Marks
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Tansel Baran Yasar
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Urs Stalder
- Department of Chemistry, UZH, Zurich, Switzerland
| | | | - Wolfger von der Behrens
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Neuroscience Center, Zurich, Switzerland
| | - Shashank R Sirsi
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland.,Department of Bioengineering, UT at Dallas, Richardson, USA
| | - Mehmet Fatih Yanik
- Institute of Neuroinformatics, D-ITET, ETH Zurich and UZH, Zurich, Switzerland. .,Neuroscience Center, Zurich, Switzerland.
| |
Collapse
|
28
|
Ingram N, McVeigh LE, Abou-Saleh RH, Maynard J, Peyman SA, McLaughlan JR, Fairclough M, Marston G, Valleley EMA, Jimenez-Macias JL, Charalambous A, Townley W, Haddrick M, Wierzbicki A, Wright A, Volpato M, Simpson PB, Treanor DE, Thomson NH, Loadman PM, Bushby RJ, Johnson BR, Jones PF, Evans JA, Freear S, Markham AF, Evans SD, Coletta PL. Ultrasound-triggered therapeutic microbubbles enhance the efficacy of cytotoxic drugs by increasing circulation and tumor drug accumulation and limiting bioavailability and toxicity in normal tissues. Theranostics 2020; 10:10973-10992. [PMID: 33042265 PMCID: PMC7532679 DOI: 10.7150/thno.49670] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Most cancer patients receive chemotherapy at some stage of their treatment which makes improving the efficacy of cytotoxic drugs an ongoing and important goal. Despite large numbers of potent anti-cancer agents being developed, a major obstacle to clinical translation remains the inability to deliver therapeutic doses to a tumor without causing intolerable side effects. To address this problem, there has been intense interest in nanoformulations and targeted delivery to improve cancer outcomes. The aim of this work was to demonstrate how vascular endothelial growth factor receptor 2 (VEGFR2)-targeted, ultrasound-triggered delivery with therapeutic microbubbles (thMBs) could improve the therapeutic range of cytotoxic drugs. Methods: Using a microfluidic microbubble production platform, we generated thMBs comprising VEGFR2-targeted microbubbles with attached liposomal payloads for localised ultrasound-triggered delivery of irinotecan and SN38 in mouse models of colorectal cancer. Intravenous injection into tumor-bearing mice was used to examine targeting efficiency and tumor pharmacodynamics. High-frequency ultrasound and bioluminescent imaging were used to visualise microbubbles in real-time. Tandem mass spectrometry (LC-MS/MS) was used to quantitate intratumoral drug delivery and tissue biodistribution. Finally, 89Zr PET radiotracing was used to compare biodistribution and tumor accumulation of ultrasound-triggered SN38 thMBs with VEGFR2-targeted SN38 liposomes alone. Results: ThMBs specifically bound VEGFR2 in vitro and significantly improved tumor responses to low dose irinotecan and SN38 in human colorectal cancer xenografts. An ultrasound trigger was essential to achieve the selective effects of thMBs as without it, thMBs failed to extend intratumoral drug delivery or demonstrate enhanced tumor responses. Sensitive LC-MS/MS quantification of drugs and their metabolites demonstrated that thMBs extended drug exposure in tumors but limited exposure in healthy tissues, not exposed to ultrasound, by persistent encapsulation of drug prior to elimination. 89Zr PET radiotracing showed that the percentage injected dose in tumors achieved with thMBs was twice that of VEGFR2-targeted SN38 liposomes alone. Conclusions: thMBs provide a generic platform for the targeted, ultrasound-triggered delivery of cytotoxic drugs by enhancing tumor responses to low dose drug delivery via combined effects on circulation, tumor drug accumulation and exposure and altered metabolism in normal tissues.
Collapse
Affiliation(s)
- Nicola Ingram
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Laura E. McVeigh
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Radwa H. Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- Department of Physics, Faculty of Science, Mansoura University, Egypt
| | - Juliana Maynard
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Sally A. Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - James R. McLaughlan
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Michael Fairclough
- Wolfson Molecular Imaging Centre, University of Manchester, Palatine Road, Manchester, M20 3LI, United Kingdom
| | - Gemma Marston
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Elizabeth M. A. Valleley
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Jorge L. Jimenez-Macias
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Antonia Charalambous
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - William Townley
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Malcolm Haddrick
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Antonia Wierzbicki
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Alexander Wright
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Milène Volpato
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Peter B. Simpson
- Medicines Discovery Catapult, Mereside, Alderley Park, Macclesfield, SK10 4TG, United Kingdom
| | - Darren E. Treanor
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Neil H. Thomson
- School of Dentistry, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, BD7 1DP, United Kingdom
| | - Richard J. Bushby
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Benjamin R.G. Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - Pamela F. Jones
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - J. Anthony Evans
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Steven Freear
- Faculty of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Alexander F. Markham
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| | - Stephen D. Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, LS9 7TF, United Kingdom
| |
Collapse
|
29
|
Chandan R, Mehta S, Banerjee R. Ultrasound-Responsive Carriers for Therapeutic Applications. ACS Biomater Sci Eng 2020; 6:4731-4747. [PMID: 33455210 DOI: 10.1021/acsbiomaterials.9b01979] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ultrasound (US)-responsive carriers have emerged as promising theranostic candidates because of their ability to enhance US-contrast, promote image-guided drug delivery, cause on-demand pulsatile release of drugs in response to ultrasound stimuli, as well as to enhance the permeability of physiological barriers such as the stratum corneum, the vascular endothelium, and the blood-brain barrier (BBB). US-responsive carriers include microbubbles MBs, liposomes, droplets, hydrogels, and nanobubble-nanoparticle complexes and have been explored for cavitation-mediated US-responsive drug delivery. Recently, a transient increase in the permeability of the BBB by microbubble (MB)-assisted low-frequency US has shown promise in enhancing the delivery of therapeutic agents in the case of neurological disorders. Further, the periodic mechanical stimulus generated by US-responsive MBs have also been explored in tissue engineering and has directly influenced the differentiation of mesenchymal stem cells into cartilage. This Review discusses the various types of US-responsive carriers and explores their emerging roles in therapeutics ranging from drug delivery to tissue engineering.
Collapse
Affiliation(s)
- Rajeet Chandan
- Nanomedicine Lab, Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sourabh Mehta
- Nanomedicine Lab, Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.,IIT Bombay-Monash Research Academy, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rinti Banerjee
- Nanomedicine Lab, Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
30
|
Stride E, Segers T, Lajoinie G, Cherkaoui S, Bettinger T, Versluis M, Borden M. Microbubble Agents: New Directions. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1326-1343. [PMID: 32169397 DOI: 10.1016/j.ultrasmedbio.2020.01.027] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 05/24/2023]
Abstract
Microbubble ultrasound contrast agents have now been in use for several decades and their safety and efficacy in a wide range of diagnostic applications have been well established. Recent progress in imaging technology is facilitating exciting developments in techniques such as molecular, 3-D and super resolution imaging and new agents are now being developed to meet their specific requirements. In parallel, there have been significant advances in the therapeutic applications of microbubbles, with recent clinical trials demonstrating drug delivery across the blood-brain barrier and into solid tumours. New agents are similarly being tailored toward these applications, including nanoscale microbubble precursors offering superior circulation times and tissue penetration. The development of novel agents does, however, present several challenges, particularly regarding the regulatory framework. This article reviews the developments in agents for diagnostic, therapeutic and "theranostic" applications; novel manufacturing techniques; and the opportunities and challenges for their commercial and clinical translation.
Collapse
Affiliation(s)
- Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| | - Tim Segers
- Physics of Fluids Group, Technical Medical (TechMed) Centre, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | - Guillaume Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Centre, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | - Samir Cherkaoui
- Bracco Suisse SA - Business Unit Imaging, Global R&D, Plan-les-Ouates, Switzerland
| | - Thierry Bettinger
- Bracco Suisse SA - Business Unit Imaging, Global R&D, Plan-les-Ouates, Switzerland
| | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Centre, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | - Mark Borden
- Mechanical Engineering Department, University of Colorado, Boulder, CO, USA
| |
Collapse
|
31
|
Dwivedi P, Kiran S, Han S, Dwivedi M, Khatik R, Fan R, Mangrio FA, Du K, Zhu Z, Yang C, Huang F, Ejaz A, Han R, Si T, Xu RX. Magnetic Targeting and Ultrasound Activation of Liposome-Microbubble Conjugate for Enhanced Delivery of Anticancer Therapies. ACS APPLIED MATERIALS & INTERFACES 2020; 12:23737-23751. [PMID: 32374147 DOI: 10.1021/acsami.0c05308] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Effective delivery of chemotherapeutics with minimal toxicity and maximal outcome is clinically important but technically challenging. Here, we synthesize a complex of doxorubicin (DOX)-loaded magneto-liposome (DOX-ML) microbubbles (DOX-ML-MBs) for magnetically responsive and ultrasonically sensitive delivery of anticancer therapies with enhanced efficiency. Citrate-stabilized iron oxide nanoparticles (MNs) of 6.8 ± 1.36 nm were synthesized, loaded with DOX in the core of oligolamellar vesicles of 172 ± 9.2 nm, and covalently conjugated with perfluorocarbon (PFC)-gas-loaded microbubbles to form DOX-ML-MBs of ∼4 μm. DOX-ML-MBs exhibited significant magnetism and were able to release chemotherapeutics and DOX-MLs instantly upon exposure to ultrasound (US) pulses. In vitro studies showed that DOX-ML-MBs in the presence of US pulses promoted apoptosis and were highly effective in killing both BxPc-3 and Panc02 pancreatic cancer cells even at a low dose. Significant reduction in the tumor volume was observed after intravenous administration of DOX-ML-MBs in comparison to the control group in a pancreatic cancer xenograft model of nude mice. Deeply penetrated iron oxide nanoparticles throughout the magnetically targeted tumor tissues in the presence of US stimulation were clearly observed. Our study demonstrated the potential of using DOX-ML-MBs for site-specific targeting and controlled drug release. It opens a new avenue for the treatment of pancreatic cancer and other tissue malignancies where precise delivery of therapeutics is necessary.
Collapse
Affiliation(s)
- Pankaj Dwivedi
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Sonia Kiran
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Shuya Han
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Monika Dwivedi
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Renuka Khatik
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| | - Rong Fan
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Farhana Akbar Mangrio
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Kun Du
- Department of Electronic Science and Technology, University of Science and Technology of China Hefei, Hefei, Anhui 230027, P. R. China
| | - Zhiqiang Zhu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Chaoyu Yang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Fangsheng Huang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Hefei National Laboratory of Physical Sciences at Microscale, Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, P. R. China
| | - Aslam Ejaz
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Renzhi Han
- Department of Surgery, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ting Si
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
| | - Ronald X Xu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, P. R. China
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
32
|
Owen J, Thomas E, Menon J, Gray M, Skaripa-Koukelli I, Gill MR, Wallington S, Miller RL, Vallis KA, Carlisle R. Indium-111 labelling of liposomal HEGF for radionuclide delivery via ultrasound-induced cavitation. J Control Release 2020; 319:222-233. [PMID: 31891732 DOI: 10.1016/j.jconrel.2019.12.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023]
Abstract
The purpose of this exploratory study was to investigate the combination of a radiopharmaceutical, nanoparticles and ultrasound (US) enhanced delivery to develop a clinically viable therapeutic strategy for tumours overexpressing the epidermal growth factor receptor (EGFR). Molecularly targeted radionuclides have great potential for cancer therapy but are sometimes associated with insufficient delivery resulting in sub-cytotoxic amounts of radioactivity being delivered to the tumour. Liposome formulations are currently used in the clinic to reduce the side effects and improve the pharmacokinetic profile of chemotherapeutic drugs. However, in contrast to non-radioactive agents, loading and release of radiotherapeutics from liposomes can be challenging in the clinical setting. US-activated cavitation agents such as microbubbles (MBs) have been used to release therapeutics from liposomes to enhance the distribution/delivery in a target area. In an effort to harness the benefits of these techniques, the development of a liposome loaded radiopharmaceutical construct for enhanced delivery via acoustic cavitation was studied. The liposomal formulation was loaded with peptide, human epidermal growth factor (HEGF), coupled to a chelator for subsequent radiolabelling with 111Indium ([111In]In3+), in a manner designed to be compatible with preparation in a radiopharmacy. Liposomes were efficiently radiolabelled (57%) within 1 h, with release of ~12% of the radiopeptide following a 20 s exposure to US-mediated cavitation in vitro. In clonogenic studies this level of release resulted in cytotoxicity specifically in cells over-expressing the epidermal growth factor receptor (EGFR), with over 99% reduction in colony survival compared to controls. The formulation extended the circulation time and changed the biodistribution compared to the non-liposomal radiopeptide in vivo, although interestingly the biodistribution did not resemble that of liposome constructs currently used in the clinic. Cavitation of MBs co-injected with liposomes into tumours expressing high levels of EGFR resulted in a 2-fold enhancement in tumour uptake within 20 min. However, owing to the poor vascularisation of the tumour model used the same level of uptake was achieved without US after 24 h. By combining acoustic-cavitation-sensitive liposomes with radiopharmaceuticals this research represents a new concept in achieving targeted delivery of radiopharmaceuticals.
Collapse
Affiliation(s)
- Joshua Owen
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Eloise Thomas
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Jyothi Menon
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK; College of Pharmacy, The University of Rhode Island, Kingston, RI 02881, USA
| | - Michael Gray
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Irini Skaripa-Koukelli
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Martin R Gill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Sheena Wallington
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Rebecca L Miller
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus, Oxford OX3 7DQ, UK
| | - Robert Carlisle
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| |
Collapse
|
33
|
Paris JL, Vallet-Regí M. Ultrasound-Activated Nanomaterials for Therapeutics. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020; 93:220-229. [PMID: 39650549 PMCID: PMC7617089 DOI: 10.1246/bcsj.20190346] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Ultrasound has attracted much attention in recent years as an external stimulus capable of activating different types of nanomaterials for therapeutic application. One of the characteristics that makes ultrasound an especially appealing triggering stimulus for nanomedicine is its capacity to be non-invasively applied in a focused manner at deep regions of the body. Combining ultrasound with nanoparticles, different biological effects can be achieved. In this work, an overview of the four main types of inducible responses will be provided: inducing drug release, producing ultrasound-derived biological effects, modifying nanoparticle biodistribution and developing theranostic agents. Several examples of each one of these applications are presented here to illustrate the key concepts underlying recent developments in the discipline.
Collapse
Affiliation(s)
- Juan L. Paris
- Department of Life Sciences, Nano4Health Unit, Nanomedicine Group. International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040-Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
34
|
Omata D, Unga J, Suzuki R, Maruyama K. Lipid-based microbubbles and ultrasound for therapeutic application. Adv Drug Deliv Rev 2020; 154-155:236-244. [PMID: 32659255 DOI: 10.1016/j.addr.2020.07.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/11/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Microbubbles with diagnostic ultrasound have had a long history of use in the medical field. In recent years, the therapeutic application of the combination of microbubbles and ultrasound, called sonoporation, has received increased attention as microbubble oscillation or collapse close to various barriers in the body was recognized to potentially open those barriers, increasing drug transport across them. In this review, we aimed to describe the development of lipid-stabilized microbubbles equipped with functions, such as long circulation and drug loading, and the therapeutic application of sonoporation for tumor-targeted therapy, brain-targeted therapy, and immunotherapy. We also attempted to discuss the current status of the field and potential future developments.
Collapse
|
35
|
Márquez MG, Dotson R, Pias S, Frolova LV, Tartis MS. Phospholipid prodrug conjugates of insoluble chemotherapeutic agents for ultrasound targeted drug delivery. Nanotheranostics 2020; 4:40-56. [PMID: 31911893 PMCID: PMC6940203 DOI: 10.7150/ntno.37738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/01/2019] [Indexed: 12/19/2022] Open
Abstract
The hydrophobicity and high potency of many therapeutic agents makes them difficult to use effectively in clinical practice. This work focuses on conjugating phospholipid tails (2T) onto podophyllotoxin (P) and its analogue (N) using a linker and characterizing the effects of their incorporation into lipid-based drug delivery vehicles for triggered ultrasound delivery. Differential Scanning Calorimetry results show that successfully synthesized lipophilic prodrugs, 2T-P (~28 % yield) and 2T-N(~26 % yield), incorporate within the lipid membranes of liposomes. As a result of this, increased stability and incorporation are observed in 2T-P and 2T-N in comparison to the parent compounds P and N. Molecular dynamic simulation results support that prodrugs remain within the lipid membrane over a relevant range of concentrations. 2T-N's (IC50: 20 nM) biological activity was retained in HeLa cells (cervical cancer), whereas 2T-P's (IC50: ~4 µM) suffered, presumably due to steric hindrance. Proof-of-concept studies using ultrasound in vitro microbubble and nanodroplet delivery vehicles establish that these prodrugs are capable of localized drug delivery. This study provides useful information about the synthesis of double tail analogues of insoluble chemotherapeutic agents to facilitate incorporation into drug delivery vehicles. The phospholipid attachment strategy presented here could be applied to other well suited drugs such as gemcitabine, commonly known for its treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Mendi G Márquez
- Materials Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA.,Chemical Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Rachel Dotson
- Departments of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Sally Pias
- Departments of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Liliya V Frolova
- Departments of Chemistry, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| | - Michaelann S Tartis
- Materials Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA.,Chemical Engineering, New Mexico Institute of Mining and Technology, 801 Leroy Place, Socorro, NM 87801, USA
| |
Collapse
|
36
|
Ding J, Chen J, Gao L, Jiang Z, Zhang Y, Li M, Xiao Q, Lee SS, Chen X. Engineered nanomedicines with enhanced tumor penetration. NANO TODAY 2019; 29:100800. [DOI: 10.1016/j.nantod.2019.100800] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
37
|
Roovers S, Deprez J, Priwitaningrum D, Lajoinie G, Rivron N, Declercq H, De Wever O, Stride E, Le Gac S, Versluis M, Prakash J, De Smedt SC, Lentacker I. Sonoprinting liposomes on tumor spheroids by microbubbles and ultrasound. J Control Release 2019; 316:79-92. [PMID: 31676384 DOI: 10.1016/j.jconrel.2019.10.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/24/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022]
Abstract
Ultrasound-triggered drug-loaded microbubbles have great potential for drug delivery due to their ability to locally release drugs and simultaneously enhance their delivery into the target tissue. We have recently shown that upon applying ultrasound, nanoparticle-loaded microbubbles can deposit nanoparticles onto cells grown in 2D monolayers, through a process that we termed "sonoprinting". However, the rigid surfaces on which cell monolayers are typically growing might be a source of acoustic reflections and aspherical microbubble oscillations, which can influence microbubble-cell interactions. In the present study, we aim to reveal whether sonoprinting can also occur in more complex and physiologically relevant tissues, by using free-floating 3D tumor spheroids as a tissue model. We show that both monospheroids (consisting of tumor cells alone) and cospheroids (consisting of tumor cells and fibroblasts, which produce an extracellular matrix) can be sonoprinted. Using doxorubicin-liposome-loaded microbubbles, we show that sonoprinting allows to deposit large amounts of doxorubicin-containing liposomes to the outer cell layers of the spheroids, followed by doxorubicin release into the deeper layers of the spheroids, resulting in a significant reduction in cell viability. Sonoprinting may become an attractive approach to deposit drug patches at the surface of tissues, thereby promoting the delivery of drugs into target tissues.
Collapse
Affiliation(s)
- S Roovers
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - J Deprez
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - D Priwitaningrum
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - N Rivron
- Institute of Molecular Biotechnology, Austrian Academy of Sciences, Vienna, Austria
| | - H Declercq
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Tissue Engineering Group, Department of Human Structure and Repair, Ghent University, Belgium
| | - O De Wever
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Laboratory Experimental Cancer Research (LECR), Ghent University, Ghent, Belgium
| | - E Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - S Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - M Versluis
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - J Prakash
- Targeted Therapeutics, Department of Biomaterials Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - S C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicine, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
38
|
Cheng CA, Chen W, Zhang L, Wu HH, Zink JI. A Responsive Mesoporous Silica Nanoparticle Platform for Magnetic Resonance Imaging-Guided High-Intensity Focused Ultrasound-Stimulated Cargo Delivery with Controllable Location, Time, and Dose. J Am Chem Soc 2019; 141:17670-17684. [PMID: 31604010 DOI: 10.1021/jacs.9b07591] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Magnetic resonance imaging (MRI) is an essential modality for clinical diagnosis, and MRI-guided high-intensity focused ultrasound (MRgHIFU) is a powerful technology for targeted therapy. Clinical applications of MRgHIFU primarily utilize hyperthermia and ablation to treat cancerous tissue, but for drug delivery applications thermal damage is undesirable. A biofriendly MRgHIFU-responsive mesoporous silica nanoparticle (MSN) platform that is stimulated within a physiological safe temperature range has been developed, reducing the possibility of thermal damage to the surrounding healthy tissues. Biocompatible polyethylene glycol (PEG) was employed to cap the pores of MSNs, and the release of cargo molecules by HIFU occurs without substantial temperature increase (∼4 °C). To visualize by MRI and measure the stimulated delivery in situ, a U.S. Food and Drug Administration (FDA)-approved gadolinium-based contrast agent, gadopentetate dimeglumine (Gd(DTPA)2-), was used as the imageable cargo. Taking advantage of the three-dimensional (3-D) imaging and targeting capabilities of MRgHIFU, the release of Gd(DTPA)2- stimulated by HIFU was pinpointed at the HIFU focal point in 3-D space in a tissue-mimicking gel phantom. The amount of Gd(DTPA)2- released was controlled by HIFU stimulation times and power levels. A positive correlation between the amount of Gd(DTPA)2- released and T1 was found. The MRgHIFU-stimulated cargo release was further imaged in a sample of ex vivo animal tissue. With this technology, the biodistribution of the nanocarriers can be tracked and the MRgHIFU-stimulated cargo release can be pinpointed, opening up an opportunity for future image-guided theranostic applications.
Collapse
Affiliation(s)
- Chi-An Cheng
- Department of Bioengineering , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| | - Wei Chen
- Department of Chemistry & Biochemistry , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| | - Le Zhang
- Department of Radiological Sciences, David Geffen School of Medicine , University of California Los Angeles , Los Angeles , California 90095 , United States
| | - Holden H Wu
- Department of Bioengineering , University of California Los Angeles , Los Angeles , California 90095 , United States.,Department of Radiological Sciences, David Geffen School of Medicine , University of California Los Angeles , Los Angeles , California 90095 , United States
| | - Jeffrey I Zink
- Department of Chemistry & Biochemistry , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| |
Collapse
|
39
|
Dhaliwal A, Zheng G. Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: Designing a new perspective in nanomedicine delivery. Theranostics 2019; 9:8091-8108. [PMID: 31754383 PMCID: PMC6857058 DOI: 10.7150/thno.37204] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
The enhanced permeability and retention (EPR) effect has underlain the predominant nanomedicine design philosophy for the past three decades. However, growing evidence suggests that it is over-represented in preclinical models, and agents designed solely using its principle of passive accumulation can only be applied to a narrow subset of clinical tumors. For this reason, strategies that can improve upon the EPR effect to facilitate nanomedicine delivery to otherwise non-responsive tumors are required for broad clinical translation. EPR-adaptive nanomedicine delivery comprises a class of chemical and physical techniques that modify tumor accessibility in an effort to increase agent delivery and therapeutic effect. In the present review, we overview the primary benefits and limitations of radiation, ultrasound, hyperthermia, and photodynamic therapy as physical strategies for EPR-adaptive delivery to EPR-insensitive tumor phenotypes, and we reflect upon changes in the preclinical research pathway that should be implemented in order to optimally validate and develop these delivery strategies.
Collapse
Affiliation(s)
- Alexander Dhaliwal
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- MD/PhD Program, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON Canada
| |
Collapse
|
40
|
Drug-Loaded Microbubbles Combined with Ultrasound for Thrombolysis and Malignant Tumor Therapy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6792465. [PMID: 31662987 PMCID: PMC6791276 DOI: 10.1155/2019/6792465] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/22/2019] [Accepted: 09/14/2019] [Indexed: 12/14/2022]
Abstract
Cardiac-cerebral thrombosis and malignant tumor endanger the safety of human life seriously. Traditional chemotherapy drugs have side effects which restrict their applications. Drug-loaded microbubbles can be destroyed by ultrasound irradiation at the focus position and be used for thrombolysis and tumor therapy. Compared with traditional drug treatment, the drug-loaded microbubbles can be excited by ultrasound and release drugs to lesion sites, increasing the local drug concentration and the exposure dose to nonfocal regions, thus reducing the cytotoxicity and side effects of drugs. This article reviews the applications of drug-loaded microbubbles combined with ultrasound for thrombolysis and tumor therapy. We focus on highlighting the advantages of using this new technique for disease treatment and concluding with recommendations for future efforts on the applications of this technology.
Collapse
|
41
|
Nittayacharn P, Yuan HX, Hernandez C, Bielecki P, Zhou H, Exner AA. Enhancing Tumor Drug Distribution With Ultrasound-Triggered Nanobubbles. J Pharm Sci 2019; 108:3091-3098. [PMID: 31095958 PMCID: PMC6708467 DOI: 10.1016/j.xphs.2019.05.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/30/2019] [Accepted: 05/07/2019] [Indexed: 01/09/2023]
Abstract
Issues with limited intratumoral drug penetration and heterogeneous drug distribution continue to impede the therapeutic efficacy of nanomedicine-based delivery systems. Ultrasound (US)-enhanced drug delivery has emerged as one effective means of overcoming these challenges. Acoustic cavitation in the presence of nanoparticles has shown to increase the cellular uptake and distribution of chemotherapeutic agents in vivo. In this study, we investigated the potential of a drug-loaded echogenic nanoscale bubbles in combination with low frequency (3 MHz), high energy (2 W/cm2) US for antitumor therapy. The doxorubicin-loaded nanobubbles (Dox-NBs) stabilized with an interpenetrating polymer mesh were 171.5 ± 20.9 nm in diameter. When used in combination with therapeutic US, Dox-NBs combined with free drug showed significantly higher (*p < 0.05) intracellular uptake and therapeutic efficacy compared with free drug. When injected intravenously in vivo, Dox-NBs + therapeutic US showed significantly higher (*p < 0.05) accumulation and better distribution of Dox in tumors when compared with free drug. This strategy provides an effective and simple method to increase the local dose and distribution of otherwise systemically toxic chemotherapeutic agents for cancer therapies.
Collapse
Affiliation(s)
- Pinunta Nittayacharn
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Hai-Xia Yuan
- Department of Ultrasound, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai 200032, China; Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Christopher Hernandez
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Peter Bielecki
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Haoyan Zhou
- GSK 1250 S. Collegeville Road, Collegeville, Pennsylvania 19426-0989
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106; Department of Radiology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106.
| |
Collapse
|
42
|
Prasad C, Banerjee R. Ultrasound-Triggered Spatiotemporal Delivery of Topotecan and Curcumin as Combination Therapy for Cancer. J Pharmacol Exp Ther 2019; 370:876-893. [PMID: 30988010 DOI: 10.1124/jpet.119.256487] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/08/2019] [Indexed: 08/30/2023] Open
Abstract
Chemotherapy is limited by the low availability of drug at the tumor site and drug resistance by the tumor. In this report we describe a combination therapy for codelivery of two anticancer drugs with spatiotemporal control by ultrasound pulses. We developed curcumin and topotecan-coencapsulated nanoconjugates Cur_Tpt_NC.MB to have an ultrasound contrast property. MDA MB 231 and B16F10 cells were incubated with Cur_Tpt_NC.MB and exposed to ultrasound. Ultrasound exposure reduced the IC50 concentration of topotecan and curcumin significantly (P < 0.05) compared with free drug. Antitumor efficacy study of the Cur_Tpt_NC.MB in B16F10 melanoma tumor-bearing C57BL/6 mice showed that ultrasound exposure of right tumor reduced growth by 3.5 times compared with the unexposed left tumor of same mice and 14.8 times compared with a group treated with a physical mixture of drugs. These results suggest that the nanotherapeutic system we developed induces site-specific inhibition of tumor growth at a high rate and has the potential to be used as a therapeutic regimen for spatiotemporal delivery of dual drugs for treatment of cancer.
Collapse
Affiliation(s)
- Chandrashekhar Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Mumbai, India
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Mumbai, India
| |
Collapse
|
43
|
Hall RL, Juan-Sing ZD, Hoyt K, Sirsi SR. Formulation and Characterization of Chemically Cross-linked Microbubble Clusters. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10977-10986. [PMID: 31310715 PMCID: PMC7061884 DOI: 10.1021/acs.langmuir.9b00475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The purpose of this study is to introduce a new concept of chemically cross-linked microbubble clusters (CCMCs), which are individual microbubble ultrasound contrast agents (UCAs) physically tethered together. We demonstrate a facile means of their production, characterize their size and stability, and describe how they can potentially be used in biomedical applications. By tethering UCAs together into CCMCs, we propose that novel methods of ultrasound mediated imaging and therapy can be developed through unique interbubble interactions in an ultrasound field. One of the major challenges in generating CCMCs is controlling aggregate sizes and maintaining stability against Ostwald ripening and coalescence. In this study, we demonstrate that chemically cross-linked microbubble clusters can produce small (<10 μm) quasi-stable complexes that slowly fuse into bubbles with individual gas cores. Furthermore, we demonstrate that this process can be driven with low-intensity ultrasound pulses, enabling a rapid fusion of clusters which could potentially be used to develop novel ultrasound contrast imaging and drug delivery strategies in future studies. The development of novel microbubble clusters presents a simple yet robust process for generating novel UCAs with a design that could allow for more versatility in contrast-enhanced ultrasound (CEUS), molecular imaging, and drug delivery applications. Additionally, microbubble clustering is a unique way to control size, shell, and gas compositions that can be used to study bubble ripening and coalescence in a highly controlled environment or study the behavior of mixed-microbubble populations.
Collapse
Affiliation(s)
- Ronald L. Hall
- University of Texas at Dallas, Richardson, Texas, 75080, United States
| | | | - Kenneth Hoyt
- University of Texas at Dallas, Richardson, Texas, 75080, United States
- University of Texas Southwestern, Dallas, Texas, 75390, United States
| | - Shashank R. Sirsi
- University of Texas at Dallas, Richardson, Texas, 75080, United States
- University of Texas Southwestern, Dallas, Texas, 75390, United States
| |
Collapse
|
44
|
Escoffre JM, Bouakaz A. Minireview: Biophysical Mechanisms of Cell Membrane Sonopermeabilization. Knowns and Unknowns. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10151-10165. [PMID: 30525655 DOI: 10.1021/acs.langmuir.8b03538] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Microbubble-assisted ultrasound has emerged as a promising method for the delivery of low-molecular-weight chemotherapeutic molecules, nucleic acids, therapeutic peptides, and antibodies in vitro and in vivo. Its clinical applications are under investigation for local delivery drug in oncology and neurology. However, the biophysical mechanisms supporting the acoustically mediated membrane permeabilization are not fully established. This review describes the present state of the investigations concerning the acoustically mediated stimuli (i.e., mechanical, chemical, and thermal stimuli) as well as the molecular and cellular actors (i.e., membrane pores and endocytosis) involved in the reversible membrane permeabilization process. The different hypotheses, which were proposed to give a biophysical description of the membrane permeabilization, are critically discussed.
Collapse
Affiliation(s)
- Jean-Michel Escoffre
- UMR 1253, iBrain, Université de Tours, Inserm , 10 bd Tonnellé , 37032 Tours Cedex 1, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm , 10 bd Tonnellé , 37032 Tours Cedex 1, France
| |
Collapse
|
45
|
Abou-Saleh RH, McLaughlan JR, Bushby RJ, Johnson BR, Freear S, Evans SD, Thomson NH. Molecular Effects of Glycerol on Lipid Monolayers at the Gas-Liquid Interface: Impact on Microbubble Physical and Mechanical Properties. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10097-10105. [PMID: 30901226 DOI: 10.1021/acs.langmuir.8b04130] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The production and stability of microbubbles (MBs) is enhanced by increasing the viscosity of both the formation and storage solution, respectively. Glycerol is a good candidate for biomedical applications of MBs, since it is biocompatible, although the exact molecular mechanisms of its action is not fully understood. Here, we investigate the influence glycerol has on lipid-shelled MB properties, using a range of techniques. Population lifetime and single bubble stability were studied using optical microscopy. Bubble stiffness measured by AFM compression is compared with lipid monolayer behavior in a Langmuir-Blodgett trough. We deduce that increasing glycerol concentrations enhances stability of MB populations through a 3-fold mechanism. First, binding of glycerol to lipid headgroups in the interfacial monolayer up to 10% glycerol increases MB stiffness but has limited impact on shell resistance to gas permeation and corresponding MB lifetime. Second, increased solution viscosity above 10% glycerol slows down the kinetics of gas transfer, markedly increasing MB stability. Third, above 10%, glycerol induces water structuring around the lipid monolayer, forming a glassy layer which also increases MB stiffness and resistance to gas loss. At 30% glycerol, the glassy layer is ablated, lowering the MB stiffness, but MB stability is further augmented. Although the molecular interactions of glycerol with the lipid monolayer modulate the MB lipid shell properties, MB lifetime continually increases from 0 to 30% glycerol, indicating that its viscosity is the dominant effect on MB solution stability. This three-fold action and biocompatibility makes glycerol ideal for therapeutic MB formation and storage and gives new insight into the action of glycerol on lipid monolayers at the gas-liquid interface.
Collapse
Affiliation(s)
- Radwa H Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
- Biophysics Group, Department of Physics, Faculty of Science , Mansoura University , Mansoura , Egypt
| | - James R McLaughlan
- School of Electronic and Electrical Engineering , University of Leeds , Leeds LS2 9JT , United Kingdom
- Leeds Institute of Medical Research , University of Leeds, St. James's University Hospital , Leeds LS9 7TF , United Kingdom
| | - Richard J Bushby
- School of Chemistry , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Benjamin R Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Steven Freear
- School of Electronic and Electrical Engineering , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Stephen D Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
| | - Neil H Thomson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy , University of Leeds , Leeds LS2 9JT , United Kingdom
- Division of Oral Biology, School of Dentistry , University of Leeds , Leeds LS2 9LU , United Kingdom
| |
Collapse
|
46
|
Yildirim A, Blum NT, Goodwin AP. Colloids, nanoparticles, and materials for imaging, delivery, ablation, and theranostics by focused ultrasound (FUS). Theranostics 2019; 9:2572-2594. [PMID: 31131054 PMCID: PMC6525987 DOI: 10.7150/thno.32424] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 12/15/2022] Open
Abstract
This review focuses on different materials and contrast agents that sensitize imaging and therapy with Focused Ultrasound (FUS). At high intensities, FUS is capable of selectively ablating tissue with focus on the millimeter scale, presenting an alternative to surgical intervention or management of malignant growth. At low intensities, FUS can be also used for other medical applications such as local delivery of drugs and blood brain barrier opening (BBBO). Contrast agents offer an opportunity to increase selective acoustic absorption or facilitate destructive cavitation processes by converting incident acoustic energy into thermal and mechanical energy. First, we review the history of FUS and its effects on living tissue. Next, we present different colloidal or nanoparticulate approaches to sensitizing FUS, for example using microbubbles, phase-shift emulsions, hollow-shelled nanoparticles, or hydrophobic silica surfaces. Exploring the science behind these interactions, we also discuss ways to make stimulus-responsive, or "turn-on" contrast agents for improved selectivity. Finally, we discuss acoustically-active hydrogels and membranes. This review will be of interest to those working in materials who wish to explore new applications in acoustics and those in acoustics who are seeking new agents to improve the efficacy of their approaches.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
- Present address: CEDAR, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239 USA
| | - Nicholas T. Blum
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
| |
Collapse
|
47
|
Horsley H, Owen J, Browning R, Carugo D, Malone-Lee J, Stride E, Rohn JL. Ultrasound-activated microbubbles as a novel intracellular drug delivery system for urinary tract infection. J Control Release 2019; 301:166-175. [PMID: 30904501 DOI: 10.1016/j.jconrel.2019.03.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/27/2022]
Abstract
The development of new modalities for high-efficiency intracellular drug delivery is a priority for a number of disease areas. One such area is urinary tract infection (UTI), which is one of the most common infectious diseases globally and which imposes an immense economic and healthcare burden. Common uropathogenic bacteria have been shown to invade the urothelial wall during acute UTI, forming latent intracellular reservoirs that can evade antimicrobials and the immune response. This behaviour likely facilitates the high recurrence rates after oral antibiotic treatments, which are not able to penetrate the bladder wall and accumulate to an effective concentration. Meanwhile, oral antibiotics may also exacerbate antimicrobial resistance and cause systemic side effects. Using a human urothelial organoid model, we tested the ability of novel ultrasound-activated lipid microbubbles to deliver drugs into the cytoplasm of apical cells. The gas-filled lipid microbubbles were decorated with liposomes containing the non-cell-permeant antibiotic gentamicin and a fluorescent marker. The microbubble suspension was added to buffer at the apical surface of the bladder model before being exposed to ultrasound (1.1 MHz, 2.5 Mpa, 5500 cycles at 20 ms pulse duration) for 20 s. Our results show that ultrasound-activated intracellular delivery using microbubbles was over 16 times greater than the control group and twice that achieved by liposomes that were not associated with microbubbles. Moreover, no cell damage was detected. Together, our data show that ultrasound-activated microbubbles can safely deliver high concentrations of drugs into urothelial cells, and have the potential to be a more efficacious alternative to traditional oral antibiotic regimes for UTI. This modality of intracellular drug delivery may prove useful in other clinical indications, such as cancer and gene therapy, where such penetration would aid in treatment.
Collapse
Affiliation(s)
- H Horsley
- Department of Renal Medicine, Division of Medicine, University College, London, UK
| | - J Owen
- Institute of Biomedical Engineering, University of Oxford, UK
| | - R Browning
- Institute of Biomedical Engineering, University of Oxford, UK
| | - D Carugo
- Faculty of Physical Sciences and Engineering, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | - J Malone-Lee
- Department of Renal Medicine, Division of Medicine, University College, London, UK
| | - E Stride
- Institute of Biomedical Engineering, University of Oxford, UK
| | - J L Rohn
- Department of Renal Medicine, Division of Medicine, University College, London, UK.
| |
Collapse
|
48
|
Manzano M, Vallet-Regí M. Ultrasound responsive mesoporous silica nanoparticles for biomedical applications. Chem Commun (Camb) 2019; 55:2731-2740. [PMID: 30694270 PMCID: PMC6667338 DOI: 10.1039/c8cc09389j] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanotechnology, which has already revolutionised many technological areas, is expected to transform life sciences. In this sense, nanomedicine could address some of the most important limitations of conventional medicine. In general, nanomedicine includes three major objectives: (1) trap and protect a great amount of therapeutic agents; (2) carry them to the specific site of disease avoiding any leakage; and (3) release on-demand high local concentrations of therapeutic agents. This feature article will make special emphasis on mesoporous silica nanoparticles that release their therapeutic cargo in response to ultrasound.
Collapse
Affiliation(s)
- Miguel Manzano
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain.
| | | |
Collapse
|
49
|
Patnaik SS, Simionescu DT, Goergen CJ, Hoyt K, Sirsi S, Finol EA. Pentagalloyl Glucose and Its Functional Role in Vascular Health: Biomechanics and Drug-Delivery Characteristics. Ann Biomed Eng 2019; 47:39-59. [PMID: 30298373 PMCID: PMC6318003 DOI: 10.1007/s10439-018-02145-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023]
Abstract
Pentagalloyl glucose (PGG) is an elastin-stabilizing polyphenolic compound that has significant biomedical benefits, such as being a free radical sink, an anti-inflammatory agent, anti-diabetic agent, enzymatic resistant properties, etc. This review article focuses on the important benefits of PGG on vascular health, including its role in tissue mechanics, the different modes of pharmacological administration (e.g., oral, intravenous and endovascular route, intraperitoneal route, subcutaneous route, and nanoparticle based delivery and microbubble-based delivery), and its potential therapeutic role in vascular diseases such as abdominal aortic aneurysms (AAA). In particular, the use of PGG for AAA suppression and prevention has been demonstrated to be effective only in the calcium chloride rat AAA model. Therefore, in this critical review we address the challenges that lie ahead for the clinical translation of PGG as an AAA growth suppressor.
Collapse
Affiliation(s)
- Sourav S Patnaik
- Vascular Biomechanics and Biofluids Laboratory, Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249-0670, USA
| | - Dan T Simionescu
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shashank Sirsi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ender A Finol
- Vascular Biomechanics and Biofluids Laboratory, Department of Mechanical Engineering, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249-0670, USA.
| |
Collapse
|
50
|
Upadhyay A, Yagnik B, Desai P, Dalvi SV. Microbubble-Mediated Enhanced Delivery of Curcumin to Cervical Cancer Cells. ACS OMEGA 2018; 3:12824-12831. [PMID: 30411020 PMCID: PMC6217580 DOI: 10.1021/acsomega.8b01737] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 09/24/2018] [Indexed: 05/19/2023]
Abstract
The major bottleneck in the current chemotherapy treatment of cancer is the low bioavailability and high cytotoxicity. Targeted delivery of drug to the cancer cells can reduce the cytotoxicity and increase the bioavailability. In this context, microbubbles are currently being explored as drug-delivery vehicles to effectively deliver drug to the tumors or cancerous cells. Microbubbles when used along with ultrasound can enhance drug uptake and inhibit the growth of tumor cells. Several potential anticancer molecules exhibit poor water solubility, which limits their use in therapeutic applications. Such poorly water soluble molecules can be coadministered with microbubbles or encapsulated within or loaded on the microbubbles surface, to enhance the effectiveness of these molecules against cancer cells. Curcumin is one of such potential anticancer molecules obtained from the rhizome of herbal spice, turmeric. In this work, curcumin-loaded protein microbubbles were synthesized and examined for effective in vitro delivery of curcumin to HeLa cells. Microbubbles in the size range of 1-10 μm were produced using perfluorobutane as core gas and bovine serum albumin (BSA) as shell material and were loaded with curcumin. The amount of curcumin loaded on the microbubble surface was estimated using UV-vis spectroscopy, and the average curcumin loading was found to be ∼54 μM/108 microbubbles. Kinetics of in vitro curcumin release from microbubble surface was also estimated, where a 4-fold increase in the rate of curcumin release was obtained in the presence of ultrasound. Sonication and incubation of HeLa cells with curcumin-loaded BSA microbubbles enhanced the uptake of curcumin by ∼250 times. Further, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay confirmed ∼71% decrease in cell viability when HeLa cells were sonicated with curcumin-loaded microbubbles and incubated for 48 h.
Collapse
Affiliation(s)
- Awaneesh Upadhyay
- Chemical
Engineering, IIT Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Bhrugu Yagnik
- B.V.
Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, Ahmedabad 380054, Gujarat, India
| | - Priti Desai
- B.V.
Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej, Ahmedabad 380054, Gujarat, India
| | - Sameer V. Dalvi
- Chemical
Engineering, IIT Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
- E-mail:
| |
Collapse
|