1
|
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
|
2
|
Tian Y, He X, Yuan Y, Zhang S, Wang C, Dong J, Liu Z, Jing H. TME-Responsive Nanoplatform with Glutathione Depletion for Enhanced Tumor-Specific Mild Photothermal/Gene/Ferroptosis Synergistic Therapy. Int J Nanomedicine 2024; 19:9145-9160. [PMID: 39258005 PMCID: PMC11386068 DOI: 10.2147/ijn.s475698] [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: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
Background Triple negative breast cancer (TNBC) is one of the worst prognosis types of breast cancer that urgently needs effective therapy methods. However, cancer is a complicated disease that usually requires multiple treatment modalities. Methods A tumor microenvironment (TME)-responsive PFC/TRIM37@Fe-TA@HA (abbreviated as PTFTH) nanoplatform was constructed by coating Fe3+ and tannic acid (TA) on the surface of TRIM37-siRNA loaded phase-transition perfluorocarbon (PFC) nanodroplets and further modifying them with hyaluronic acid (HA) to achieve tumor-specific mild photothermal/gene/ferroptosis synergistic therapy (MPTT/GT/ Ferroptosis) in vitro. Once internalized into tumor cells through CD44 receptor-mediated active targeting, the HA shell of PTFTH would be preliminarily disassembled by hyaluronidase (HAase) to expose the Fe-TA metal-phenolic networks (MPNs), which would further degrade in response to an acidic lysosomal environment, leading to HAase/pH dual-responsive release of Fe3+ and PFC/TRIM37. Results PTFTH showed good biocompatibility in vitro. On the one hand, the released Fe3+ could deplete the overexpressed glutathione (GSH) through redox reactions and produce Fe2+, which in turn converts endogenous H2O2 into highly cytotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT). On the other hand, the local hyperthermia generated by PTFTH under 808 nm laser irradiation could not only improve CDT efficacy through accelerating the Fe2+-mediated Fenton reaction, but also enhance TRIM37-siRNA delivery for gene therapy (GT). The consumption of GSH and accumulation of •OH synergistically augmented intracellular oxidative stress, resulting in substantial tumor cell ferroptosis. Moreover, PTFTH possessed outstanding contrast enhanced ultrasound (CEUS), photoacoustic imaging (PAI) and magnetic resonance imaging (MRI) ability. Conclusion This PTFTH based multiple-mode therapeutic strategy has successfully achieved a synergistic anticancer effect in vitro and has the potential to be translated into clinical application for tumor therapy in future.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Shijie Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| |
Collapse
|
3
|
Rajankar N, Aalhate M, Mahajan S, Maji I, Gupta U, Nair R, Paul P, Singh PK. Unveiling multifaceted avenues of echogenic liposomes: Properties, preparation, and potential applications. J Drug Deliv Sci Technol 2024; 99:105931. [DOI: 10.1016/j.jddst.2024.105931] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Li X, Cai J, Zhang H, Sun S, Zhao S, Wang Z, Nie X, Xu C, Zhang Y, Xiao H. A Trisulfide Bond Containing Biodegradable Polymer Delivering Pt(IV) Prodrugs to Deplete Glutathione and Donate H 2S to Boost Chemotherapy and Antitumor Immunity. ACS NANO 2024; 18:7852-7867. [PMID: 38437513 DOI: 10.1021/acsnano.3c06194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
The clinical application of cisplatin (CisPt) is limited by its dose-dependent toxicity. To overcome this, we developed reduction-responsive nanoparticles (NP(3S)s) for the targeted delivery of a platinum(IV) (Pt(IV)) prodrug to improve efficacy and reduce the toxicity. NP(3S)s could release Pt(II) and hydrogen sulfide (H2S) upon encountering intracellular glutathione, leading to potent anticancer effects. Notably, NP(3S)s induced DNA damage and activated the STING pathway, which is a known promoter for T cell activation. Comparative RNA profiling revealed that NP(3S)s outperformed CisPt in enhancing T cell immunity, antitumor immunity, and oxidative stress pathways. In vivo experiments showed that NP(3S)s accumulated in tumors, promoting CD8+ T cell infiltration and boosting antitumor immunity. Furthermore, NP(3S)s exhibited robust in vivo anticancer efficacy while minimizing the CisPt-induced liver toxicity. Overall, the results indicate NP(3S)s hold great promise for clinical translation due to their low toxicity profile and potent anticancer activity.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Simei Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiu Nie
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane 4006, Australia
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [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] [Received: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Duncan B, Al-Kassas R, Zhang G, Hughes D, Qiu Y. Ultrasound-Mediated Ocular Drug Delivery: From Physics and Instrumentation to Future Directions. MICROMACHINES 2023; 14:1575. [PMID: 37630111 PMCID: PMC10456754 DOI: 10.3390/mi14081575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023]
Abstract
Drug delivery to the anterior and posterior segments of the eye is impeded by anatomical and physiological barriers. Increasingly, the bioeffects produced by ultrasound are being proven effective for mitigating the impact of these barriers on ocular drug delivery, though there does not appear to be a consensus on the most appropriate system configuration and operating parameters for this application. In this review, the fundamental aspects of ultrasound physics most pertinent to drug delivery are presented; the primary phenomena responsible for increased drug delivery efficacy under ultrasound sonication are discussed; an overview of common ocular drug administration routes and the associated ocular barriers is also given before reviewing the current state of the art of ultrasound-mediated ocular drug delivery and its potential future directions.
Collapse
Affiliation(s)
- Blair Duncan
- School of Engineering, Faculty of Engineering & Technology, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| | - Raida Al-Kassas
- School of Pharmacy & Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| | - Guangming Zhang
- School of Engineering, Faculty of Engineering & Technology, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| | - Dave Hughes
- Novosound Ltd., Biocity, BoNess Road, Newhouse, Glasgow ML1 5UH, UK
| | - Yongqiang Qiu
- School of Engineering, Faculty of Engineering & Technology, Liverpool John Moores University, James Parsons Building, Byrom Street, Liverpool L3 3AF, UK
| |
Collapse
|
7
|
Muthukutty P, Yoo SY. Oncolytic Virus Engineering and Utilizations: Cancer Immunotherapy Perspective. Viruses 2023; 15:1645. [PMID: 37631987 PMCID: PMC10459766 DOI: 10.3390/v15081645] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Oncolytic viruses have positively impacted cancer immunotherapy over the past 20 years. Both natural and genetically modified viruses have shown promising results in treating various cancers. Various regulatory authorities worldwide have approved four commercial oncolytic viruses, and more are being developed to overcome this limitation and obtain better anti-tumor responses in clinical trials at various stages. Faster advancements in translating research into the commercialization of cancer immunotherapy and a comprehensive understanding of the modification strategies will widen the current knowledge of future technologies related to the development of oncolytic viruses. In this review, we discuss the strategies of virus engineering and the progress of clinical trials to achieve virotherapeutics.
Collapse
Affiliation(s)
| | - So Young Yoo
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
8
|
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
|
9
|
Cardoso RV, Pereira PR, Freitas CS, Paschoalin VMF. Trends in Drug Delivery Systems for Natural Bioactive Molecules to Treat Health Disorders: The Importance of Nano-Liposomes. Pharmaceutics 2022; 14:2808. [PMID: 36559301 PMCID: PMC9785269 DOI: 10.3390/pharmaceutics14122808] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Drug delivery systems are believed to increase pharmaceutical efficacy and the therapeutic index by protecting and stabilizing bioactive molecules, such as protein and peptides, against body fluids' enzymes and/or unsuitable physicochemical conditions while preserving the surrounding healthy tissues from toxicity. Liposomes are biocompatible and biodegradable and do not cause immunogenicity following intravenous or topical administration. Still, their most important characteristic is the ability to load any drug or complex molecule uncommitted to its hydrophobic or hydrophilic character. Selecting lipid components, ratios and thermo-sensitivity is critical to achieve a suitable nano-liposomal formulation. Nano-liposomal surfaces can be tailored to interact successfully with target cells, avoiding undesirable associations with plasma proteins and enhancing their half-life in the bloodstream. Macropinocytosis-dynamin-independent, cell-membrane-cholesterol-dependent processes, clathrin, and caveolae-independent mechanisms are involved in liposome internalization and trafficking within target cells to deliver the loaded drugs to modulate cell function. A successful translation from animal studies to clinical trials is still an important challenge surrounding the approval of new nano-liposomal drugs that have been the focus of investigations. Precision medicine based on the design of functionalized nano-delivery systems bearing highly specific molecules to drive therapies is a promising strategy to treat degenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Vania Margaret Flosi Paschoalin
- Programa de Pós-Graduação em Ciência de Alimentos e Programa de Pós-Graduação em Quimica, Instituto de Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos 149-sala 545-Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| |
Collapse
|
10
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Bahutair WN, Abuwatfa WH, Husseini GA. Ultrasound Triggering of Liposomal Nanodrugs for Cancer Therapy: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12173051. [PMID: 36080088 PMCID: PMC9458162 DOI: 10.3390/nano12173051] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/27/2022] [Accepted: 08/28/2022] [Indexed: 05/11/2023]
Abstract
Efficient conventional chemotherapy is limited by its nonspecific nature, which causes severe systemic toxicity that can lead to patient discomfort and low therapeutic efficacy. The emergence of smart drug delivery systems (SDDSs) utilizing nanoparticles as drug nanocarriers has shown great potential in enhancing the targetability of anticancer agents and limiting their side effects. Liposomes are among the most investigated nanoplatforms due to their promising capabilities of encapsulating hydrophilic, lipophilic, and amphiphilic drugs, biocompatibility, physicochemical and biophysical properties. Liposomal nanodrug systems have demonstrated the ability to alter drugs' biodistribution by sufficiently delivering the entrapped chemotherapeutics at the targeted diseased sites, sparing normal cells from undesired cytotoxic effects. Combining liposomal treatments with ultrasound, as an external drug release triggering modality, has been proven effective in spatially and temporally controlling and stimulating drug release. Therefore, this paper reviews recent literature pertaining to the therapeutic synergy of triggering nanodrugs from liposomes using ultrasound. It also highlights the effects of multiple physical and chemical factors on liposomes' sonosensetivity, several ultrasound-induced drug release mechanisms, and the efficacy of ultrasound-responsive liposomal systems in cancer therapy. Overall, liposomal nanodrug systems triggered by ultrasound are promising cancer therapy platforms that can potentially alleviate the detriments of conventional cancer treatments.
Collapse
Affiliation(s)
- Wafa N. Bahutair
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box. 26666, United Arab Emirates
- Correspondence:
| |
Collapse
|
12
|
Ji Q, Wu Y, Albers A, Fang M, Qian X. Strategies for Advanced Oncolytic Virotherapy: Current Technology Innovations and Clinical Approaches. Pharmaceutics 2022; 14:1811. [PMID: 36145559 PMCID: PMC9504140 DOI: 10.3390/pharmaceutics14091811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 12/22/2022] Open
Abstract
Oncolytic virotherapy is a type of nanomedicine with a dual antitumor mechanism. Viruses are engineered to selectively infect and lyse cancer cells directly, leading to the release of soluble antigens which induce systemic antitumor immunity. Representative drug Talimogene laherparepvec has showed promising therapeutic effects in advanced melanoma, especially when combined with immune checkpoint inhibitors with moderate adverse effects. Diverse viruses like herpes simplex virus, adenovirus, vaccina virus, and so on could be engineered as vectors to express different transgenic payloads, vastly expanding the therapeutic potential of oncolytic virotherapy. A number of related clinical trials are under way which are mainly focusing on solid tumors. Studies about further optimizing the genome of oncolytic viruses or improving the delivering system are in the hotspot, indicating the future development of oncolytic virotherapy in the clinic. This review introduces the latest progress in clinical trials and pre-clinical studies as well as technology innovations directed at oncolytic viruses. The challenges and perspectives of oncolytic virotherapy towards clinical application are also discussed.
Collapse
Affiliation(s)
- Qing Ji
- Department of Rare and Head & Neck Oncology, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yuchen Wu
- Department of Clinical Laboratory, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Andreas Albers
- Department of Otolaryngology, Head and Neck Surgery, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Meiyu Fang
- Department of Rare and Head & Neck Oncology, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Xu Qian
- Department of Clinical Laboratory, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
13
|
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: 30] [Impact Index Per Article: 10.0] [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
|
14
|
Aslam S, Akhtar A, Nirmal N, Khalid N, Maqsood S. Recent Developments in Starch-Based Delivery Systems of Bioactive Compounds: Formulations and Applications. FOOD ENGINEERING REVIEWS 2022. [DOI: 10.1007/s12393-022-09311-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
15
|
Investigation of fouling mechanism in membrane distillation using in-situ optical coherence tomography with green regeneration of fouled membrane. J Memb Sci 2022. [DOI: 10.1016/j.memsci.2021.119894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
16
|
Keller SB, Averkiou MA. The Role of Ultrasound in Modulating Interstitial Fluid Pressure in Solid Tumors for Improved Drug Delivery. Bioconjug Chem 2021; 33:1049-1056. [PMID: 34514776 DOI: 10.1021/acs.bioconjchem.1c00422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The unique microenvironment of solid tumors, including desmoplasia within the extracellular matrix, enhanced vascular permeability, and poor lymphatic drainage, leads to an elevated interstitial fluid pressure which is a major barrier to drug delivery. Reducing tumor interstitial fluid pressure is one proposed method of increasing drug delivery to the tumor. The goal of this topical review is to describe recent work using focused ultrasound with or without microbubbles to modulate tumor interstitial fluid pressure, through either thermal or mechanical effects on the extracellular matrix and the vasculature. Furthermore, we provide a review on techniques in which ultrasound imaging may be used to diagnose elevated interstitial fluid pressure within solid tumors. Ultrasound-based techniques show high promise in diagnosing and treating elevated interstitial pressure to enhance drug delivery.
Collapse
Affiliation(s)
- Sara B Keller
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Michalakis A Averkiou
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
17
|
Silvani G, Basirun C, Wu H, Mehner C, Poole K, Bradbury P, Chou J. A 3D‐Bioprinted Vascularized Glioblastoma‐on‐a‐Chip for Studying the Impact of Simulated Microgravity as a Novel Pre‐Clinical Approach in Brain Tumor Therapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Giulia Silvani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| | - Carin Basirun
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| | - Hanjie Wu
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| | - Christine Mehner
- Department of Physiology and Biomedical Engineering Mayo Clinic Jacksonville FL USA
| | - Kate Poole
- EMBL Australia node in Single Molecule Science, School of Medical Sciences, Faculty of Medicine University of New South Wales Sydney 2052 Australia
| | - Peta Bradbury
- Institut Curie, Paris Sciences et Lettres Research University Mechanics and Genetics of Embryonic and Tumoral Development Group Paris France
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology University of Technology Sydney Sydney Australia
| |
Collapse
|
18
|
Xu J, Salari A, Wang Y, He X, Kerr L, Darbandi A, de Leon AC, Exner AA, Kolios MC, Yuen D, Tsai SSH. Microfluidic Generation of Monodisperse Nanobubbles by Selective Gas Dissolution. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100345. [PMID: 33811441 DOI: 10.1002/smll.202100345] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/26/2021] [Indexed: 06/12/2023]
Abstract
Nanotechnology currently enables the fabrication of uniform solid nanoparticles and liquid nano-emulsions, but not uniform gaseous nanobubbles (NBs). In this article, for the first time, a method based on microfluidics that directly produces monodisperse NBs is reported. Specifically, a two-component gas mixture of water-soluble nitrogen and water-insoluble octafluoropropane as the gas phase are used in a microfluidic bubble generator. First, monodisperse microbubbles (MBs) with a classical microfluidic flow-focusing junction is generated, then the MBs shrink down to ≈100 nm diameter, due to the dissolution of the water-soluble components in the gas mixture. The degree of shrinkage is controlled by tuning the ratio of water-soluble to water-insoluble gas components. This technique maintains the monodispersity of the NBs, and enables precise control of the final NB size. It is found that the monodisperse NBs show better homogeneity than polydisperse NBs in in vitro ultrasound imaging experiments. Proof-of-concept in vivo kidney imaging is performed in live mice, demonstrating enhanced contrast using the monodisperse NBs. The NB monodispersity and imaging results make microfluidically generated NBs promising candidates as ultrasound contrast and molecular imaging agents.
Collapse
Affiliation(s)
- Jiang Xu
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Alinaghi Salari
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Biomedical Engineering Graduate Program, Ryerson University, Toronto, Ontario, M5B 2K2, Canada
| | - Yanjie Wang
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Department of Physics, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Xiaolin He
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Division of Nephrology, Department of Medicine, Unity Health Toronto and University of Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Liam Kerr
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Ali Darbandi
- Nanoimaging Centre, The Hospital for Sick Children, Peter Gilgan Centre for Research & Learning, Toronto, Ontario, M5G 0A4, Canada
| | - Al C de Leon
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Michael C Kolios
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Department of Physics, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
| | - Darren Yuen
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
- Division of Nephrology, Department of Medicine, Unity Health Toronto and University of Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Scott S H Tsai
- Institute for Biomedical Engineering, Science and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada
- Department of Mechanical and Industrial Engineering, Ryerson University, Toronto, Ontario, M5B 2K3, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| |
Collapse
|
19
|
Telichko AV, Lee T, Jakovljevic M, Dahl JJ. Passive Cavitation Mapping by Cavitation Source Localization From Aperture-Domain Signals-Part I: Theory and Validation Through Simulations. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:1184-1197. [PMID: 33141665 PMCID: PMC8486001 DOI: 10.1109/tuffc.2020.3035696] [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] [Indexed: 05/21/2023]
Abstract
Passive cavitation mapping (PCM) algorithms for diagnostic ultrasound arrays based on time exposure acoustics (TEA) exhibit poor axial resolution, which is in part due to the diffraction-limited point spread function of the imaging system and poor rejection by the delay-and-sum beamformer. In this article, we adapt a method for speed of sound estimation to be utilized as a cavitation source localization (CSL) approach. This method utilizes a hyperbolic fit to the arrival times of the cavitation signals in the aperture domain, and the coefficients of the fit are related to the position of the cavitation source. Wavefronts exhibiting poor fit to the hyperbolic function are corrected to yield improved source localization. We demonstrate through simulations that this method is capable of accurate estimation of the origin of coherent spherical waves radiating from cavitation/point sources. The average localization error from simulated microbubble sources was 0.12 ± 0.12mm ( 0.15 ± 0.14λ0 for a 1.78-MHz transmit frequency). In simulations of two simultaneous cavitation sources, the proposed technique had an average localization error of 0.2mm ( 0.23λ0 ), whereas conventional TEA had an average localization error of 0.81mm ( 0.97λ0 ). The reconstructed PCM-CSL image showed a significant improvement in resolution compared with the PCM-TEA approach.
Collapse
|
20
|
Telichko AV, Lee T, Hyun D, Chowdhury SM, Bachawal S, Herickhoff CD, Paulmurugan R, Dahl JJ. Passive Cavitation Mapping by Cavitation Source Localization From Aperture-Domain Signals-Part II: Phantom and In Vivo Experiments. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:1198-1212. [PMID: 33141666 PMCID: PMC8528486 DOI: 10.1109/tuffc.2020.3035709] [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: 05/04/2023]
Abstract
Passive cavitation mapping (PCM) techniques typically utilize a time-exposure acoustic (TEA) approach, where the received radio frequency data are beamformed, squared, and integrated over time. Such PCM-TEA cavitation maps typically suffer from long-tail artifacts and poor axial resolution with pulse-echo diagnostic arrays. Here, we utilize a recently developed PCM technique based on cavitation source localization (CSL), which fits a hyperbolic function to the received cavitation wavefront. A filtering method based on the root-mean-square error (rmse) of the hyperbolic fit is utilized to filter out spurious signals. We apply a wavefront correction technique to the signals with poor fit quality to recover additional cavitation signals and improve cavitation localization. Validation of the PCM-CSL technique with rmse filtering and wavefront correction was conducted in experiments with a tissue-mimicking flow phantom and an in vivo mouse model of cancer. It is shown that the quality of the hyperbolic fit, necessary for the PCM-CSL, requires an rmse < 0.05 mm2 in order to accurately localize the cavitation sources. A detailed study of the wavefront correction technique was carried out, and it was shown that, when applied to experiments with high noise and interference from multiple cavitating microbubbles, it was capable of effectively correcting noisy wavefronts without introducing spurious cavitation sources, thereby improving the quality of the PCM-CSL images. In phantom experiments, the PCM-CSL was capable of precisely localizing sources on the therapy beam axis and off-axis sources. In vivo cavitation experiments showed that PMC-CSL showed a significant improvement over PCM-TEA and yielded acceptable localization of cavitation signals in mice.
Collapse
|
21
|
Bharatula LD, Marsili E, Rice SA, Kwan JJ. Influence of High Intensity Focused Ultrasound on the Microstructure and c-di-GMP Signaling of Pseudomonas aeruginosa Biofilms. Front Microbiol 2020; 11:599407. [PMID: 33384674 PMCID: PMC7769819 DOI: 10.3389/fmicb.2020.599407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/23/2020] [Indexed: 01/13/2023] Open
Abstract
Bacterial biofilms are typically more tolerant to antimicrobials compared to bacteria in the planktonic phase and therefore require alternative treatment approaches. Mechanical biofilm disruption from ultrasound may be such an alternative by circumventing rapid biofilm adaptation to antimicrobial agents. Although ultrasound facilitates biofilm dispersal and may enhance the effectiveness of antimicrobial agents, the resulting biological response of bacteria within the biofilms remains poorly understood. To address this question, we investigated the microstructural effects of Pseudomonas aeruginosa biofilms exposed to high intensity focused ultrasound (HIFU) at different acoustic pressures and the subsequent biological response. Confocal microscopy images indicated a clear microstructural response at peak negative pressures equal to or greater than 3.5 MPa. In this pressure amplitude range, HIFU partially reduced the biomass of cells and eroded exopolysaccharides from the biofilm. These pressures also elicited a biological response; we observed an increase in a biomarker for biofilm development (cyclic-di-GMP) proportional to ultrasound induced biofilm removal. Cyclic-di-GMP overproducing mutant strains were also more resilient to disruption from HIFU at these pressures. The biological response was further evidenced by an increase in the relative abundance of cyclic-di-GMP overproducing variants present in the biofilm after exposure to HIFU. Our results, therefore, suggest that both physical and biological effects of ultrasound on bacterial biofilms must be considered in future studies.
Collapse
Affiliation(s)
- Lakshmi Deepika Bharatula
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Enrico Marsili
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- Department of Chemical and Materials Engineering, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Scott A. Rice
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - James J. Kwan
- Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Amin M, Huang W, Seynhaeve ALB, ten Hagen TLM. Hyperthermia and Temperature-Sensitive Nanomaterials for Spatiotemporal Drug Delivery to Solid Tumors. Pharmaceutics 2020; 12:E1007. [PMID: 33105816 PMCID: PMC7690578 DOI: 10.3390/pharmaceutics12111007] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nanotechnology has great capability in formulation, reduction of side effects, and enhancing pharmacokinetics of chemotherapeutics by designing stable or long circulating nano-carriers. However, effective drug delivery at the cellular level by means of such carriers is still unsatisfactory. One promising approach is using spatiotemporal drug release by means of nanoparticles with the capacity for content release triggered by internal or external stimuli. Among different stimuli, interests for application of external heat, hyperthermia, is growing. Advanced technology, ease of application and most importantly high level of control over applied heat, and as a result triggered release, and the adjuvant effect of hyperthermia in enhancing therapeutic response of chemotherapeutics, i.e., thermochemotherapy, make hyperthermia a great stimulus for triggered drug release. Therefore, a variety of temperature sensitive nano-carriers, lipid or/and polymeric based, have been fabricated and studied. Importantly, in order to achieve an efficient therapeutic outcome, and taking the advantages of thermochemotherapy into consideration, release characteristics from nano-carriers should fit with applicable clinical thermal setting. Here we introduce and discuss the application of the three most studied temperature sensitive nanoparticles with emphasis on release behavior and its importance regarding applicability and therapeutic potentials.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Wenqiu Huang
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
| | - Ann L. B. Seynhaeve
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
| | - Timo L. M. ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| |
Collapse
|
23
|
Sengupta S, Khatua C, Pal A, Bodhak S, Balla VK. Influence of Ultrasound and Magnetic Field Treatment Time on Carcinoma Cell Inhibition with Drug Carriers: An in Vitro Study. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:2752-2764. [PMID: 32654916 DOI: 10.1016/j.ultrasmedbio.2020.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/10/2020] [Accepted: 06/18/2020] [Indexed: 06/11/2023]
Abstract
The influence of exposing carcinoma cells to a static magnetic field (SMF) and low-intensity pulsed ultrasound (LIPUS), for different durations (15-45 min/d), in the presence of magnetic and non-magnetic drug carriers, on their in vitro inhibition is examined. Increasing the exposure time by 15 min/d decreased the culture duration by 24 h to achieve the same level of inhibition in colon (HCT116) and hepatocellular (HepG2) cells. Cell cycle analysis revealed enhanced cellular blockage in G1 and S phases with SMF + LIPUS exposure, and exposure for 45 min/d completely suppressed the S → G2 transition. Apoptosis of both types of cells increased with SMF + LIPUS treatment time, and HepG2 cells exhibited elevated necrosis with >30 min/d exposure. HepG2 cells also had higher amounts of reactive oxygen species (seven- to eightfold) than HCT116 cells (two- to sixfold), suggesting treatment effectiveness is cell and drug carrier dependent. The accelerated cellular activities are attributed to the enhanced internalization of drug carriers as a consequence of destabilized cellular membranes caused by the SMF + LIPUS-generated mechanical and electrical stimuli.
Collapse
Affiliation(s)
- Somoshree Sengupta
- Bioceramics & Coating Division, CSIR-Central Glass & Ceramic Research Institute, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Glass & Ceramic Research Institute Campus, Kolkata, India
| | - Chandra Khatua
- Bioceramics & Coating Division, CSIR-Central Glass & Ceramic Research Institute, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Glass & Ceramic Research Institute Campus, Kolkata, India
| | - Aniruddha Pal
- Bioceramics & Coating Division, CSIR-Central Glass & Ceramic Research Institute, Kolkata, India
| | - Subhadip Bodhak
- Bioceramics & Coating Division, CSIR-Central Glass & Ceramic Research Institute, Kolkata, India
| | - Vamsi Krishna Balla
- Bioceramics & Coating Division, CSIR-Central Glass & Ceramic Research Institute, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Central Glass & Ceramic Research Institute Campus, Kolkata, India.
| |
Collapse
|
24
|
Yang Y, Li Q, Guo X, Tu J, Zhang D. Mechanisms underlying sonoporation: Interaction between microbubbles and cells. ULTRASONICS SONOCHEMISTRY 2020; 67:105096. [PMID: 32278246 DOI: 10.1016/j.ultsonch.2020.105096] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 05/04/2023]
Abstract
The past several decades have witnessed great progress in "smart drug delivery", an advance technology that can deliver genes or drugs into specific locations of patients' body with enhanced delivery efficiency. Ultrasound-activated mechanical force induced by the interactions between microbubbles and cells, which can stimulate so-called "sonoporation" process, has been regarded as one of the most promising candidates to realize spatiotemporal-controllable drug delivery to selected regions. Both experimental and numerical studies were performed to get in-depth understanding on how the microbubbles interact with cells during sonoporation processes, under different impact parameters. The current work gives an overview of the general mechanism underlying microbubble-mediated sonoporation, and the possible impact factors (e.g., the properties of cavitation agents and cells, acoustical driving parameters and bubble/cell micro-environment) that could affect sonoporation outcomes. Finally, current progress and considerations of sonoporation in clinical applications are reviewed also.
Collapse
Affiliation(s)
- Yanye Yang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Qunying Li
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiasheng Guo
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China
| | - Juan Tu
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China.
| | - Dong Zhang
- Key Laboratory of Modern Acoustics (MOE), Department of Physics, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210093, China; The State Key Laboratory of Acoustics, Chinese Academy of Science, Beijing 10080, China
| |
Collapse
|
25
|
Seynhaeve A, Amin M, Haemmerich D, van Rhoon G, ten Hagen T. Hyperthermia and smart drug delivery systems for solid tumor therapy. Adv Drug Deliv Rev 2020; 163-164:125-144. [PMID: 32092379 DOI: 10.1016/j.addr.2020.02.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Chemotherapy is a cornerstone of cancer therapy. Irrespective of the administered drug, it is crucial that adequate drug amounts reach all cancer cells. To achieve this, drugs first need to be absorbed, then enter the blood circulation, diffuse into the tumor interstitial space and finally reach the tumor cells. Next to chemoresistance, one of the most important factors for effective chemotherapy is adequate tumor drug uptake and penetration. Unfortunately, most chemotherapeutic agents do not have favorable properties. These compounds are cleared rapidly, distribute throughout all tissues in the body, with only low tumor drug uptake that is heterogeneously distributed within the tumor. Moreover, the typical microenvironment of solid cancers provides additional hurdles for drug delivery, such as heterogeneous vascular density and perfusion, high interstitial fluid pressure, and abundant stroma. The hope was that nanotechnology will solve most, if not all, of these drug delivery barriers. However, in spite of advances and decades of nanoparticle development, results are unsatisfactory. One promising recent development are nanoparticles which can be steered, and release content triggered by internal or external signals. Here we discuss these so-called smart drug delivery systems in cancer therapy with emphasis on mild hyperthermia as a trigger signal for drug delivery.
Collapse
|
26
|
Tian Y, Liu Z, Tan H, Hou J, Wen X, Yang F, Cheng W. New Aspects of Ultrasound-Mediated Targeted Delivery and Therapy for Cancer. Int J Nanomedicine 2020; 15:401-418. [PMID: 32021187 PMCID: PMC6982438 DOI: 10.2147/ijn.s201208] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Ultrasound-mediated targeted delivery (UMTD), a novel delivery modality of therapeutic materials based on ultrasound, shows great potential in biomedical applications. By coupling ultrasound contrast agents with therapeutic materials, UMTD combines the advantages of ultrasound imaging and carrier, which benefit deep tissue penetration and high concentration aggregation. In this paper we introduced recent advances in ultrasound contrast agents and applications in tumor therapy. Ultrasound contrast agents were categorized by their functions, mainly including thermosensitive, pH-sensitive and photosensitive ultrasound contrast agents. The various applications of UMTD in tumor treatment were summarized as follows: drug therapy, transfection of anti-oncogene, RNA interference, vaccine immunotherapy, monoclonal antibody immunotherapy, adoptive cellular immunotherapy, cytokine immunotherapy, and so on. In the end, we elaborated on the current challenges and provided perspectives of UMTD for clinical applications.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Haoyan Tan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Jiahui Hou
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Xin Wen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Fan Yang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| |
Collapse
|
27
|
Drug-Loaded Microbubbles Combined with Ultrasound for Thrombolysis and Malignant Tumor Therapy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6792465. [PMID: 31662987 PMCID: PMC6791276 DOI: 10.1155/2019/6792465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/22/2019] [Accepted: 09/14/2019] [Indexed: 12/14/2022]
Abstract
Cardiac-cerebral thrombosis and malignant tumor endanger the safety of human life seriously. Traditional chemotherapy drugs have side effects which restrict their applications. Drug-loaded microbubbles can be destroyed by ultrasound irradiation at the focus position and be used for thrombolysis and tumor therapy. Compared with traditional drug treatment, the drug-loaded microbubbles can be excited by ultrasound and release drugs to lesion sites, increasing the local drug concentration and the exposure dose to nonfocal regions, thus reducing the cytotoxicity and side effects of drugs. This article reviews the applications of drug-loaded microbubbles combined with ultrasound for thrombolysis and tumor therapy. We focus on highlighting the advantages of using this new technique for disease treatment and concluding with recommendations for future efforts on the applications of this technology.
Collapse
|
28
|
Su X, Thomas RG, Bharatula LD, Kwan JJ. Remote targeted implantation of sound-sensitive biodegradable multi-cavity microparticles with focused ultrasound. Sci Rep 2019; 9:9612. [PMID: 31270380 PMCID: PMC6610131 DOI: 10.1038/s41598-019-46022-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/20/2019] [Indexed: 01/04/2023] Open
Abstract
Ultrasound-enhanced drug delivery has shown great promise in providing targeted burst release of drug at the site of the disease. Yet current solid ultrasound-responsive particles are non-degradable with limited potential for drug-loading. Here, we report on an ultrasound-responsive multi-cavity poly(lactic-co-glycolic acid) microparticle (mcPLGA MP) loaded with rhodamine B (RhB) with or without 4',6-diamidino-2-phenylindole (DAPI) to represent small molecule therapeutics. After exposure to high intensity focused ultrasound (HIFU), these delivery vehicles were remotely implanted into gel and porcine tissue models, where the particles rapidly released their payload within the first day and sustained release for at least seven days. RhB-mcPLGA MPs were implanted with HIFU into and beyond the sub-endothelial space of porcine arteries without observable damage to the artery. HIFU also guided the location of implantation; RhB-mcPLGA MPs were only observed at the focus of the HIFU away from the direction of ultrasound. Once implanted, DAPI co-loaded RhB-mcPLGA MPs released DAPI into the arterial wall, staining the nucleus of the cells. Our work shows the potential for HIFU-guided implantation of drug-loaded particles as a strategy to improve the local and sustained delivery of a therapeutic for up to two weeks.
Collapse
Affiliation(s)
- Xiaoqian Su
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Reju George Thomas
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Lakshmi Deepika Bharatula
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - James J Kwan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore.
| |
Collapse
|
29
|
Wallis J, Shenton DP, Carlisle RC. Novel approaches for the design, delivery and administration of vaccine technologies. Clin Exp Immunol 2019; 196:189-204. [PMID: 30963549 PMCID: PMC6468175 DOI: 10.1111/cei.13287] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
It is easy to argue that vaccine development represents humankind's most important and successful endeavour, such is the impact that vaccination has had on human morbidity and mortality over the last 200 years. During this time the original method of Jenner and Pasteur, i.e. that of injecting live-attenuated or inactivated pathogens, has been developed and supplemented with a wide range of alternative approaches which are now in clinical use or under development. These next-generation technologies have been designed to produce a vaccine that has the effectiveness of the original live-attenuated and inactivated vaccines, but without the associated risks and limitations. Indeed, the method of development has undoubtedly moved away from Pasteur's three Is paradigm (isolate, inactivate, inject) towards an approach of rational design, made possible by improved knowledge of the pathogen-host interaction and the mechanisms of the immune system. These novel vaccines have explored methods for targeted delivery of antigenic material, as well as for the control of release profiles, so that dosing regimens can be matched to the time-lines of immune system stimulation and the realities of health-care delivery in dispersed populations. The methods by which vaccines are administered are also the subject of intense research in the hope that needle and syringe dosing, with all its associated issues regarding risk of injury, cross-infection and patient compliance, can be replaced. This review provides a detailed overview of new vaccine vectors as well as information pertaining to the novel delivery platforms under development.
Collapse
Affiliation(s)
- J. Wallis
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| | - D. P. Shenton
- Defence Science and Technology LaboratoryPorton DownUK
| | - R. C. Carlisle
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| |
Collapse
|
30
|
Abstract
Since the second half of the 20th century, bioceramics are used for bone repair and regeneration. Inspired by bones and teeth, and aimed at mimicking their structure and composition, several artificial bioceramics were developed for biomedical applications. And nowadays, in the 21st century, with the increasing prominence of nanoscience and nanotechnology, certain bioceramics are being used to build smart drug delivery systems, among other applications. This minireview will mainly describe both tendencies through the research work carried out by the research team of María Vallet-Regí.
Collapse
Affiliation(s)
- María Vallet-Regí
- Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12,
Plaza Ramón y Cajal s/n, E-28040, Madrid, Spain; and Networking Research
Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid,
Spain
| |
Collapse
|
31
|
Alzaidi AI, Yahya A, Rava M, Swee TT, Idris N. A SYSTEMATIC REVIEW ON CURRENT RESEARCH TRENDS IN ELECTROSURGICAL SYSTEMS. BIOMEDICAL ENGINEERING: APPLICATIONS, BASIS AND COMMUNICATIONS 2019. [DOI: 10.4015/s1016237219500042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The use of electrosurgery (also referred to as radiosurgery) is a type of surgery that uses electrical currents in order to perform the surgery. This type of surgery dates back to more than 100 years. For over five decades, different reviews have been conducted in the field of electrosurgery. This has led to a dramatic increase in interest in electrosurgery, resulting to an incredible intervention in microvascular surgery that has provoked the most noteworthy use of radio frequency instruments. The controlled and exact use of these radio frequency electrical current on delicate tissue sites to be cut is accomplished by methods for deliberately composed anodes. This is a persistently advancing field with dynamic research going on in various new applications. There are different sorts of surgery, which can be delivered with high recurrence instruments such as Diathermy, Bipolar, and Monopolar. This paper performs a systematic review by extracting information from a wide variety of research papers in electrosurgery topics. The purpose of a systematic review is to classify and categorize the field of electrosurgery in an unbiased way, listing the current existing trends in electrosurgery, can lead to the classification and understanding of electrosurgery as a whole, and to predict the future direction of the field by analyzing the research interest over the last couple of years.
Collapse
Affiliation(s)
- Ali Idham Alzaidi
- Faculty of Biomedical Engineering, Universiti Teknologi Malaysia, 81310 UTM Skudai, Johor, Malaysia
| | - Azli Yahya
- Faculty of Electrical Engineering, Universiti Teknologi Malaysia 81310 UTM Skudai, Johor, Malaysia
| | - Mohammad Rava
- Faculty of Biomedical Engineering, Universiti Teknologi Malaysia, 81310 UTM Skudai, Johor, Malaysia
- Faculty of Computing, Universiti Teknologi Malaysia, 81310 UTM Skudai, Johor, Malaysia
| | - Tan Tian Swee
- Faculty of Biomedical Engineering, Universiti Teknologi Malaysia, 81310 UTM Skudai, Johor, Malaysia
| | - Norhalimah Idris
- Faculty of Management, University Teknologi Malaysia, 81310 Skudai Johor, Malaysia
| |
Collapse
|
32
|
Saccomandi P, Lapergola A, Longo F, Schena E, Quero G. Thermal ablation of pancreatic cancer: A systematic literature review of clinical practice and pre-clinical studies. Int J Hyperthermia 2018; 35:398-418. [PMID: 30428728 DOI: 10.1080/02656736.2018.1506165] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
PURPOSE Pancreatic cancer is a challenging malignancy with low treatment option and poor life expectancy. Thermal ablation techniques were proposed as alternative treatment options, especially in advanced stages and for unfit-for-surgery patients. This systematic review describes the thermal ablative techniques -i.e., Laser (LA), Radiofrequency (RFA), Microwave (MWA) Ablation, High-Intensity Focused Ultrasound (HIFU) and cryoablation- available for pancreatic cancer treatment. Additionally, an analysis of the efficacy, complication rate and overall survival for each technique is conducted. MATERIAL AND METHODS This review collects the ex vivo, preclinical and clinical studies presenting the use of thermal techniques in the pancreatic cancer treatment, searched up to March 2018 in PubMed and Medline. Abstracts, letters-to-the-editor, expert opinions, reviews and non-English language manuscripts were excluded. RESULTS Sixty-five papers were included. For the ex vivo and preclinical studies, there are: 12 records for LA, 8 for RFA, 0 for MWA, 6 for HIFU, 1 for cryoablation and 3 for hybrid techniques. For clinical studies, 1 paper for LA, 14 for RFA, 1 for MWA, 17 for HIFU, 1 for cryoablation and 1 for hybrid techniques. CONCLUSIONS Important technological advances are presented in ex vivo and preclinical studies, as the real-time thermometry, nanotechnology and hybrid techniques to enhance the thermal outcome. Conversely, a lack of standardization in the clinical employment of the procedures emerged, leading to contrasting results on the safety and feasibility of some analyzed techniques. Uniform conclusions on the safety and feasibility of these techniques for pancreatic cancer will require further structured investigation.
Collapse
Affiliation(s)
- Paola Saccomandi
- a IHU-Strasbourg Institute of Image-Guided Surgery , Strasbourg , France.,b Departement of Mechanical Engineering, Politecnico di Milano , Milan , Italy
| | - Alfonso Lapergola
- a IHU-Strasbourg Institute of Image-Guided Surgery , Strasbourg , France.,c Università G. D'Annunzio , Chieti , Italy
| | - Fabio Longo
- a IHU-Strasbourg Institute of Image-Guided Surgery , Strasbourg , France.,d Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome , Italy
| | | | - Giuseppe Quero
- d Fondazione Policlinico Universitario Agostino Gemelli IRCCS , Rome , Italy
| |
Collapse
|
33
|
Tian Y, Liu Z, Zhang L, Zhang J, Han X, Wang Q, Cheng W. Apatinib-loaded lipid nanobubbles combined with ultrasound-targeted nanobubble destruction for synergistic treatment of HepG2 cells in vitro. Onco Targets Ther 2018; 11:4785-4795. [PMID: 30127626 PMCID: PMC6091478 DOI: 10.2147/ott.s170786] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Apatinib, an oral small-molecule antiangiogenetic medicine, is used to treat patients with advanced hepatocellular carcinoma. However, its systemic toxic side effects cannot be ignored. The ultrasound (US)-targeted nanobubble destruction technology can minimize systemic drug exposure and maximize therapeutic efficacy. The aim of this study was to develop novel GPC3-targeted and drug-loaded nanobubbles (NBs) and further assess the associated therapeutic effects on hepatocellular carcinoma cells in vitro. Materials and methods Apatinib-loaded NBs were prepared by a mechanical vibration method. GPC3, a liver tumor homing peptide, was coated onto the surface of apatinib-loaded NBs through biotin–avidin interactions to target liver cancer HepG2 cells. The effects of different treatment groups on cell proliferation, cell cycle, and apoptosis of HepG2 cells were tested. Results The NBs could achieve 68% of optimal drug encapsulation. In addition, ligand binding assays demonstrated that attachment of targeted NBs to human HepG2 liver cancer cells was highly efficient. Furthermore, cell proliferation assays indicated that the antiproliferative activities of GPC3-targeted and apatinib-loaded NBs in combination with US (1 MHz, 1 W/cm2, 30 s) were, respectively, 44.11%±2.84%, 57.09%±6.38%, and 67.51%±2.89% after 24, 48, and 72 h of treatment. Treatment with GPC3-targeted and apatinib-loaded NBs also resulted in a higher proportion of cells in the G1 phase compared with other treatment groups such as apatinib only and nontargeted apatinib-loaded NBs when US was utilized. Conclusion US-targeted and drug-loaded nanobubble destruction successfully achieved selective growth inhibition and apoptosis in HepG2 cells in vitro. Therefore, GPC3-targeted and apatinib-loaded NBs can be considered a novel chemotherapeutic approach for treating liver cancer in combination with US.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150080, People's Republic of China,
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150080, People's Republic of China,
| | - Lei Zhang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150080, People's Republic of China,
| | - Jia Zhang
- Department of Microsystems and Microstructure Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150080, People's Republic of China
| | - Xue Han
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150080, People's Republic of China,
| | - Qiucheng Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150080, People's Republic of China,
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150080, People's Republic of China,
| |
Collapse
|
34
|
Kline-Schoder A, Le Z, Zderic V. Ultrasound-Enhanced Drug Delivery for Treatment of Onychomycosis. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2018; 37:1743-1752. [PMID: 29288596 DOI: 10.1002/jum.14526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 09/11/2017] [Accepted: 10/07/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES The aim of our study was to determine the effectiveness of using ultrasound (US) to increase the permeability of the nail, with the goal of improving outcomes in the treatment of onychomycosis. METHODS Porcine nails were used because of their similarity to human nails. A hydrophilic blue dye was used as a drug-mimicking compound. Two sets of experiments were performed: luminosity experiments to assess the dye levels inside the nail after US and sham treatments and diffusion cell experiments for determination of changes in nail permeability due to US application. In both sets of experiments, planar US transducers were used to sonicate the nails at frequencies of 400, 600, and 800 kHz and 1 MHz, an intensity of 1 W/cm2 , and a duration of 5 min in a continuous mode. Modeling studies were also performed to assess the safety of US application to the human toe for later clinical studies. RESULTS In the luminosity experiments, application of US at frequencies of 600 and 800 kHz led to statistically significant results (P < .05), with an increase in dye delivery into the nail of up to 95% compared to control values. The diffusion cell results found statistical significance (P < .05) at all applied frequencies, with up to a 70% increase in the nail permeability compared to the control. Safety modeling studies found a maximal temperature increase of 4.4 °C in the bone. CONCLUSIONS Our proposed US method may offer an alternative for improved treatment of onychomycosis. The current maximal temperature increase was found to be at the safety limit, and so pulsing and other alternatives will be investigated to minimize this temperature increase.
Collapse
Affiliation(s)
- Alina Kline-Schoder
- Departments of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Zung Le
- Podiatry, Medical Faculty Associates, George Washington University, Washington, DC, USA
| | - Vesna Zderic
- Departments of Biomedical Engineering, George Washington University, Washington, DC, USA
| |
Collapse
|
35
|
Sengupta S, Khatua C, Balla VK. In Vitro Carcinoma Treatment Using Magnetic Nanocarriers under Ultrasound and Magnetic Fields. ACS OMEGA 2018; 3:5459-5469. [PMID: 30023921 PMCID: PMC6044950 DOI: 10.1021/acsomega.8b00105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/07/2018] [Indexed: 06/01/2023]
Abstract
Nowadays, tumor hypoxia has become a more predominant problem for diagnosis as well as treatment of cancer due to difficulties in delivering chemotherapeutic drugs and their carriers to these regions with reduced vasculature and oxygen supply. In such cases, external physical stimulus-mediated drug delivery, such as ultrasound and magnetic fields, would be effective. In this work, the effect of simultaneous exposure of low-intensity pulsed ultrasound and static magnetic field on colon (HCT116) and hepatocellular (HepG2) carcinoma cell inhibition was assessed in vitro. The treatment, in the presence of anticancer drug, with and without magnetic carrier, significantly increased the reactive oxygen species production and hyperpolarized the cancer cells. As a result, a significant increase in cell inhibition, up to 86%, was observed compared to 50% inhibition with bare anticancer drug. The treatment appears to have relatively more effect on HepG2 cells during the initial 24 h than on HCT116 cells. The proposed treatment was also found to reduce cancer cell necrosis and did not show any inhibitory effect on healthy cells (MC3T3). Our in vitro results suggest that this approach has strong application potential to treat cancer at lower drug dosage to achieve similar inhibition and can reduce health risks associated with drugs.
Collapse
Affiliation(s)
- Somoshree Sengupta
- Bioceramics
& Coating Division, CSIR-Central
Glass & Ceramic Research Institute, 196 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Central
Glass & Ceramic Research Institute Campus, 196 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Chandra Khatua
- Bioceramics
& Coating Division, CSIR-Central
Glass & Ceramic Research Institute, 196 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Central
Glass & Ceramic Research Institute Campus, 196 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Vamsi K. Balla
- Bioceramics
& Coating Division, CSIR-Central
Glass & Ceramic Research Institute, 196 Raja S.C. Mullick Road, Kolkata 700032, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Central
Glass & Ceramic Research Institute Campus, 196 Raja S.C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
36
|
Sonodynamic therapy (SDT): a novel strategy for cancer nanotheranostics. SCIENCE CHINA-LIFE SCIENCES 2018; 61:415-426. [PMID: 29666990 DOI: 10.1007/s11427-017-9262-x] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Sonodynamic therapy (SDT) is a promising non-invasive therapeutic modality. Compared to photo-inspired therapy, SDT provides many opportunities and benefits, including deeper tissue penetration, high precision, less side effects, and good patient compliance. Thanks to the facile engineerable nature of nanotechnology, nanoparticles-based sonosensitizers exhibit predominant advantages, such as increased SDT efficacy, binding avidity, and targeting specificity. This review aims to summarize the possible mechanisms of SDT, which can be expected to provide the theoretical basis for SDT development in the future. We also extensively discuss nanoparticle-assisted sonosensitizers to enhance the outcome of SDT. Additionally, we focus on the potential strategy of combinational SDT with other therapeutic modalities and discuss the limitations and challenges of SDT toward clinical applications.
Collapse
|
37
|
Myers R, Grundy M, Rowe C, Coviello CM, Bau L, Erbs P, Foloppe J, Balloul JM, Story C, Coussios CC, Carlisle R. Ultrasound-mediated cavitation does not decrease the activity of small molecule, antibody or viral-based medicines. Int J Nanomedicine 2018; 13:337-349. [PMID: 29391793 PMCID: PMC5768183 DOI: 10.2147/ijn.s141557] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The treatment of cancer using nanomedicines is limited by the poor penetration of these potentially powerful agents into and throughout solid tumors. Externally controlled mechanical stimuli, such as the generation of cavitation-induced microstreaming using ultrasound (US), can provide a means of improving nanomedicine delivery. Notably, it has been demonstrated that by focusing, monitoring and controlling the US exposure, delivery can be achieved without damage to surrounding tissue or vasculature. However, there is a risk that such stimuli may disrupt the structure and thereby diminish the activity of the delivered drugs, especially complex antibody and viral-based nanomedicines. In this study, we characterize the impact of cavitation on four different agents, doxorubicin (Dox), cetuximab, adenovirus (Ad) and vaccinia virus (VV), representing a scale of sophistication from a simple small-molecule drug to complex biological agents. To achieve tight regulation of the level and duration of cavitation exposure, a “cavitation test rig” was designed and built. The activity of each agent was assessed with and without exposure to a defined cavitation regime which has previously been shown to provide effective and safe delivery of agents to tumors in preclinical studies. The fluorescence profile of Dox remained unchanged after exposure to cavitation, and the efficacy of this drug in killing a cancer cell line remained the same. Similarly, the ability of cetuximab to bind its epidermal growth factor receptor target was not diminished following exposure to cavitation. The encoding of the reporter gene luciferase within the Ad and VV constructs tested here allowed the infectivity of these viruses to be easily quantified. Exposure to cavitation did not impact on the activity of either virus. These data provide compelling evidence that the US parameters used to safely and successfully delivery nanomedicines to tumors in preclinical models do not detrimentally impact on the structure or activity of these nanomedicines.
Collapse
Affiliation(s)
| | - Megan Grundy
- BUBBL, IBME, Department of Engineering Science, University of Oxford, Oxford, UK
| | | | | | - Luca Bau
- BUBBL, IBME, Department of Engineering Science, University of Oxford, Oxford, UK
| | | | | | | | | | | | - Robert Carlisle
- BUBBL, IBME, Department of Engineering Science, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Browning RJ, Reardon PJT, Parhizkar M, Pedley RB, Edirisinghe M, Knowles JC, Stride E. Drug Delivery Strategies for Platinum-Based Chemotherapy. ACS NANO 2017; 11:8560-8578. [PMID: 28829568 DOI: 10.1021/acsnano.7b04092] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Few chemotherapeutics have had such an impact on cancer management as cis-diamminedichloridoplatinum(II) (CDDP), also known as cisplatin. The first member of the platinum-based drug family, CDDP's potent toxicity in disrupting DNA replication has led to its widespread use in multidrug therapies, with particular benefit in patients with testicular cancers. However, CDDP also produces significant side effects that limit the maximum systemic dose. Various strategies have been developed to address this challenge including encapsulation within micro- or nanocarriers and the use of external stimuli such as ultrasound to promote uptake and release. The aim of this review is to look at these strategies and recent scientific and clinical developments.
Collapse
Affiliation(s)
- Richard J Browning
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford , Oxford OX1 2JD, United Kingdom
| | | | | | | | | | - Jonathan C Knowles
- Department of Nanobiomedical Science and BK21 Plus NBM, Global Research Center for Regenerative Medicine, Dankook University , 518-10 Anseo-dong, Dongnam-gu, Cheonan, Chungcheongnam-do, Republic of Korea
- The Discoveries Centre for Regenerative and Precision Medicine, UCL Campus , Gower Street, London WC1E 6BT, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford , Oxford OX1 2JD, United Kingdom
| |
Collapse
|
39
|
Wu P, Jia Y, Qu F, Sun Y, Wang P, Zhang K, Xu C, Liu Q, Wang X. Ultrasound-Responsive Polymeric Micelles for Sonoporation-Assisted Site-Specific Therapeutic Action. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25706-25716. [PMID: 28741924 DOI: 10.1021/acsami.7b05469] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Targeting drug delivery remains a challenge in various disease treatment including cancer. The local drug deposit could be greatly enhanced by some external stimuli-responsive systems. Here we develop pluronic P123/F127 polymeric micelles (M) encapsulating curcumin (Cur) that are permeabilized directly by focused ultrasound, in which ultrasound triggers drug release. Tumor preferential accumulation and site-specific sonochemotherapy were then evaluated. Cur-loaded P123/F127 mixed micelles (Cur-M) exhibited longer circulating time and increased cellular uptake compared to free Cur. With the assistance of focused ultrasound treatment, Cur-M showed tumor-targeting deposition in a time-dependent manner following systemic administration. This was due to enhanced permeabilization of tumor regions and increased penetration of Cur-M in irradiated tumor cells by ultrasound sonoporation. Furthermore, Cur-M self-assembly could be regulated by ultrasound irradiation. In vitro Cur release from mixed micelles was greatly dependent on ultrasound intensity but not on duration, suggesting the cavitational threshold was necessary to initiate subsequent sonochemotherapy. In vivo site-specific drug release was demonstrated in dual-tumor models, which showed spatial-temporal release of entrapped drugs following intratumoral injection. The sonoporation-assisted site-specific chemotherapy significantly inhibited tumor growth and the decrease in tumor weight was approximately 6.5-fold more than without exposure to ultrasound irradiation. In conclusion, the established ultrasound-guided nanomedicine targeting deposit and local release may represent a new strategy to improve chemotherapy efficiency.
Collapse
Affiliation(s)
- Pengying Wu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Yali Jia
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Fei Qu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Yue Sun
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Pan Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Kun Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Chuanshan Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong , Shatin, Hong Kong 999077, China
| | - Quanhong Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| | - Xiaobing Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University , Xi'an, Shaanxi 710119, China
| |
Collapse
|
40
|
Apoptotic and genotoxic effects of low-intensity ultrasound on healthy and leukemic human peripheral mononuclear blood cells. J Med Ultrason (2001) 2017; 45:31-39. [DOI: 10.1007/s10396-017-0805-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/05/2017] [Indexed: 12/31/2022]
|
41
|
Bian S, Seth A, Daly D, Carlisle R, Stride E. A multimodal instrument for real-time in situ study of ultrasound and cavitation mediated drug delivery. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2017; 88:034302. [PMID: 28372398 DOI: 10.1063/1.4978811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The development of a multimodal instrument capable of real-time in situ measurements of cavitation activity and effect in tissue mimicking phantoms during ultrasound and cavitation mediated drug delivery experiments is described here. The instrument features an acoustic arm that can expose phantoms to high-intensity focused-ultrasound while measuring cavitation activity and an optical arm that monitors cavitation effect using confocal microscopy. This combination of modalities allows real-time in situ characterisation of drug delivery in tissue and tissue mimicking phantoms during ultrasound and cavitation mediated drug delivery experiments. A representative result, obtained with a tissue mimicking phantom and acoustically activated droplets, is presented here as a demonstration of the instrument's capabilities and potential applications.
Collapse
Affiliation(s)
- Shuning Bian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Anjali Seth
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Dan Daly
- Lein Applied Diagnostics, Reading, United Kingdom
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
42
|
Aw MS, Paniwnyk L. Overcoming T. gondii infection and intracellular protein nanocapsules as biomaterials for ultrasonically controlled drug release. Biomater Sci 2017; 5:1944-1961. [DOI: 10.1039/c7bm00425g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
One of the pivotal matters of concern in intracellular drug delivery is the preparation of biomaterials containing drugs that are compatible with the host target.
Collapse
Affiliation(s)
- M. S. Aw
- School of Life Sciences
- Biomolecular and Sports Science
- Faculty of Health and Life Sciences
- Coventry University
- Coventry
| | - L. Paniwnyk
- School of Life Sciences
- Biomolecular and Sports Science
- Faculty of Health and Life Sciences
- Coventry University
- Coventry
| |
Collapse
|
43
|
Bayir E, Bilgi E, Urkmez AS. Implementation of Nanoparticles in Cancer Therapy. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Cancer is a wide group of diseases and generally characterized by uncontrolled proliferation of cells whose metabolic activities are disrupted. Conventionally, chemotherapy, radiotherapy, and surgery are used in the treatment of cancer. However, in theory, even a single cancer cell may trigger recurrence. Therefore, these treatments cannot provide high survival rate for deadly types. Identification of alternative methods in treatment of cancers is inevitable because of adverse effects of conventional methods. In the last few decades, nanotechnology developed by scientists working in different disciplines—physics, chemistry, and biology—offers great opportunities. It is providing elimination of both circulating tumor cells and solid cancer cells by targeting cancer cells. In this chapter, inadequate parts of conventional treatment methods, nanoparticle types used in new treatment methods of cancer, and targeting methods of nanoparticles are summarized; furthermore, recommendations of future are provided.
Collapse
|
44
|
Lea-Banks H, Teo B, Stride E, Coussios CC. The effect of particle density on ultrasound-mediated transport of nanoparticles. Phys Med Biol 2016; 61:7906-7918. [DOI: 10.1088/0031-9155/61/22/7906] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
45
|
Nwokeoha S, Carlisle R, Cleveland RO. The Application of Clinical Lithotripter Shock Waves to RNA Nucleotide Delivery to Cells. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2478-2492. [PMID: 27444864 DOI: 10.1016/j.ultrasmedbio.2016.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/15/2016] [Accepted: 06/02/2016] [Indexed: 06/06/2023]
Abstract
The delivery of genes into cells through the transfer of ribonucleic acids (RNAs) has been found to cause a change in the level of target protein expression. RNA-based transfection is conceptually more efficient than commonly delivered plasmid DNA because it does not require division or damage of the nuclear envelope, thereby increasing the chances of the cell remaining viable. Shock waves (SWs) have been found to induce cellular uptake by transiently altering the permeability of the plasma membrane, thereby overcoming a critical step in gene therapy. However, accompanying SW bio-effects include dose-dependent irreversible cell injury and cytotoxicity. Here, the effect of SWs generated by a clinical lithotripter on the viability and permeabilisation of three different cell lines in vitro was investigated. Comparison of RNA stability before and after SW exposure revealed no statistically significant difference. Optimal SW exposure parameters were identified to minimise cell death and maximise permeabilisation, and applied to enhanced green fluorescent protein (eGFP) messenger RNA (mRNA) or anti-eGFP small interfering RNA delivery. As a result, eGFP mRNA expression levels increased up to 52-fold in CT26 cells, whereas a 2-fold decrease in GFP expression was achieved after anti-eGFP small interfering RNA delivery to MCF-7/GFP cells. These results indicate that SW parameters can be employed to achieve effective nucleotide delivery, laying the foundation for non-invasive and high-tolerability RNA-based gene therapy.
Collapse
Affiliation(s)
- Sandra Nwokeoha
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom.
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Robin O Cleveland
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
46
|
Qian X, Zheng Y, Chen Y. Micro/Nanoparticle-Augmented Sonodynamic Therapy (SDT): Breaking the Depth Shallow of Photoactivation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:8097-8129. [PMID: 27384408 DOI: 10.1002/adma.201602012] [Citation(s) in RCA: 537] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/28/2016] [Indexed: 05/08/2023]
Abstract
The fast development of photoactivation for cancer treatment provides an efficient photo-therapeutic strategy for cancer treatment, but traditional photodynamic or photothermal therapy suffers from the critical issue of low in vivo penetration depth of tissues. As a non-invasive therapeutic modality, sonodynamic therapy (SDT) can break the depth barrier of photoactivation because ultrasound has an intrinsically high tissue-penetration performance. Micro/nanoparticles can efficiently augment the SDT efficiency based on nanobiotechnology. The state-of-art of the representative achievements on micro/nanoparticle-enhanced SDT is summarized, and specific functions of micro/nanoparticles for SDT are discussed, from the different viewpoints of ultrasound medicine, material science and nanobiotechnology. Emphasis is put on the relationship of structure/composition-SDT performance of micro/nanoparticle-based sonosensitizers. Three types of micro/nanoparticle-augmented SDT are discussed, including organic and inorganic sonosensitizers and micro/nanoparticle-based but sonosensitizer-free strategies to enhance the SDT outcome. SDT-based synergistic cancer therapy augmented by micro/nanoparticles and their biosafety are also included. Some urgent critical issues and potential developments of micro/nanoparticle-augmented SDT for efficient cancer treatment are addressed. It is highly expected that micro/nanoparticle-augmented SDT will be quickly developed as a new and efficient therapeutic modality which will find practical applications in cancer treatment. At the same time, fundamental disciplines regarding materials science, chemistry, medicine and nanotechnology will be advanced.
Collapse
Affiliation(s)
- Xiaoqin Qian
- Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Yuanyi Zheng
- Sixth Affiliated Hospital of Shanghai Jiaotong University & Shanghai Institute of Ultrasound in Medicine, Shanghai, 200233, P. R. China.
| | - Yu Chen
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China.
| |
Collapse
|
47
|
Exploitation of sub-micron cavitation nuclei to enhance ultrasound-mediated transdermal transport and penetration of vaccines. J Control Release 2016; 238:22-30. [DOI: 10.1016/j.jconrel.2016.07.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/13/2016] [Accepted: 07/10/2016] [Indexed: 01/18/2023]
|
48
|
Polymeric Cups for Cavitation-mediated Delivery of Oncolytic Vaccinia Virus. Mol Ther 2016; 24:1627-33. [PMID: 27375160 PMCID: PMC5113106 DOI: 10.1038/mt.2016.139] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/21/2016] [Indexed: 12/15/2022] Open
Abstract
Oncolytic viruses (OV) could become the most powerful and selective cancer therapies. However, the limited transport of OV into and throughout tumors following intravenous injection means their clinical administration is often restricted to direct intratumoral dosing. Application of physical stimuli, such as focused ultrasound, offers a means of achieving enhanced mass transport. In particular, shockwaves and microstreaming resulting from the instigation of an ultrasound-induced event known as inertial cavitation can propel OV hundreds of microns. We have recently developed a polymeric cup formulation which, when delivered intravenously, provides the nuclei for instigation of sustained inertial cavitation events within tumors. Here we report that exposure of tumors to focused ultrasound after intravenous coinjection of cups and oncolytic vaccinia virus , leads to substantial and significant increases in activity. When cavitation was instigated within SKOV-3 or HepG2 xenografts, reporter gene expression from vaccinia virus was enhanced 1,000-fold (P < 0.0001) or 10,000-fold (P < 0.001), respectively. Similar increases in the number of vaccinia virus genomes recovered from tumors were also observed. In survival studies, the application of cup mediated cavitation to a vaccinia virus expressing a prodrug converting enzyme provided significant (P < 0.05) retardation of tumor growth. This technology could improve the clinical utility of all biological therapeutics including OV.
Collapse
|
49
|
Lyka E, Coviello C, Kozick R, Coussios CC. Sum-of-harmonics method for improved narrowband and broadband signal quantification during passive monitoring of ultrasound therapies. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2016; 140:741. [PMID: 27475195 DOI: 10.1121/1.4958991] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Passive Acoustic Mapping (PAM) enables real-time monitoring of ultrasound therapies by beamforming acoustic emissions emanating from the ultrasound focus. Reconstruction of the narrowband or broadband acoustic emissions component enables mapping of different physical phenomena, with narrowband emissions arising from non-linear propagation and scattering, non-inertial cavitation or tissue boiling, and broadband (generally, of significantly lower amplitude) indicating inertial cavitation. Currently, accurate classification of the received signals based on pre-defined frequency-domain comb filters cannot be guaranteed because varying levels of leakage occur as a function of signal amplitude and the choice of windowing function. This work presents a time-domain parametric model aimed at enabling accurate estimation of the amplitude of time-varying narrowband components in the presence of broadband signals. Conversely, the method makes it possible to recover a weak broadband signal in the presence of a dominant harmonic or other narrowband component. Compared to conventional comb filtering, the proposed sum-of-harmonics method enables PAM of cavitation sources that better reflect their physical location and extent.
Collapse
Affiliation(s)
- Erasmia Lyka
- Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Christian Coviello
- Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Richard Kozick
- Department of Electrical and Computer Engineering, Bucknell University, Lewisburg, Pennsylvania 17837, USA
| | - Constantin-C Coussios
- Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
50
|
Prieur F, Pillon A, Mestas JL, Cartron V, Cèbe P, Chansard N, Lafond M, Lafon C. Enhancement of Fluorescent Probe Penetration into Tumors In Vivo Using Unseeded Inertial Cavitation. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1706-1713. [PMID: 27087691 DOI: 10.1016/j.ultrasmedbio.2016.01.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 01/18/2016] [Accepted: 01/27/2016] [Indexed: 06/05/2023]
Abstract
Ultrasound-induced cavitation has found many applications in the field of cancer therapy. One of its beneficial effects is the enhancement of drug intake by tumor cells. Our group has developed a device that can create and control unseeded cavitation in tissue using ultrasound. We conducted experiments on tumor-bearing mice using our device to assess the impact of sonication on the penetration of fluorescent probes into tumor cells. We studied the influence of pressure level, timing of sonication and sonication duration on treatment efficiency. Our results indicate that fluorescent probes penetrate better into tumors exposed to ultrasound. The best results revealed an increase in penetration of 61% and were obtained when sonicating the tumor in presence of the probes with a peak negative pressure at focus of 19 MPa. At this pressure level, the treatment generated only minor skin damage. Treatments could be significantly accelerated as equivalent enhanced penetration of probes was achieved when multiplying the initial raster scan speed by a factor of four.
Collapse
Affiliation(s)
- Fabrice Prieur
- Inserm, U1032, LabTau, Lyon, France; Université de Lyon, Lyon, France.
| | - Arnaud Pillon
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Jean-Louis Mestas
- Inserm, U1032, LabTau, Lyon, France; Université de Lyon, Lyon, France; Caviskills SAS, Vaulx-en-Velin, France
| | - Valérie Cartron
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Patrick Cèbe
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Nathalie Chansard
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Maxime Lafond
- Inserm, U1032, LabTau, Lyon, France; Université de Lyon, Lyon, France
| | - Cyril Lafon
- Inserm, U1032, LabTau, Lyon, France; Université de Lyon, Lyon, France; Caviskills SAS, Vaulx-en-Velin, France
| |
Collapse
|