1
|
Elkhashab M, Barreto G, Fauconnier M, Le Bourlout Y, Creemers LB, Nieminen HJ, Howard KA. ADAMTS5-specific gapmer release from an albumin biomolecular assembly and cartilage internalization triggered by ultrasound. Drug Deliv 2025; 32:2464921. [PMID: 39967271 PMCID: PMC11841101 DOI: 10.1080/10717544.2025.2464921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE Antisense oligonucleotides (ASOs) have reached the clinic; however, they lack tissue specificity. Albumin is a plasma-abundant macromolecule that has been shown to accumulate in inflamed tissues. In this work, we have designed a recombinant human albumin (rHA)-based biomolecular assembly incorporating a DNase-resistant phosphorothioate-based complementary oligonucleotide (cODN) and an anti-ADAMTS5 ASO for potential delivery to inflamed sites. Ultrasound (US) was used to trigger ASO release from the assembly and enhance internalization into articular cartilage. METHODS A phosphorothioate cODN was conjugated to rHA through a maleimide cross-linker after which, a therapeutic ADAMTS5-specific gapmer ASO was annealed to the cODN. ASO release was assessed after exposing the biomolecular assembly to different US conditions using an US-actuated medical needle operating at 32.2 kHz. Gene silencing efficiency of US-treated anti-ADAMTS5 ASO was assessed in human primary chondrocytes isolated from osteoarthritic patients. US-mediated ASO penetration into articular cartilage was assessed on ex vivo bovine articular cartilage. RESULTS ASO release was observed after exposure to US waves in continuous mode conditions that did not compromise ASO gene silencing efficiency in human chondrocytes. Furthermore, US increased ASO internalization into bovine articular cartilage after 30 min of application without detrimental effects on chondrocyte viability. CONCLUSION A medical needle driven by continuous US waves at 32.2 kHz has the capability of disassembling a duplex oligonucleotide and enhancing released ASOs internalization into articular cartilage. This work offers the potential delivery and the local triggered release of ASOs at the surface of articular cartilage providing potential benefits for the treatment of diverse cartilage pathologies.
Collapse
Affiliation(s)
- Marwa Elkhashab
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
| | - Goncalo Barreto
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Orton Orthopedic Hospital, Helsinki, Finland
| | - Maxime Fauconnier
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Yohann Le Bourlout
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Laura B. Creemers
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Heikki J. Nieminen
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
2
|
Ghosh R, Kumar M, Kumar S, Komal K, Sharma R, Kurmi BD. Small molecule therapeutics for receptor-mediated targeting through liposomes in breast cancer treatment: A comprehensive review. Bioorg Chem 2025; 160:108442. [PMID: 40199009 DOI: 10.1016/j.bioorg.2025.108442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer (BC) remains a significant global health challenge, with conventional treatment approaches such as surgery, chemotherapy, and radiation therapy. These approaches face limitations in targeting, toxicity, and efficacy. Liposomal drug delivery systems have emerged as promising tools for targeted breast cancer therapies. Liposomes can encapsulate both hydrophilic and hydrophobic drugs, improve drug distribution, and reduce the side effects. Passive targeting exploits the enhanced permeability and retention effect in tumor tissues, whereas active targeting employs small molecule ligands such as aptamers, folic acid (FA), transferrin, and monoclonal antibodies to specifically bind to overexpressed receptors on cancer cells. Aptamer-functionalized liposomes exhibit high specificity and affinity, folate and transferrin receptor targeting enhances cellular uptake and cytotoxicity, and antibody-conjugated liposomes improve drug delivery and efficacy by targeting specific antigens. Dual-responsive liposomes are sensitive to multiple stimuli and further enhance targeting precision. However, challenges remain, including tumor heterogeneity, limited penetration, and potential immunogenicity. Current research has focused on developing stable and effective formulations and exploring combination-targeting strategies to overcome these limitations. With further advancements, targeted liposomal drug delivery systems hold great promise in improving breast cancer treatment outcomes and reducing adverse effects.
Collapse
Affiliation(s)
- Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India.
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Rohit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| |
Collapse
|
3
|
Einen C, Snipstad S, Wesche HF, Nordlund V, Devold EJ, Amini N, Hansen R, Sulheim E, Davies CDL. Impact of the tumor microenvironment on delivery of nanomedicine in tumors treated with ultrasound and microbubbles. J Control Release 2025; 378:656-670. [PMID: 39701458 DOI: 10.1016/j.jconrel.2024.12.037] [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: 09/04/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
The delivery of nanoparticles to tumors has been shown preclinically to be improved by microbubble-mediated ultrasound. However, the mechanisms and biological effects are not fully understood. In this study, we explored the influence of the tumor microenvironment on nanoparticle uptake and microdistribution both with and without ultrasound and microbubble treatment. Three murine tumor models, KPC (pancreatic ductal adenocarcinoma), 4T1 (triple negative mammary carcinoma) and CT26 (colon carcinoma), were characterized with respect to extracellular matrix composition, tumor stiffness and perfusion. KPC and 4T1 tumors presented higher levels of collagen and hyaluronic acid and were stiffer compared to CT26, whereas all three tumors had similar levels of sulfated glycosaminoglycans. Furthermore, the 4T1 tumors appeared poorly vascularized with a lower cell density compared to KPC and CT26. All three tumors presented similar nanoparticle uptake, but extravasated nanoparticles traveled significantly shorter in KPC tumors compared to 4T1 and CT26. The effect of ultrasound and microbubble treatment on the tumor uptake and penetration of polymer nanoparticles into the extracellular matrix were evaluated using a treatment protocol previously shown to increase nanoparticle delivery to tumors. Interestingly, we found a significant increase in nanoparticle uptake in the soft CT26 tumor, but no effect of the ultrasound treatment in the stiff KPC and 4T1 tumors, suggesting that tumor stiffness is an important parameter for treatment with ultrasound and microbubbles. Ultrasound treatment resulted in a modest but not statistically significant improvement in nanoparticle penetration through the extracellular matrix. In tumors demonstrating increased uptake of nanoparticles following ultrasound treatment, the uptake correlated positively with blood volume. These findings emphasize the importance of taking the tumor microenvironment into consideration when optimizing ultrasound parameters for delivery of nanomedicine.
Collapse
Affiliation(s)
- Caroline Einen
- Porelab and Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Håkon F Wesche
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Veronica Nordlund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ella J Devold
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | | |
Collapse
|
4
|
Yazdan M, Naghib SM. Smart Ultrasound-responsive Polymers for Drug Delivery: An Overview on Advanced Stimuli-sensitive Materials and Techniques. Curr Drug Deliv 2025; 22:283-309. [PMID: 38288800 DOI: 10.2174/0115672018283792240115053302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 04/11/2025]
Abstract
In recent years, a notable advancement has occurred in the domain of drug delivery systems via the integration of intelligent polymers that respond to ultrasound. The implementation of this groundbreaking methodology has significantly revolutionised the controlled and precise delivery of therapeutic interventions. An in-depth investigation is conducted into the most recent developments in ultrasonic stimulus-responsive materials and techniques for the purpose of accomplishing precise medication administration. The investigation begins with an exhaustive synopsis of the foundational principles underlying drug delivery systems that react to ultrasonic stimuli, focusing specifically on the complex interplay between polymers and ultrasound waves. Significant attention is devoted to the development of polymers that demonstrate tailored responsiveness to ultrasound, thereby exemplifying their versatility in generating controlled drug release patterns. Numerous classifications of intelligent polymers are examined in the discussion, including those that react to variations in temperature, pH, and enzymes. When coupled with ultrasonic stimuli, these polymers offer a sophisticated framework for the precise manipulation of drug release in terms of both temporal and spatial dimensions. The present study aims to examine the synergistic effects of responsive polymers and ultrasound in overcoming biological barriers such as the blood-brain barrier and the gastrointestinal tract. By doing so, it seeks to shed light on the potential applications of these materials in intricate clinical scenarios. The issues and future prospects of intelligent ultrasound-responsive polymers in the context of drug delivery are critically analysed in this article. The objective of this study is to offer valuable perspectives on the challenges that must be overcome to enable the effective implementation of these technologies. The primary objective of this comprehensive review is to furnish researchers, clinicians, and pharmaceutical scientists with a wealth of information that will serve as a guide for forthcoming developments in the development and enhancement of intelligent drug delivery systems that employ ultrasound-responsive polymers to attain superior therapeutic outcomes.
Collapse
Affiliation(s)
- Mostafa Yazdan
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| |
Collapse
|
5
|
Zhu S, Jin G, He X, Li Y, Xu F, Guo H. Mechano-assisted strategies to improve cancer chemotherapy. Life Sci 2024; 359:123178. [PMID: 39471901 DOI: 10.1016/j.lfs.2024.123178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
Chemotherapy remains a cornerstone in cancer treatment; however, its effectiveness is frequently undermined by the development of drug resistance. Recent studies underscores the pivotal role of the tumor mechanical microenvironment (TMME) and the emerging field of mechanical nanomedicine in tackling chemo-resistance. This review offers an in-depth analysis of mechano-assisted strategies aimed at mitigating chemo-resistance through the modification of the TMME and the refinement of mechanical nanomedicine delivery systems. We explore the potential of targeting abnormal tumor mechanical properties as a promising avenue for enhancing the efficacy of cancer chemotherapy, which offers novel directions for advancing future cancer therapies, especially from the mechanomedicine perspective.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, PR China.
| |
Collapse
|
6
|
Maciulevičius M, Palepšienė R, Vykertas S, Raišutis R, Rafanavičius A, Krilavičius T, Šatkauskas S. The comparison of the dynamics of Ca 2+ and bleomycin intracellular delivery after cell sonoporation and electroporation in vitro. Bioelectrochemistry 2024; 158:108708. [PMID: 38636366 DOI: 10.1016/j.bioelechem.2024.108708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
Ca2+, in combination with SP or EP, induces cell cytotoxicity much faster compared to BLM. The application of BLM in combination with, SP or EP, reaches the level of cell death, induced by similar combination with Ca2+, only after 72 h. The methods of SP and EP were calibrated according to the level of differential cytotoxicity, determined after 6 days (using cell clonogenic assay). The combination of Ca2+ SP induces cell death faster than Ca2+ EP - after Ca2+ SP it increases to a maximum level after 15 min and remains constant for up to 6 days, while the cytotoxic efficiency after Ca2+ EP increases to the level of Ca2+ SP only after 72 h. The combination of BLM SP shows a very similar dynamics to BLM EP - both reach maximal level of cytotoxicity after 48-72 h. Ca2+ and BLM in combination with SP have shown similar levels of cytotoxicity at higher acoustic pressures (≥250 kPa); therefore, Ca2+ SP can be used to induce immediate and maximal level of cytotoxic effect. The faster cytotoxic efficiency of Ca2+ in combination with SP than EP was determined to be due to the involvement of microbubble inertial cavitation.
Collapse
Affiliation(s)
- Martynas Maciulevičius
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania; Ultrasound Research Institute, Kaunas University of Technology, K. Baršausko st. 59, LT-51423 Kaunas, Lithuania.
| | - Rūta Palepšienė
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Salvijus Vykertas
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Renaldas Raišutis
- Ultrasound Research Institute, Kaunas University of Technology, K. Baršausko st. 59, LT-51423 Kaunas, Lithuania; Department of Electrical Power Systems, Faculty of Electrical and Electronics Engineering, Kaunas University of Technology, Studentų st. 48, LT-51367 Kaunas, Lithuania.
| | - Aras Rafanavičius
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Tomas Krilavičius
- Faculty of Informatics, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| | - Saulius Šatkauskas
- Biophysical Research Group, Faculty of Natural Sciences, Vytautas Magnus University, Vileikos st. 8, LT-44404, Kaunas, Lithuania.
| |
Collapse
|
7
|
Zhu P, Simon I, Kokalari I, Kohane DS, Rwei AY. Miniaturized therapeutic systems for ultrasound-modulated drug delivery to the central and peripheral nervous system. Adv Drug Deliv Rev 2024; 208:115275. [PMID: 38442747 PMCID: PMC11031353 DOI: 10.1016/j.addr.2024.115275] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ultrasound is a promising technology to address challenges in drug delivery, including limited drug penetration across physiological barriers and ineffective targeting. Here we provide an overview of the significant advances made in recent years in overcoming technical and pharmacological barriers using ultrasound-assisted drug delivery to the central and peripheral nervous system. We commence by exploring the fundamental principles of ultrasound physics and its interaction with tissue. The mechanisms of ultrasonic-enhanced drug delivery are examined, as well as the relevant tissue barriers. We highlight drug transport through such tissue barriers utilizing insonation alone, in combination with ultrasound contrast agents (e.g., microbubbles), and through innovative particulate drug delivery systems. Furthermore, we review advances in systems and devices for providing therapeutic ultrasound, as their practicality and accessibility are crucial for clinical application.
Collapse
Affiliation(s)
- Pancheng Zhu
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands; State Key Laboratory of Mechanics and Control of Aerospace Structures, Nanjing University of Aeronautics & Astronautics, 210016, Nanjing, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ignasi Simon
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Ida Kokalari
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Alina Y Rwei
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands.
| |
Collapse
|
8
|
Shakya G, Cattaneo M, Guerriero G, Prasanna A, Fiorini S, Supponen O. Ultrasound-responsive microbubbles and nanodroplets: A pathway to targeted drug delivery. Adv Drug Deliv Rev 2024; 206:115178. [PMID: 38199257 DOI: 10.1016/j.addr.2023.115178] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/21/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Ultrasound-responsive agents have shown great potential as targeted drug delivery agents, effectively augmenting cell permeability and facilitating drug absorption. This review focuses on two specific agents, microbubbles and nanodroplets, and provides a sequential overview of their drug delivery process. Particular emphasis is given to the mechanical response of the agents under ultrasound, and the subsequent physical and biological effects on the cells. Finally, the state-of-the-art in their pre-clinical and clinical implementation are discussed. Throughout the review, major challenges that need to be overcome in order to accelerate their clinical translation are highlighted.
Collapse
Affiliation(s)
- Gazendra Shakya
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Marco Cattaneo
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Giulia Guerriero
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Anunay Prasanna
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Samuele Fiorini
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland
| | - Outi Supponen
- Institute of Fluid Dynamics, D-MAVT, Sonneggstrasse 3, ETH Zurich, Zurich, 8092, Switzerland.
| |
Collapse
|
9
|
Bouakaz A, Michel Escoffre J. From concept to early clinical trials: 30 years of microbubble-based ultrasound-mediated drug delivery research. Adv Drug Deliv Rev 2024; 206:115199. [PMID: 38325561 DOI: 10.1016/j.addr.2024.115199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Ultrasound mediated drug delivery, a promising therapeutic modality, has evolved remarkably over the past three decades. Initially designed to enhance contrast in ultrasound imaging, microbubbles have emerged as a main vector for drug delivery, offering targeted therapy with minimized side effects. This review addresses the historical progression of this technology, emphasizing the pivotal role microbubbles play in augmenting drug extravasation and targeted delivery. We explore the complex mechanisms behind this technology, from stable and inertial cavitation to diverse acoustic phenomena, and their applications in medical fields. While the potential of ultrasound mediated drug delivery is undeniable, there are still challenges to overcome. Balancing therapeutic efficacy and safety and establishing standardized procedures are essential areas requiring attention. A multidisciplinary approach, gathering collaborations between researchers, engineers, and clinicians, is important for exploiting the full potential of this technology. In summary, this review highlights the potential of using ultrasound mediated drug delivery in improving patient care across various medical conditions.
Collapse
Affiliation(s)
- Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | | |
Collapse
|
10
|
Chang M, Zhang L, Wang Z, Chen L, Dong Y, Yang J, Chen Y. Nanomedicine/materdicine-enabled sonocatalytic therapy. Adv Drug Deliv Rev 2024; 205:115160. [PMID: 38110153 DOI: 10.1016/j.addr.2023.115160] [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: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/20/2023]
Abstract
The advent of numerous treatment modalities with desirable therapeutic efficacy has been made possible by the fast development of nanomedicine and materdicine, among which the ultrasound (US)-triggered sonocatalytic process as minimal or non-invasive method has been frequently employed for diagnostic and therapeutic purposes. In comparison to phototherapeutic approaches with inherent penetration depth limitations, sonocatalytic therapy shatters the depth limit of photoactivation and offers numerous remarkable prospects and advantages, including mitigated side effects and appropriate tissue-penetration depth. Nevertheless, the optimization of sonosensitizers and therapies remains a significant issue in terms of precision, intelligence and efficiency. In light of the fact that nanomedicine and materdicine can effectively enhance the theranostic efficiency, we herein aim to furnish a cutting-edge review on the latest progress and development of nanomedicine/materdicine-enabled sonocatalytic therapy. The design methodologies and biological features of nanomedicine/materdicine-based sonosensitizers are initially introduced to reveal the underlying relationship between composition/structure, sonocatalytic function and biological effect, in accompany with a thorough discussion of nanomedicine/materdicine-enabled synergistic therapy. Ultimately, the facing challenges and future perspectives of this intriguing sonocatalytic therapy are highlighted and outlined to promote technological advancements and clinical translation in efficient disease treatment.
Collapse
Affiliation(s)
- Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, PR China
| | - Lu Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Hebei University, Baoding 071000, PR China
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China
| | - Yang Dong
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| | - Jishun Yang
- Naval Medical Center of PLA, Medical Security Center, Shanghai 200052, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, PR China.
| |
Collapse
|
11
|
Kim G, Won J, Kim CW, Park JR, Park D. Fabrication and Evaluation of Ultrasound-Responsive Emulsion Loading Paclitaxel for Targeted Chemotherapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:91-99. [PMID: 38146661 DOI: 10.1021/acs.langmuir.3c02005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Chemotherapy is the most widely used cancer treatment, but it has several drawbacks such as adverse side effects and low bioavailability. To address these limitations, various drug delivery systems have been investigated, including liposomes, micelles, and emulsions. These drug delivery technologies have been improving the efficacy and safety of conventional chemotherapy. This study presents an emerging drug delivery technology for targeted chemotherapy using drug-loaded ultrasound-responsive emulsion (URE) as a drug carrier and ultrasound technology for external activation. URE was designed to be responsive to ultrasound energy and fabricated by using an emulsification technique. To investigate this technology, paclitaxel, as a model drug, was used and encapsulated into URE. The size distribution, morphology, and drug release behavior of paclitaxel-loaded URE (PTX-URE) were characterized, and the echogenicity of PTX-URE was assessed by using ultrasound imaging equipment. The cellular uptake and cytotoxicity of PTX-URE with ultrasound were evaluated in breast cancer cells (MDA-MB-231). Our in vitro results indicate that the combination of PTX-URE and ultrasound significantly enhanced cellular uptake by 10.6-fold and improved cytotoxicity by 24.1% compared to PTX alone. These findings suggest that the URE platform combined with ultrasound is a promising technology to improve the drug delivery efficiency for chemotherapy.
Collapse
Affiliation(s)
- Gayoung Kim
- Bioinfra Life Science Inc., Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul 03080, South Korea
| | - Jongho Won
- Bioinfra Life Science Inc., Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul 03080, South Korea
| | - Chul-Woo Kim
- Bioinfra Life Science Inc., Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul 03080, South Korea
| | - Jong-Ryul Park
- Bioinfra Life Science Inc., Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul 03080, South Korea
| | - Donghee Park
- Bioinfra Life Science Inc., Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul 03080, South Korea
| |
Collapse
|
12
|
Lin CW, Fan CH, Yeh CK. The relationship between surface drug distribution of Dox-loaded microbubbles and drug release/cavitation behaviors with ultrasound. ULTRASONICS SONOCHEMISTRY 2024; 102:106728. [PMID: 38103369 PMCID: PMC10765110 DOI: 10.1016/j.ultsonch.2023.106728] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/23/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Ultrasound (US)-triggered microbubbles (MBs) drug delivery is a promising tool for noninvasive and localized therapy. Several studies have shown the potential of drug-loaded MBs to boost the delivery of therapeutic substances to target tissue effectively. Nevertheless, little is known about the surface payload distribution affecting the cavitation activity and drug release behavior of the drug-loaded MBs. In this study, we designed a common chemodrug (Doxorubicin, Dox)-loaded MB (Dox-MBs) and regulated the payload distribution as uniform or cluster onto the outer surface of MBs. The Dox distribution on the MB shells was assessed by confocal fluorescence microscopic imaging. The acoustic properties of the Dox-MBs with different Dox distributions were evaluated by their acoustic stability and cavitation activities. The payload release and the fragments from Dox-MBs in response to different US parameters were measured and visualized by column chromatography and cryo-electron microscopy, respectively. By amalgamating these methodologies, we found that stable cavitation was sufficient for triggering uniform-loaded MBs to release their payload, but stable cavitation and inertial cavitation were required for cluster-loaded MBs. The released substances included free Dox and Dox-containing micelle/liposome; their portions depended on the payload distribution, acoustic pressure, cycle number, and sonication duration. Furthermore, we also revealed that the Dox-containing micelle/liposome in cluster-loaded MBs had the potential for multiple drug releases upon US sonication. This study compared uniform-loaded MBs and cluster-loaded MBs to enhance our comprehension of drug-loaded MBs mediated drug delivery.
Collapse
Affiliation(s)
- Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
13
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
14
|
Micaletti F, Escoffre JM, Kerneis S, Bouakaz A, Galvin JJ, Boullaud L, Bakhos D. Microbubble-assisted ultrasound for inner ear drug delivery. Adv Drug Deliv Rev 2024; 204:115145. [PMID: 38042259 DOI: 10.1016/j.addr.2023.115145] [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: 09/21/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Treating pathologies of the inner ear is a major challenge. To date, a wide range of procedures exists for administering therapeutic agents to the inner ear, with varying degrees of success. The key is to deliver therapeutics in a way that is minimally invasive, effective, long-lasting, and without adverse effects on vestibular and cochlear function. Microbubble-assisted ultrasound ("sonoporation") is a promising new modality that can be adapted to the inner ear. Combining ultrasound technology with microbubbles in the middle ear can increase the permeability of the round window, enabling therapeutic agents to be delivered safely and effectively to the inner ear in a targeted manner. As such, sonoporation is a promising new approach to treat hearing loss and vertigo. This review summarizes all studies on the delivery of therapeutic molecules to the inner ear using sonoporation.
Collapse
Affiliation(s)
- Fabrice Micaletti
- ENT and Cervico-Facial Surgery Department, University Hospital Center of Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
| | | | - Sandrine Kerneis
- ENT and Cervico-Facial Surgery Department, University Hospital Center of Tours, 2 Boulevard Tonnellé, 37044 Tours, France
| | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - John J Galvin
- Faculty of medicine, Université de Tours, 10 boulevard Tonnellé, 37044 Tours, France; House Institute Foundation, 2100 W 3rd Street, Suite 111, Los Angeles, CA 90057, USA
| | - Luc Boullaud
- ENT and Cervico-Facial Surgery Department, University Hospital Center of Tours, 2 Boulevard Tonnellé, 37044 Tours, France
| | - David Bakhos
- ENT and Cervico-Facial Surgery Department, University Hospital Center of Tours, 2 Boulevard Tonnellé, 37044 Tours, France; UMR 1253, iBrain, Université de Tours, Inserm, Tours, France; Faculty of medicine, Université de Tours, 10 boulevard Tonnellé, 37044 Tours, France; House Institute Foundation, 2100 W 3rd Street, Suite 111, Los Angeles, CA 90057, USA
| |
Collapse
|
15
|
Zhang W, Wang D, Ostertag-Hill CA, Han Y, Li X, Zheng Y, Lu B, Kohane DS. On-Demand Opioid Effect Reversal with an Injectable Light-Triggered Polymer-Naloxone Conjugate. NANO LETTERS 2023; 23:10545-10553. [PMID: 37937844 PMCID: PMC10949359 DOI: 10.1021/acs.nanolett.3c03426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Misuse of opioids can lead to a potential lethal overdose. Timely administration of naloxone is critical for survival. Here, we designed a polymer-naloxone conjugate that can provide on-demand phototriggered opioid reversal. Naloxone was attached to the polymer poly(lactic-co-glycolic acid) via a photocleavable coumarin linkage and formulated as injectable nanoparticles. In the absence of irradiation, the formulation did not release naloxone. Upon irradiation with blue (400 nm) light, the nanoparticles released free naloxone, reversing the effect of morphine in mice. Such triggered events could be performed days and weeks after the initial administration of the nanoparticles and could be performed repeatedly.
Collapse
Affiliation(s)
- Wei Zhang
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Dali Wang
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Claire A. Ostertag-Hill
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yiyuan Han
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Xiyu Li
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yueqin Zheng
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Berwyn Lu
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, The Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
16
|
Hoseini B, Jaafari MR, Golabpour A, Momtazi-Borojeni AA, Karimi M, Eslami S. Application of ensemble machine learning approach to assess the factors affecting size and polydispersity index of liposomal nanoparticles. Sci Rep 2023; 13:18012. [PMID: 37865639 PMCID: PMC10590434 DOI: 10.1038/s41598-023-43689-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 10/23/2023] Open
Abstract
Liposome nanoparticles have emerged as promising drug delivery systems due to their unique properties. Assessing particle size and polydispersity index (PDI) is critical for evaluating the quality of these liposomal nanoparticles. However, optimizing these parameters in a laboratory setting is both costly and time-consuming. This study aimed to apply a machine learning technique to assess the impact of specific factors, including sonication time, extrusion temperature, and compositions, on the size and PDI of liposomal nanoparticles. Liposomal solutions were prepared and subjected to sonication with varying values for these parameters. Two compositions: (A) HSPC:DPPG:Chol:DSPE-mPEG2000 at 55:5:35:5 molar ratio and (B) HSPC:Chol:DSPE-mPEG2000 at 55:40:5 molar ratio, were made using remote loading method. Ensemble learning (EL), a machine learning technique, was employed using the Least-squares boosting (LSBoost) algorithm to accurately model the data. The dataset was randomly split into training and testing sets, with 70% allocated for training. The LSBoost algorithm achieved mean absolute errors of 1.652 and 0.0105 for modeling the size and PDI, respectively. Under conditions where the temperature was set at approximately 60 °C, our EL model predicted a minimum particle size of 116.53 nm for composition (A) with a sonication time of approximately 30 min. Similarly, for composition (B), the model predicted a minimum particle size of 129.97 nm with sonication times of approximately 30 or 55 min. In most instances, a PDI of less than 0.2 was achieved. These results highlight the significant impact of optimizing independent factors on the characteristics of liposomal nanoparticles and demonstrate the potential of EL as a decision support system for identifying the best liposomal formulation. We recommend further studies to explore the effects of other independent factors, such as lipid composition and surfactants, on liposomal nanoparticle characteristics.
Collapse
Affiliation(s)
- Benyamin Hoseini
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Golabpour
- Department of Health Information Technology, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, School of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Maryam Karimi
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, USA
| | - Saeid Eslami
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Informatics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Zheng Y, Li C, Zhang C, He J, Jiang X, Ta D. Distinct thermal effect on biological tissues using subwavelength ultrasound metalens at megahertz. iScience 2023; 26:107929. [PMID: 37810209 PMCID: PMC10551838 DOI: 10.1016/j.isci.2023.107929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/21/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Ultrasound focusing plays an important role in biomedical therapy and diagnosis. Acoustic metalens has showcased remarkable focusing performance but yet to be implemented to the practical ultrasound therapeutic applications. We design a planar metalens operating at megahertz and experimentally demonstrate the distinct thermal effect on biological tissues induced by the high-resolution focusing. A prominent temperature rise of 50°C is experimentally observed in the biological phantom, with a much lower input ultrasound power of 4 W compared with the traditional methods. We further study the thermal effect on fresh porcine liver and investigate the morphological changes under different physical parameters. Visible lesions are observed in in vitro tissues at the lowest input ultrasound power of 2.6 W within 10 s. This study facilitates the practical biomedical application of acoustic metalens, providing a feasible approach for the precise, safe, and reliable therapeutic ultrasound with the simple and compact metalens.
Collapse
Affiliation(s)
- Yan Zheng
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Chen Li
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Chuanxin Zhang
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Jiajie He
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Xue Jiang
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200433, China
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
18
|
Dutt Y, Pandey RP, Dutt M, Gupta A, Vibhuti A, Raj VS, Chang CM, Priyadarshini A. Liposomes and phytosomes: Nanocarrier systems and their applications for the delivery of phytoconstituents. Coord Chem Rev 2023; 491:215251. [DOI: 10.1016/j.ccr.2023.215251] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
19
|
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
|
20
|
Miranda TC, Andrade JFM, Gelfuso GM, Cunha-Filho M, Oliveira LA, Gratieri T. Novel technologies to improve the treatment of endodontic microbial infections: Inputs from a drug delivery perspective. Int J Pharm 2023; 635:122794. [PMID: 36870400 DOI: 10.1016/j.ijpharm.2023.122794] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Endodontic microbial infections are still a challenge for an effective treatment for being biofilm-mediated and very refractory to conventional therapies. Biomechanical preparation and chemical irrigants cannot fully eradicate biofilms due to the anatomic structure of the root canal system. Instruments employed in biomechanical preparation and irrigants solution cannot reach the narrow and deepest portion of root canals, especially the apical thirds. In addition, aside from the dentin surface, biofilms can also infiltrate dentine tubules and periapical tissues, compromising treatment success. Therefore, different technologies have been investigated to achieve a more effective outcome in the control of endodontic infections. However, these technologies continue to face great difficulties in reaching the apical region and eradicating biofilms to avoid the recurrence of infection. Here, we present an overview of the fundamentals of endodontics infections and review technologies currently available for root canal treatment. We discuss them from a drug delivery perspective, highlighting each technology's strength to envision the best use of these technologies.
Collapse
Affiliation(s)
- Thamires C Miranda
- Laboratory of Food, Drugs, and Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Jayanaraian F M Andrade
- Laboratory of Food, Drugs, and Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Guilherme M Gelfuso
- Laboratory of Food, Drugs, and Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Marcilio Cunha-Filho
- Laboratory of Food, Drugs, and Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Laudimar A Oliveira
- Department of Dentistry, Faculty of Health Sciences, University of Brasilia, 70910-900, Brasília, DF, Brazil
| | - Tais Gratieri
- Laboratory of Food, Drugs, and Cosmetics (LTMAC), University of Brasilia, 70910-900, Brasília, DF, Brazil.
| |
Collapse
|
21
|
Abstract
The conventional microbubble-based ultrasound biomedicine clinically plays a vital role in providing the dynamic detection of macro and microvasculature and disease theranostics. However, the intrinsic limitation of particle size severely decreases the treatment effectiveness due to their vascular transport characteristics, which promotes the development and application of multifunctional ultrasound-responsive nanomaterials. Herein, we put forward a research field of "ultrasound nanomedicine and materdicine", referring to the interdiscipline of ultrasound, nanobiotechnology and materials, which seeks to produce specific biological effects for addressing the challenges faced and dilemma of conventional ultrasound medicine. We comprehensively summarize the state-of-the-art scientific advances in the latest progress in constructing ultrasound-based platforms and ultrasound-activated sonosensitizers, ranging from the synthesis strategies, biological functions to ultrasound-triggered therapeutic applications. Ultimately, the unresolved challenges and clinical-translation potentials of ultrasound nanomedicine and materdicine are discussed and prospected in this evolving field.
Collapse
Affiliation(s)
- Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Xue Wang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Meiqi Chang
- Central Laboratory of Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China.
| | - Jia Guo
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| |
Collapse
|
22
|
Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. "Smart" drug delivery: A window to future of translational medicine. Front Chem 2023; 10:1095598. [PMID: 36688039 PMCID: PMC9846181 DOI: 10.3389/fchem.2022.1095598] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 01/05/2023] Open
Abstract
Chemotherapy is the mainstay of cancer treatment today. Chemotherapeutic drugs are non-selective and can harm both cancer and healthy cells, causing a variety of adverse effects such as lack of specificity, cytotoxicity, short half-life, poor solubility, multidrug resistance, and acquiring cancer stem-like characteristics. There is a paradigm shift in drug delivery systems (DDS) with the advent of smarter ways of targeted cancer treatment. Smart Drug Delivery Systems (SDDSs) are stimuli responsive and can be modified in chemical structure in response to light, pH, redox, magnetic fields, and enzyme degradation can be future of translational medicine. Therefore, SDDSs have the potential to be used as a viable cancer treatment alternative to traditional chemotherapy. This review focuses mostly on stimuli responsive drug delivery, inorganic nanocarriers (Carbon nanotubes, gold nanoparticles, Meso-porous silica nanoparticles, quantum dots etc.), organic nanocarriers (Dendrimers, liposomes, micelles), antibody-drug conjugates (ADC) and small molecule drug conjugates (SMDC) based SDDSs for targeted cancer therapy and strategies of targeted drug delivery systems in cancer cells.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Meheli Adhikary
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Praveen Kumar Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Bhudev C. Das
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,*Correspondence: Seema Bhatnagar,
| |
Collapse
|
23
|
Geng S, Guo M, Zhan G, Shi D, Shi L, Gan L, Zhao Y, Yang X. NIR-triggered ligand-presenting nanocarriers for enhancing synergistic photothermal-chemotherapy. J Control Release 2023; 353:229-240. [PMID: 36427657 DOI: 10.1016/j.jconrel.2022.11.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/14/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Surface PEGylation of nanomedicine is effective for prolonging blood circulation time and facilitating the EPR effect, whereas the hydrophilic stealth surface inhibits effective cellular uptake and hinders active targeting. To address the dilemma, herein, a NIR light-triggered dePEGylation/ligand-presenting strategy based on thermal decomposition of azo bonds is developed, whereby Dox/Pz-IR nanoparticle is self-assembled from thermo-labile azo molecule-linked long PEG chain polymer (Pz-IR), cRGD-conjugated IR783 with short PEG chains (rP-IR) and doxorubicin. The long PEG chains could mask cRGD peptides in the blood circulation, preventing serum degradation and nonspecific interaction with normal cells. Once exposed to NIR laser, the PEG corona is stripped off owing to the rupture of azo bonds through the photothermal effect of IR783, and the masked cRGD peptides are exposed, which remarkably enhances cellular uptake by tumor cells and improves tumor accumulation. Dox/Pz-IR achieves the optimal synergy of photothermal-chemotherapy at mild temperature through progressive tumor accumulation, precisely regulated photothermal effect and NIR-PTT induced pulsated drug release. The strategy of NIR photo-driven dePEGylation/targeting offers a new approach to overcoming the "PEG dilemma", and provides a noval avenue for programmed tumor-targeted drug delivery.
Collapse
Affiliation(s)
- Shinan Geng
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Mengqin Guo
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Guiting Zhan
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dingwen Shi
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liyun Shi
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou 310015, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yanbing Zhao
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China; GBA Research Innovation Institute for Nanotechnology, Guangdong 510530, China.
| |
Collapse
|
24
|
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
|
25
|
van Wamel A, Mühlenpfordt M, Hansen R, Healey A, Villanueva FS, Kotopoulis S, Davies CDL, Chen X. Ultrafast Microscopy Imaging of Acoustic Cluster Therapy Bubbles: Activation and Oscillation. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1840-1857. [PMID: 35773079 DOI: 10.1016/j.ultrasmedbio.2022.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/15/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Acoustic Cluster Therapy (ACT®) is a platform for improving drug delivery and has had promising pre-clinical results. A clinical trial is ongoing. ACT® is based on microclusters of microbubbles-microdroplets that, when sonicated, form a large ACT® bubble. The aim of this study was to obtain new knowledge on the dynamic formation and oscillations of ACT® bubbles by ultrafast optical imaging in a microchannel. The high-speed recordings revealed the microbubble-microdroplet fusion, and the gas in the microbubble acted as a vaporization seed for the microdroplet. Subsequently, the bubble grew by gas diffusion from the surrounding medium and became a large ACT® bubble with a diameter of 5-50 μm. A second ultrasound exposure at lower frequency caused the ACT® bubble to oscillate. The recorded oscillations were compared with simulations using the modified Rayleigh-Plesset equation. A term accounting for the physical boundary imposed by the microchannel wall was included. The recorded oscillation amplitudes were approximately 1-2 µm, hence similar to oscillations of smaller contrast agent microbubbles. These findings, together with our previously reported promising pre-clinical therapeutic results, suggest that these oscillations covering a large part of the vessel wall because of the large bubble volume can substantially improve therapeutic outcome.
Collapse
Affiliation(s)
- Annemieke van Wamel
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Flordeliza S Villanueva
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Spiros Kotopoulis
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Xucai Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
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
|
27
|
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]
|
28
|
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]
|
29
|
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
|
30
|
Liu X, Zhang W, Jing Y, Yi S, Farooq U, Shi J, Pang N, Rong N, Xu L. Non-Cavitation Targeted Microbubble-Mediated Single-Cell Sonoporation. MICROMACHINES 2022; 13:mi13010113. [PMID: 35056278 PMCID: PMC8780975 DOI: 10.3390/mi13010113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023]
Abstract
Sonoporation employs ultrasound accompanied by microbubble (MB) cavitation to induce the reversible disruption of cell membranes and has been exploited as a promising intracellular macromolecular delivery strategy. Due to the damage to cells resulting from strong cavitation, it is difficult to balance efficient delivery and high survival rates. In this paper, a traveling surface acoustic wave (TSAW) device, consisting of a TSAW chip and a polydimethylsiloxane (PDMS) channel, was designed to explore single-cell sonoporation using targeted microbubbles (TMBs) in a non-cavitation regime. A TSAW was applied to precisely manipulate the movement of the TMBs attached to MDA-MB-231 cells, leading to sonoporation at a single-cell level. The impact of input voltage and the number of TMBs on cell sonoporation was investigated. In addition, the physical mechanisms of bubble cavitation or the acoustic radiation force (ARF) for cell sonoporation were analyzed. The TMBs excited by an ARF directly propelled cell membrane deformation, leading to reversible perforation in the cell membrane. When two TMBs adhered to the cell surface and the input voltage was 350 mVpp, the cell sonoporation efficiency went up to 83%.
Collapse
Affiliation(s)
- Xiufang Liu
- College of Medicine and Biological Information Engineering, Northeastern University, 195 Innovation Road, Shenyang 110016, China; (X.L.); (N.P.)
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Wenjun Zhang
- Department of Mechanical and Electrical Engineering, Gannan University of Science and Technology, 156 Kejia Avenue, Ganzhou 341000, China;
| | - Yanshu Jing
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
- Department of Biomedical Engineering, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Shasha Yi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Umar Farooq
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Jingyao Shi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Na Pang
- College of Medicine and Biological Information Engineering, Northeastern University, 195 Innovation Road, Shenyang 110016, China; (X.L.); (N.P.)
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
| | - Ning Rong
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (Y.J.); (S.Y.); (U.F.); (J.S.)
- Correspondence: (N.R.); (L.X.); Tel.: +86-024-83683200 (L.X.)
| | - Lisheng Xu
- College of Medicine and Biological Information Engineering, Northeastern University, 195 Innovation Road, Shenyang 110016, China; (X.L.); (N.P.)
- Neusoft Research of Intelligent Healthcare Technology, Co., Ltd., Shenyang 110167, China
- Correspondence: (N.R.); (L.X.); Tel.: +86-024-83683200 (L.X.)
| |
Collapse
|
31
|
Michon S, Rodier F, Yu FTH. Targeted Anti-Cancer Provascular Therapy Using Ultrasound, Microbubbles, and Nitrite to Increase Radiotherapy Efficacy. Bioconjug Chem 2022; 33:1093-1105. [PMID: 34990112 DOI: 10.1021/acs.bioconjchem.1c00510] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hypoxia is an important mechanism of resistance to radiation therapy in many human malignancies including prostate cancer. It has been recently shown that ultrasound targeted microbubble cavitation (UTMC) can increase blood perfusion in skeletal muscle by triggering nitric oxide signaling. Interestingly, this effect was amplified with a sodium nitrite coinjection. Since sodium nitrite has been shown to synergize with radiotherapy (RT), we hypothesized that UTMC with a sodium nitrite coinjection could further radiosensitize solid tumors by increasing blood perfusion and thus reduce tumor hypoxia. We evaluated (1) the ability of UTMC with and without nitrite to increase perfusion in muscle (mouse hindlimbs) and human prostate tumors using different pulse lengths and pressure; (2) the efficacy of this approach as a provascular therapy given directly before RT in the human prostate subcutaneous xenografts PC3 tumor model. Using long pulses with various pressures, in muscle, the provascular response following UTMC was strong (6.61 ± 4.41-fold increase in perfusion post-treatment). In tumors, long pulses caused an increase in perfusion (2.42 ± 1.38-fold) at lower mechanical index (MI = 0.25) but not at higher MI (0.375, 0.5, and 0.750) when compared to control (no UTMC). However, when combined with RT, UTMC with long pulses (MI = 0.25) did not improve tumor growth inhibition. With short pulses, in muscle, the provascular response following UTMC (SONOS) + nitrite was strong (13.74 ± 8.60-fold increase in perfusion post-treatment). In tumors, UTMC (SONOS) + nitrite also caused a provascular response (1.94 ± 1.20-fold increase in perfusion post-treatment) that lasted for at least 10 min, but not with nitrite alone. Interestingly, the blunted provascular response observed for long pulses at higher MI without nitrite was reversed with the addition of nitrite. UTMC (SONOS) with and without nitrite caused an increase in perfusion in tumors. The provascular response observed for UTMC (SONOS) + nitrite was confirmed by histology. Finally, there was an improved growth inhibition for the 8 Gy RT dose + nitrite + UTMC group vs 8 Gy RT + nitrite alone. This effect was not significant with mice treated by UTMC + nitrite and receiving doses of 0 or 2 Gy RT. In conclusion, UTMC + nitrite increased blood flow leading to an increased efficacy of higher doses of RT in our tumor model, warranting further study of this strategy.
Collapse
Affiliation(s)
- Simon Michon
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, Québec H2X 0A9, Canada.,Institut de Génie Biomédical, Université de Montréal, Montréal, Québec H3T 1J4, Canada.,Département de Radiologie, Radio-Oncologie Et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, Québec H2X 0A9, Canada.,Département de Radiologie, Radio-Oncologie Et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - François T H Yu
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) et Institut du Cancer de Montréal, Montréal, Québec H2X 0A9, Canada.,Institut de Génie Biomédical, Université de Montréal, Montréal, Québec H3T 1J4, Canada.,Département de Radiologie, Radio-Oncologie Et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
32
|
Shen LM, Li MC, Wei WJ, Guan X, Liu J. In Vitro Neuroprotective Effects of Macrophage Membrane-Derived Curcumin-Loaded Carriers against 1-Methyl-4-phenylpyridinium-Induced Neuronal Damage. ACS OMEGA 2021; 6:32133-32141. [PMID: 34870034 PMCID: PMC8637945 DOI: 10.1021/acsomega.1c04894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
Curcumin (CUR) possesses neuroprotective effects. However, its clinical therapeutic efficacy is limited because of its low systemic bioavailability due to poor water solubility and fast metabolism. Herein, we designed biomimetic therapeutic nanovesicles (NVs) with enhanced performance and biocompatibility for the intracellular delivery of hydrophobic CUR. Cell membrane NVs were constructed to function as drug carriers by the serial extrusion of macrophages using filters with decreasing pore sizes. Various CUR loading strategies were also evaluated. Furthermore, the neuroprotective effects of the CUR-loaded NVs (NVs-CUR) against 1-methyl-4-phenylpyridinium (MPP+)-induced neuronal degeneration were studied thoroughly. CUR-loaded NVs were readily taken up by neurons in vitro, and the survival rate of MPP+-induced primary neurons increased from 65.37 ± 6.37 to 90.91 ± 3.18% after pretreatment with NVs-CUR. Compared with traditional Parkinson's disease chemotherapeutic treatment, NV formulations can improve the bioavailability of this drug. NVs are expected to become a new and effective drug-delivery platform for further applications in the field of central nervous system therapy.
Collapse
Affiliation(s)
- Li-Ming Shen
- Stem
Cell Clinical Research Center, The First
Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian 116011, China
| | - Meng-Chu Li
- Stem
Cell Clinical Research Center, The First
Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian 116011, China
| | - Wen-Juan Wei
- Stem
Cell Clinical Research Center, The First
Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian 116011, China
| | - Xin Guan
- Stem
Cell Clinical Research Center, The First
Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian 116011, China
| | - Jing Liu
- Stem
Cell Clinical Research Center, The First
Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian 116011, China
- Dalian
Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, Dalian High-Tech
Park, Dalian 116023, China
| |
Collapse
|
33
|
Sridharan A, Hwang M, Kutty S, McCarville MB, Paltiel HJ, Piskunowicz M, Shellikeri S, Silvestro E, Taylor GA, Didier RA. Translational research in pediatric contrast-enhanced ultrasound. Pediatr Radiol 2021; 51:2425-2436. [PMID: 33991196 PMCID: PMC11459366 DOI: 10.1007/s00247-021-05095-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/21/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
The role of contrast-enhanced ultrasound (CEUS) imaging is being widely explored by various groups for its use in the pediatric population. Clinical implementation of new diagnostic or therapeutic techniques requires extensive and meticulous preclinical testing and evaluation. The impact of CEUS will be determined in part by the extent to which studies are oriented specifically toward a pediatric population. Rather than simply applying principles and techniques used in the adult population, these studies are expected to advance and augment preexisting knowledge with pediatric-specific information. To further develop this imaging modality for use in children, pediatric-focused preclinical research is essential. In this paper we describe the development and implementation of the pediatric-specific preclinical animal and phantom models that are being used to evaluate CEUS with the goal of clinical translation to children.
Collapse
Affiliation(s)
- Anush Sridharan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.
| | - Misun Hwang
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Shelby Kutty
- Taussig Heart Center, Johns Hopkins University, Baltimore, MD, USA
| | - M Beth McCarville
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Harriet J Paltiel
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Sphoorti Shellikeri
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Elizabeth Silvestro
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - George A Taylor
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Harvard Medical School, Boston, MA, USA
| | - Ryne A Didier
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Kim J, Wang Q, Zhang S, Yoon S. Compressed Sensing-Based Super-Resolution Ultrasound Imaging for Faster Acquisition and High Quality Images. IEEE Trans Biomed Eng 2021; 68:3317-3326. [PMID: 33793396 PMCID: PMC8609474 DOI: 10.1109/tbme.2021.3070487] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
GOAL Typical SRUS images are reconstructed by localizing ultrasound microbubbles (MBs) injected in a vessel using normalized 2-dimensional cross-correlation (2DCC) between MBs signals and the point spread function of the system. However, current techniques require isolated MBs in a confined area due to inaccurate localization of densely populated MBs. To overcome this limitation, we developed the ℓ1-homotopy based compressed sensing (L1H-CS) based SRUS imaging technique which localizes densely populated MBs to visualize microvasculature in vivo. METHODS To evaluate the performance of L1H-CS, we compared the performance of 2DCC, interior-point method based compressed sensing (CVX-CS), and L1H-CS algorithms. Localization efficiency was compared using axially and laterally aligned point targets (PTs) with known distances and randomly distributed PTs generated by simulation. We developed post-processing techniques including clutter reduction, noise equalization, motion compensation, and spatiotemporal noise filtering for in vivo imaging. We then validated the capabilities of L1H-CS based SRUS imaging technique with high-density MBs in a mouse tumor model, kidney, and zebrafish dorsal trunk, and brain. RESULTS Compared to 2DCC and CVX-CS algorithms, L1H-CS achieved faster data acquisition time and considerable improvement in SRUS image quality. CONCLUSIONS AND SIGNIFICANCE These results demonstrate that the L1H-CS based SRUS imaging technique has the potential to examine microvasculature with reduced acquisition and reconstruction time to acquire enhanced SRUS image quality, which may be necessary to translate it into clinics.
Collapse
|
35
|
Lin B, Du H, Fan J, Huang D, Gao F, Li J, Zhang Y, Feng G, Dai T, Du X. Radioimmunotherapy Combined With Low-Intensity Ultrasound and Microbubbles: A Potential Novel Strategy for Treatment of Solid Tumors. Front Oncol 2021; 11:750741. [PMID: 34745976 PMCID: PMC8570127 DOI: 10.3389/fonc.2021.750741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
The prognosis of advanced malignant tumors is very poor, and effective treatment is limited. Radioimmunotherapy (RIT) is a novel treatment method. However, its anti-tumor effect is relatively low in solid tumors, which is mainly due to the blood-tumor barrier preventing RIT from penetrating the tumor, resulting in an insufficient dose. Low-intensity ultrasound with microbubbles (USMB) has proven capable of opening the blood-tumor barrier. The combination of the two technologies may overcome the poor anti-tumor effect of RIT and promote the clinical application of RIT in solid tumors. In this article, we reviewed the current research status of RIT in the treatment of solid tumors and the opportunities and challenges of USMB combined with RIT.
Collapse
Affiliation(s)
- Binwei Lin
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Huan Du
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jinjia Fan
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Dan Huang
- Radiology Department, Mianyang Central Hospital, Mianyang, China
| | - Feng Gao
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Jie Li
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Yu Zhang
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Gang Feng
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Tangzhi Dai
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| | - Xiaobo Du
- Department of Oncology, Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang Central Hospital, Mianyang, China
| |
Collapse
|
36
|
Peng T, Huang Y, Feng X, Zhu C, Yin S, Wang X, Bai X, Pan X, Wu C. TPGS/hyaluronic acid dual-functionalized PLGA nanoparticles delivered through dissolving microneedles for markedly improved chemo-photothermal combined therapy of superficial tumor. Acta Pharm Sin B 2021; 11:3297-3309. [PMID: 34729317 PMCID: PMC8546669 DOI: 10.1016/j.apsb.2020.11.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 12/18/2022] Open
Abstract
Nanoparticles (NPs) have shown potential in cancer therapy, while a single administration conferring a satisfactory outcome is still unavailable. To address this issue, the dissolving microneedles (DMNs) were developed to locally deliver functionalized NPs with combined chemotherapy and photothermal therapy (PTT). α-Tocopheryl polyethylene glycol succinate (TPGS)/hyaluronic acid (HA) dual-functionalized PLGA NPs (HD10 NPs) were fabricated to co-load paclitaxel and indocyanine green. HD10 NPs significantly enhanced the cytotoxicity of low-dose paclitaxel because of active and mitochondrial targeting by HA and TPGS, respectively. PTT could further sensitize tumor cells toward chemotherapy by promoting apoptosis into the advanced period, highly activating caspase 3 enzyme, and significantly reducing the expression of survivin and MMP-9 proteins. Further, the anti-tumor effects of HD10 NPs delivered through different administration routes were conducted on the 4T1 tumor-bearing mice. After a single administration, HD10 NPs delivered with DMNs showed the best anti-tumor effect when giving chemotherapy alone. As expected, the anti-tumor effect was profoundly enhanced after combined therapy, and complete tumor ablation was achieved in the mice treated with DMNs and intra-tumor injection. Moreover, DMNs showed better safety due to moderate hyperthermia. Therefore, the DMNs along with combined chemo-photothermal therapy provide a viable treatment option for superficial tumors.
Collapse
Affiliation(s)
- Tingting Peng
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yao Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiaoqian Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chune Zhu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shi Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinyi Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Xuequn Bai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Corresponding authors.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 510632, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Corresponding authors.
| |
Collapse
|
37
|
Snipstad S, Vikedal K, Maardalen M, Kurbatskaya A, Sulheim E, Davies CDL. Ultrasound and microbubbles to beat barriers in tumors: Improving delivery of nanomedicine. Adv Drug Deliv Rev 2021; 177:113847. [PMID: 34182018 DOI: 10.1016/j.addr.2021.113847] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022]
Abstract
Successful delivery of drugs and nanomedicine to tumors requires a functional vascular network, extravasation across the capillary wall, penetration through the extracellular matrix, and cellular uptake. Nanomedicine has many merits, but penetration deep into the tumor interstitium remains a challenge. Failure of cancer treatment can be caused by insufficient delivery of the therapeutic agents. After intravenous administration, nanomedicines are often found in off-target organs and the tumor extracellular matrix close to the capillary wall. With circulating microbubbles, ultrasound exposure focused toward the tumor shows great promise in improving the delivery of therapeutic agents. In this review, we address the impact of focused ultrasound and microbubbles to overcome barriers for drug delivery such as perfusion, extravasation, and transport through the extracellular matrix. Furthermore, we discuss the induction of an immune response with ultrasound and delivery of immunotherapeutics. The review discusses mainly preclinical results and ends with a summary of ongoing clinical trials.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Krister Vikedal
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Matilde Maardalen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna Kurbatskaya
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | | |
Collapse
|
38
|
Chung IJ, Moon H, Jeon SI, Lee HJ, Ahn CH. Ultrasound-triggered imaging and drug delivery using microbubble-self-aggregate complexes. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:57-76. [PMID: 34503403 DOI: 10.1080/09205063.2021.1976362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Co-delivery of microbubbles (MBs) with anticancer drugs is a promising theranostic approach that can enhance both the ultrasound contrast and local extravasation of drugs with the sonoporation effect. The simultaneous administration of MBs and hydrophobic drugs, however, is still challenging due to the limitations in drug loading or undesirable stabilization of MBs. In this research, MB-self-aggregate complexes (MB-SAs) were newly fabricated for the encapsulation of hydrophobic drugs, and their theranostic properties are investigated in vitro and in vivo. Glycol chitosan self-aggregates (GC-SAs) loaded with hydrophobic drugs or dyes were chemically conjugated on the surface MBs. Their conjugation ratio was determined to be 73.9%, and GC-SAs on MBs did not affect the stability of MBs. GC-SA attached MBs (GC@MBs) were successfully visualized with low-intensity insonation and showed enhanced cellular uptake via the sonoporation effect. In vivo biodistribution of GC@MBs was examined with tumor-bearing mice, confirming that their accumulation at the tumor site increased by 1.85 times after ultrasound irradiation. The anticancer drug-loaded GC@MBs also exhibited 10% higher cytotoxicity under ultrasound flash. In conclusion, it was expected that GC@MBs could be used both as an ultrasound contrast agent and a drug carrier even with conventional ultrasonic devices.
Collapse
Affiliation(s)
- In Jae Chung
- Department of Materials Science and Engineering, Research Institute of Advanced Materials (RIAM), Seoul National University, Gwanak-gu, Seoul, Korea
| | | | - Seong Ik Jeon
- Department of Materials Science and Engineering, Research Institute of Advanced Materials (RIAM), Seoul National University, Gwanak-gu, Seoul, Korea
| | - Hak Jong Lee
- IMGT Co., Ltd, Seongnam, Korea.,Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol-Hee Ahn
- Department of Materials Science and Engineering, Research Institute of Advanced Materials (RIAM), Seoul National University, Gwanak-gu, Seoul, Korea
| |
Collapse
|
39
|
Diltemiz SE, Tavafoghi PhD M, Roberto de Barros N, Kanada M, Heinamaki J, Contag C, Seidlits S, Ashammakhi N. USE OF ARTIFICIAL CELLS AS DRUG CARRIERS. MATERIALS CHEMISTRY FRONTIERS 2021; 5:6672-6692. [PMID: 38344270 PMCID: PMC10857888 DOI: 10.1039/d1qm00717c] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Cells are the fundamental functional units of biological systems and mimicking their size, function and complexity is a primary goal in the development of new therapeutic strategies. Recent advances in chemistry, synthetic biology and material science have enabled the development of cell membrane-based drug delivery systems (DDSs), often referred to as "artificial cells" or protocells. Artificial cells can be made by removing functions from natural systems in a top-down manner, or assembly from synthetic, organic or inorganic materials, through a bottom-up approach where simple units are integrated to form more complex structures. This review covers the latest advances in the development of artificial cells as DDSs, highlighting how their designs have been inspired by natural cells or cell membranes. Advancement of artificial cell technologies has led to a set of drug carriers with effective and controlled release of a variety of therapeutics for a range of diseases, and with increasing complexity they will have a greater impact on therapeutic designs.
Collapse
Affiliation(s)
- Sibel Emir Diltemiz
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Chemistry, Eskisehir Technical University, Eskisehir, Turkey
| | - Maryam Tavafoghi PhD
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Natan Roberto de Barros
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Bioprocess and Biotechnology Engineering, São Paulo State University (Unesp), School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil
| | - Masamitsu Kanada
- Institute for Quantitative Health Science and Engineering (IQ), Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Jyrki Heinamaki
- Institute of Pharmacy, Faculty of Medicine, University of Tartu, Nooruse Str. 1, EE-50411 Tartu, Estonia
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering (IQ) and Departments of Biomedical Engineering (BME), and Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Stephanie Seidlits
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
40
|
Krafft MP, Riess JG. Therapeutic oxygen delivery by perfluorocarbon-based colloids. Adv Colloid Interface Sci 2021; 294:102407. [PMID: 34120037 DOI: 10.1016/j.cis.2021.102407] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
After the protocol-related indecisive clinical trial of Oxygent, a perfluorooctylbromide/phospholipid nanoemulsion, in cardiac surgery, that often unduly assigned the observed untoward effects to the product, the development of perfluorocarbon (PFC)-based O2 nanoemulsions ("blood substitutes") has come to a low. Yet, significant further demonstrations of PFC O2-delivery efficacy have continuously been reported, such as relief of hypoxia after myocardial infarction or stroke; protection of vital organs during surgery; potentiation of O2-dependent cancer therapies, including radio-, photodynamic-, chemo- and immunotherapies; regeneration of damaged nerve, bone or cartilage; preservation of organ grafts destined for transplantation; and control of gas supply in tissue engineering and biotechnological productions. PFC colloids capable of augmenting O2 delivery include primarily injectable PFC nanoemulsions, microbubbles and phase-shift nanoemulsions. Careful selection of PFC and other colloid components is critical. The basics of O2 delivery by PFC nanoemulsions will be briefly reminded. Improved knowledge of O2 delivery mechanisms has been acquired. Advanced, size-adjustable O2-delivering nanoemulsions have been designed that have extended room-temperature shelf-stability. Alternate O2 delivery options are being investigated that rely on injectable PFC-stabilized microbubbles or phase-shift PFC nanoemulsions. The latter combine prolonged circulation in the vasculature, capacity for penetrating tumor tissues, and acute responsiveness to ultrasound and other external stimuli. Progress in microbubble and phase-shift emulsion engineering, control of phase-shift activation (vaporization), understanding and control of bubble/ultrasound/tissue interactions is discussed. Control of the phase-shift event and of microbubble size require utmost attention. Further PFC-based colloidal systems, including polymeric micelles, PFC-loaded organic or inorganic nanoparticles and scaffolds, have been devised that also carry substantial amounts of O2. Local, on-demand O2 delivery can be triggered by external stimuli, including focused ultrasound irradiation or tumor microenvironment. PFC colloid functionalization and targeting can help adjust their properties for specific indications, augment their efficacy, improve safety profiles, and expand the range of their indications. Many new medical and biotechnological applications involving fluorinated colloids are being assessed, including in the clinic. Further uses of PFC-based colloidal nanotherapeutics will be briefly mentioned that concern contrast diagnostic imaging, including molecular imaging and immune cell tracking; controlled delivery of therapeutic energy, as for noninvasive surgical ablation and sonothrombolysis; and delivery of drugs and genes, including across the blood-brain barrier. Even when the fluorinated colloids investigated are designed for other purposes than O2 supply, they will inevitably also carry and deliver a certain amount of O2, and may thus be considered for O2 delivery or co-delivery applications. Conversely, O2-carrying PFC nanoemulsions possess by nature a unique aptitude for 19F MR imaging, and hence, cell tracking, while PFC-stabilized microbubbles are ideal resonators for ultrasound contrast imaging and can undergo precise manipulation and on-demand destruction by ultrasound waves, thereby opening multiple theranostic opportunities.
Collapse
Affiliation(s)
- Marie Pierre Krafft
- University of Strasbourg, Institut Charles Sadron (CNRS), 23 rue du Loess, 67034 Strasbourg, France.
| | - Jean G Riess
- Harangoutte Institute, 68160 Ste Croix-aux-Mines, France
| |
Collapse
|
41
|
Zhang C, Zhao Y, Zhang E, Jiang M, Zhi D, Chen H, Cui S, Zhen Y, Cui J, Zhang S. Co-delivery of paclitaxel and anti-VEGF siRNA by tripeptide lipid nanoparticle to enhance the anti-tumor activity for lung cancer therapy. Drug Deliv 2021; 27:1397-1411. [PMID: 33096948 PMCID: PMC7594708 DOI: 10.1080/10717544.2020.1827085] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The combination of chemotherapeutic drug paclitaxel (PTX) and VEGF siRNA could inhibit cancer development with synergistic efficacy. However, efficient and safe delivery systems with high encapsulation efficiency of PTX and a long-time release of drugs are urgently needed. In this study, novel nanoparticles (PTX/siRNA/FALS) were constructed by using tripeptide lipid (L), sucrose laurate (S), and folate-PEG2000-DSPE (FA) to co-deliver PTX and siRNA. The cancer cell targeting nanoparticle carrier (PTX/siRNA/FALS) showed anticipated PTX encapsulation efficiency, siRNA retardation ability, improved cell uptake and sustained and controlled drug release. It led to significant anti-tumor activity in vitro and in vivo by efficient inhibition of VEGF expression and induction of cancer cell apoptosis. Importantly, the biocompatibility of the carriers and low dosage of PTX required for effective therapy greatly reduced the toxicity to mice. The targeting nanoparticles show potential as an effective co-delivery platform for RNAi and chemotherapy drugs, aiming to improve the efficacy of cancer therapy.
Collapse
Affiliation(s)
- Chuanmin Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China.,Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Enxia Zhang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Meilin Jiang
- College of Postgraduate, Jinzhou Medical University, Jinzhou, China
| | - Defu Zhi
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Huiying Chen
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Shaohui Cui
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Jingnan Cui
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| |
Collapse
|
42
|
Drug Delivery by Ultrasound-Responsive Nanocarriers for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13081135. [PMID: 34452096 PMCID: PMC8397943 DOI: 10.3390/pharmaceutics13081135] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Conventional cancer chemotherapies often exhibit insufficient therapeutic outcomes and dose-limiting toxicity. Therefore, there is a need for novel therapeutics and formulations with higher efficacy, improved safety, and more favorable toxicological profiles. This has promoted the development of nanomedicines, including systems for drug delivery, but also for imaging and diagnostics. Nanoparticles loaded with drugs can be designed to overcome several biological barriers to improving efficiency and reducing toxicity. In addition, stimuli-responsive nanocarriers are able to release their payload on demand at the tumor tissue site, preventing premature drug loss. This review focuses on ultrasound-triggered drug delivery by nanocarriers as a versatile, cost-efficient, non-invasive technique for improving tissue specificity and tissue penetration, and for achieving high drug concentrations at their intended site of action. It highlights aspects relevant for ultrasound-mediated drug delivery, including ultrasound parameters and resulting biological effects. Then, concepts in ultrasound-mediated drug delivery are introduced and a comprehensive overview of several types of nanoparticles used for this purpose is given. This includes an in-depth compilation of the literature on the various in vivo ultrasound-responsive drug delivery systems. Finally, toxicological and safety considerations regarding ultrasound-mediated drug delivery with nanocarriers are discussed.
Collapse
|
43
|
Zhang Y, Tang N, Huang L, Qiao W, Zhu Q, Liu Z. Effect of diagnostic ultrasound and microbubble-enhanced chemotherapy on metastasis of rabbit VX2 tumor. Med Phys 2021; 48:3927-3935. [PMID: 33774845 DOI: 10.1002/mp.14867] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Ultrasound-targeted microbubble destruction (UTMD) has been widely applied to enhance chemotherapy of tumors, yet few studies have focused on the metastatic potential induced by UTMD. This study aimed to explore the metastasis of VX2 tumors after treatment with UTMD and chemotherapy. METHODS Forty-four New Zealand rabbits bearing subcutaneous VX2 tumors were enrolled for the treatment of UTMD with chemotherapy. For UTMD, the tumors were insonated using two pulsing protocols of diagnostic ultrasound (DUS, VINNO and ECARE) with a mechanical index (MI) of 0.29-0.33, tone burst of 8.0 cycles, and frequencies of 3-4 MHz. A total dose of 2 ml SonoVue® was injected intermittently during 10-min UTMD exposure. The combination therapy was treated using doxorubicin (DOX, 2 mg/kg) and DUS, while the tumors treated using DOX only served as the control. Tumor size was measured using the tumor volume formula. Survival time was observed until animal death or the end of the study (120 days). Specific organs (lung, liver, kidney, and brain) were removed for metastatic evaluation. RESULTS There were no statistical differences in overall metastasis classification and individual organ metastases among all groups (P > 0.05). The tumor growth rate only showed inhibition on the 5th day (P < 0.01). The survival time did not demonstrate any significant difference between UTMD and chemotherapy only (P > 0.05). CONCLUSIONS UTMD using long-pulse DUS with commercial microbubbles did not pose a risk of metastasis enhancement in DOX chemotherapy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Najiao Tang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Leidan Huang
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wei Qiao
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China.,General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
44
|
Abstract
Contrary to the fact that capillary action is ubiquitous in our daily lives, its role in drug delivery has not attracted attention. Therefore, its application in medicine and disease treatment has not been actively developed. This perspective begins by reviewing the principles, advantages, and limitations of the three existing drug delivery strategies: non-covalent interaction, cavity loading, and covalent conjugation. Then, we discussed the principle of capillary action in drug delivery and the influencing factors that determine its performance. To illustrate the advantages of capillary action over existing drug delivery strategies and how the capillary action could potentially address the shortcomings of the existing drug delivery strategies, we described five examples of using capillary action to design drug delivery platforms for disease treatment: marker pen for topical and transdermal drug delivery, microneedle patch with a sponge container for pulsatile drug delivery, core-shell scaffold for sustained release of growth factors, oral bolus for insulin delivery to the esophagus, and semi-hollow floating ball for intravesical and gastroprotective drug delivery. Each of the five drug delivery platforms exhibits certain unique functions that existing drug delivery technologies cannot easily achieve, hence expected to solve specific practical medical problems that are not satisfactorily resolved. As people pay more attention to capillary action and develop more drug delivery platforms, more unique functions and characteristics of capillary action in drug delivery will be explored. Thus, capillary action could become an important choice for drug delivery systems to improve therapeutic drug efficacy, treat diseases, and improve human health.
Collapse
Affiliation(s)
- Xiaosi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Yue Zhao
- School of Software, Northwestern Polytechnical University, Taicang, Jiangsu 215400, China
| | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| |
Collapse
|
45
|
Aydın M, Özdemir E, Altun Z, Kılıç S, Aktaş S. Evaluation of Liposomal and Microbubbles Mediated Delivery of Doxorubicin in Two-Dimensional (2D) and Three-Dimensional (3D) Models for Breast Cancer. Eur J Breast Health 2021; 17:274-282. [PMID: 34263156 DOI: 10.4274/ejbh.galenos.2021.6255] [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: 11/20/2020] [Accepted: 02/28/2021] [Indexed: 12/12/2022]
Abstract
Objective Liposomal cancer treatment strategies are useful in removing the side effects that were the main concern in recent years. In this study, we prepared microbubble (MBs) conjugated with DOX-loaded liposomes (DOX-loaded MBs) and investigated their effectiveness in in vitro breast cancer cells in two dimensions (2D) and three dimensions (3D). Materials and Methods With this aim, breast cancer cells with different features (4T1, MDA-MB231, MCF-7) were growth in 2D and 3D dimensions. The cytotoxic and cell death effects under different conditions, durations and doses were evaluated with WST-1, trypan-blue, colony counts. Apoptotic effects were investigated with flow cytometric Annexin-V-PI and immunohistochemical (Ki-67, caspase 3, 8, 9) methods. Results After free DOX and LipoDOX were applied, the proliferation index of three cell lines reduced. Intrinsic and extrinsic apoptotic pathways were activated in both 2D and 3D models. However, this effect was observed at lower levels in the 3D model due to the difficulty of diffusion of DOX into the spheroids. Additionally, the suitability of the 3D model for breast cancer cells was supported by formation of ductus-like structures and spheroids. Cell deaths were not observed significantly with the DOX-loaded microbubbles due to rising of MBs to the surface and not reaching spheroids held in matrigel of 3D model. Conclusion DOX and LipoDOX showed anti-proliferative and apoptosis-inducing effects in breast cancer cells. However, these effects indicated variability depending on the cell lines and 2D or 3D model types.
Collapse
Affiliation(s)
- Melek Aydın
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Turkey
| | - Ekrem Özdemir
- Department of Chemical Engineering, İzmir Institute of Technology, İzmir, Turkey
| | - Zekiye Altun
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Turkey
| | - Sevgi Kılıç
- Department of Chemical Engineering, İzmir Institute of Technology, İzmir, Turkey
| | - Safiye Aktaş
- Department of Basic Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Turkey
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Wang L, Zhu B, Huang J, Xiang X, Tang Y, Ma L, Yan F, Cheng C, Qiu L. Ultrasound-targeted microbubble destruction augmented synergistic therapy of rheumatoid arthritis via targeted liposomes. J Mater Chem B 2021; 8:5245-5256. [PMID: 32432638 DOI: 10.1039/d0tb00430h] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Rheumatoid arthritis (RA) can lead to joint destruction and deformity, which is a significant cause of the loss of the young and middle-aged labor force. However, the treatment of RA is still filled with challenges. Though dexamethasone, one of the glucocorticoids, is commonly used in the treatment of RA, its clinical use is limited because of the required high-dose and long-term use, unsatisfactory therapeutic effects, and various side-effects. Ultrasound-targeted microbubble destruction (UTMD) can augment the ultrasonic cavitation effects and trigger drug release from targeted nanocarriers in the synovial cavity, which makes it a more effective synergistic treatment strategy for RA. In this work, we aim to utilize the UTMD effect to augment the synergistic therapy of RA by using polyethylene glycol (PEG)-modified folate (FA)-conjugated liposomes (LPs) loaded with dexamethasone sodium phosphate (DexSP) (DexSP@LPs-PEG-FA). The UTMD-mediated DexSP@LPs-PEG-FA for targeted delivery of DexSP including a synergistic ultrasonic cavitation effect and drug therapy were investigated through in vitro RAW264.7 cell experiments and in vivo collagen-induced arthritis SD rat model animal experiments. The results show the DexSP release from targeted liposomes was improved under the UTMD effect. Likewise, the folate-conjugated liposomes displayed targeting association to RAW264.7 cells. Together with the application of ultrasound and microbubbles, liposomes-delivered DexSP potently reduced joints swelling, bone erosion, and inflammation in both joints and serum with a low dose. These results demonstrated that UTMD-mediated folate-conjugated liposomes are not only a promising method for targeted synergistic treatment of RA but also may show high potential for serving as nanomedicines for many other biomedical fields.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bihui Zhu
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jianbo Huang
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xi Xiang
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuanjiao Tang
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lang Ma
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Feng Yan
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China and Department of Chemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Li Qiu
- Department of Medical Ultrasound, Laboratory of Ultrasound Imaging Drug, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
49
|
Liu Y, Zhou J, Li Q, Li L, Jia Y, Geng F, Zhou J, Yin T. Tumor microenvironment remodeling-based penetration strategies to amplify nanodrug accessibility to tumor parenchyma. Adv Drug Deliv Rev 2021; 172:80-103. [PMID: 33705874 DOI: 10.1016/j.addr.2021.02.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Remarkable advances in nano delivery systems have provided new hope for tumor prevention, diagnosis and treatment. However, only limited clinical therapeutic effects against solid tumors were achieved. One of the main reasons is the presence of abundant physiological and pathological barriers in vivo that impair tumoral penetration and distribution of the nanodrugs. These barriers are related to the components of tumor microenvironment (TME) including abnormal tumor vasculature, rich composition of the extracellular matrix (ECM), and abundant stroma cells. Herein, we review the advanced strategies of TME remodeling to overcome these biological obstacles against nanodrug delivery. This review aims to offer a perspective guideline for the implementation of promising approaches to facilitate intratumoral permeation of nanodrugs through alleviation of biological barriers. At the same time, we analyze the advantages and disadvantages of the corresponding methods and put forward possible directions for the future researches.
Collapse
Affiliation(s)
- Yanhong Liu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jiyuan Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Qiang Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Lingchao Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yue Jia
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Feiyang Geng
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
50
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|