1
|
Italiya KS, Mullins-Dansereau V, Geoffroy K, Gilchrist VH, Alain T, Bourgeois-Daigneault MC, Yu F. Ultrasound and Microbubble Mediated Delivery of Virus-Sensitizing Drugs Improves In Vitro Oncolytic Virotherapy Against Breast Cancer Cells. ULTRASOUND IN MEDICINE & BIOLOGY 2025:S0301-5629(25)00089-4. [PMID: 40234152 DOI: 10.1016/j.ultrasmedbio.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/17/2025]
Abstract
OBJECTIVE Oncolytic virotherapy is an emerging form of cancer treatment that uses replication-competent viruses to kill cancer cells. However, as for other cancer therapies, oncolytic viruses are not effective against all cancers and there is a need to further improve treatment efficacy while maintaining low toxicity. Viral sensitizers are drugs that potentiate viral replication in tumor cells. While various studies have shown their synergy with oncolytic virotherapy, the risks associated with systemic toxicities that vary according to the drug used limit the clinical translation of the approach. In this study, we used an ultrasound and image-guided approach in which we loaded viral-sensitizing drugs onto microbubbles which are then cavitated by ultrasound to deliver the encapsulated drugs to tumor cells, which improves in vitro oncolytic virotherapy efficacy in the 4T1 breast cancer model. METHODS In this study, we loaded two viral sensitizers, paclitaxel and volasertib, onto lipid microbubbles and comprehensively characterized their effect on the infection of 4T1 murine mammary carcinoma cells by oncolytic Vesicular stomatitis virus in vitro. RESULTS We synthesized lipid microbubbles with high and moderate encapsulation efficiency for paclitaxel (83.7%) and volasertib (28.6%), respectively. Stability assessments demonstrated excellent retention in various conditions, highlighting their potential for in vivo use. In vitro studies confirmed their acoustic responsiveness essential for controlled drug release at targeted sites. Paclitaxel and volasertib release from viral sensitizer-loaded microbubbles following ultrasound-triggered cavitation significantly increased viral replication (57-fold, p < 0.0001 and 27-fold, p < 0.01, respectively), as well as tumor cell killing compared to virus-infected untreated cells. CONCLUSION Altogether, our data show that drug-loaded microbubble cavitation and free drugs both sensitize cancer cells to oncolytic viruses to equivalent levels. These findings provide a proof of concept for the use of ultrasound-guided microbubble drug delivery in combination with oncolytic virotherapy.
Collapse
Affiliation(s)
- Kishan Shamjibhai Italiya
- Imaging and Engineering axis, CRCHUM, Montreal, Quebec, Canada; Cancer axis and Institut du cancer de Montréal, CRCHUM, Montreal, Quebec, Canada
| | - Victor Mullins-Dansereau
- Cancer axis and Institut du cancer de Montréal, CRCHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Immunopathology axis, CRCHUM, Montreal, Quebec, Canada
| | - Karen Geoffroy
- Cancer axis and Institut du cancer de Montréal, CRCHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Immunopathology axis, CRCHUM, Montreal, Quebec, Canada
| | - Victoria Heather Gilchrist
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada; Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Tommy Alain
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada; Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Cancer axis and Institut du cancer de Montréal, CRCHUM, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Immunopathology axis, CRCHUM, Montreal, Quebec, Canada
| | - François Yu
- Imaging and Engineering axis, CRCHUM, Montreal, Quebec, Canada; Cancer axis and Institut du cancer de Montréal, CRCHUM, Montreal, Quebec, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Jayasankar G, Koilpillai J, Narayanasamy D. A Systematic Study on Long-acting Nanobubbles: Current Advancement and Prospects on Theranostic Properties. Adv Pharm Bull 2024; 14:278-301. [PMID: 39206408 PMCID: PMC11347731 DOI: 10.34172/apb.2024.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 09/04/2024] Open
Abstract
Delivery of diagnostic drugs via nanobubbles (NBs) has shown to be an emerging field of study. Due to their small size, NBs may more easily travel through constricted blood vessels and precisely target certain bodily parts. NB is considered the major treatment for cancer treatment and other diseases which are difficult to diagnose. The field of NBs is dynamic and continues to grow as researchers discover new properties and seek practical applications in various fields. The predominant usage of NBs in novel drug delivery is to enhance the bioavailability, and controlled drug release along with imaging properties NBs are important because they may change interfacial characteristics including surface force, lubrication, and absorption. The quick diffusion of gas into the water was caused by a hypothetical film that was stimulated and punctured by a strong acting force at the gas/water contact of the bubble. In this article, various prominent aspects of NBs have been discussed, along with the long-acting nature, and the theranostical aspect which elucidates the potential marketed drugs along with clinical trial products. The article also covers quality by design aspects, different production techniques that enable method-specific therapeutic applications, increasing the floating time of the bubble, and refining its properties to enhance the prepared NB's quality. NB containing both analysis and curing properties makes it special from other nano-carriers. This work includes all the possible methods of preparing NB, its application, all marketed drugs, and products in clinical trials.
Collapse
Affiliation(s)
| | | | - Damodharan Narayanasamy
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institution of Science and Technology, Kattankulathur, Chengalpattu, India
| |
Collapse
|
3
|
Han J, Dong H, Zhu T, Wei Q, Wang Y, Wang Y, Lv Y, Mu H, Huang S, Zeng K, Xu J, Ding J. Biochemical hallmarks-targeting antineoplastic nanotherapeutics. Bioact Mater 2024; 36:427-454. [PMID: 39044728 PMCID: PMC11263727 DOI: 10.1016/j.bioactmat.2024.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 07/25/2024] Open
Abstract
Tumor microenvironments (TMEs) have received increasing attention in recent years as they play pivotal roles in tumorigenesis, progression, metastases, and resistance to the traditional modalities of cancer therapy like chemotherapy. With the rapid development of nanotechnology, effective antineoplastic nanotherapeutics targeting the aberrant hallmarks of TMEs have been proposed. The appropriate design and fabrication endow nanomedicines with the abilities for active targeting, TMEs-responsiveness, and optimization of physicochemical properties of tumors, thereby overcoming transport barriers and significantly improving antineoplastic therapeutic benefits. This review begins with the origins and characteristics of TMEs and discusses the latest strategies for modulating the TMEs by focusing on the regulation of biochemical microenvironments, such as tumor acidosis, hypoxia, and dysregulated metabolism. Finally, this review summarizes the challenges in the development of smart anti-cancer nanotherapeutics for TME modulation and examines the promising strategies for combination therapies with traditional treatments for further clinical translation.
Collapse
Affiliation(s)
- Jing Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - He Dong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Tianyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Yongheng Wang
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Yun Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Yu Lv
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Shandeng Huang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Ke Zeng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
4
|
Huang J, Zhang L, Zheng J, Lin Y, Leng X, Wang C, Li P, Feng L. Microbubbles-assisted ultrasonication to promote tumor accumulation of therapeutics and modulation of tumor microenvironment for enhanced cancer treatments. Biomaterials 2023; 299:122181. [PMID: 37276797 DOI: 10.1016/j.biomaterials.2023.122181] [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: 12/28/2022] [Revised: 05/10/2023] [Accepted: 05/26/2023] [Indexed: 06/07/2023]
Abstract
Abnormal tumor vasculature is reported to severely hinder the therapeutic potency of diverse cancer therapeutics by restricting their intratumoral accumulation and/or causing therapeutic resistance. Herein, a microbubble-assisted ultrasonication technology (MAUT) of systemic administration of octafluoropropane-filled microbubbles together with tumor localized ultrasound (US) exposure is developed to generally promote intratumoral accumulation efficacy of three kinds of anti-tumor drugs with varying sizes through the cavitation effect-induced disruption of tumor blood vessels. MAUT was further shown to enable selective tumor hypoxia attenuation by filling microbubbles with high-purity oxygen and thus reducing the production of immunosuppressive lactic acids by suppressing glycolysis in cancer cells. Resultantly, MAUT markedly enhanced the therapeutic outcome of systemically administered anti-programmed death-1 (anti-PD-1) and chemotherapeutic doxorubicin (DOX) with and without using nanoscale liposomes as delivery vehicles. This work highlights that MAUT is a biocompatible yet versatile strategy to effectively reinforce the therapeutic potency of a broad range of cancer therapeutics, promising for future clinical usage.
Collapse
Affiliation(s)
- Ju Huang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, PR China
| | - Jun Zheng
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yi Lin
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, PR China
| | - Xiaojing Leng
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Chunjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren' Ai Road, Suzhou, Jiangsu, 215123, PR China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren' Ai Road, Suzhou, Jiangsu, 215123, PR China.
| |
Collapse
|
5
|
Kancheva M, Aronson L, Pattilachan T, Sautto F, Daines B, Thommes D, Shar A, Razavi M. Bubble-Based Drug Delivery Systems: Next-Generation Diagnosis to Therapy. J Funct Biomater 2023; 14:373. [PMID: 37504868 PMCID: PMC10382061 DOI: 10.3390/jfb14070373] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
Current radiologic and medication administration is systematic and has widespread side effects; however, the administration of microbubbles and nanobubbles (MNBs) has the possibility to provide therapeutic and diagnostic information without the same ramifications. Microbubbles (MBs), for instance, have been used for ultrasound (US) imaging due to their ability to remain in vessels when exposed to ultrasonic waves. On the other hand, nanobubbles (NBs) can be used for further therapeutic benefits, including chronic treatments for osteoporosis and cancer, gene delivery, and treatment for acute conditions, such as brain infections and urinary tract infections (UTIs). Clinical trials are also being conducted for different administrations and utilizations of MNBs. Overall, there are large horizons for the benefits of MNBs in radiology, general medicine, surgery, and many more medical applications. As such, this review aims to evaluate the most recent publications from 2016 to 2022 to report the current uses and innovations for MNBs.
Collapse
Affiliation(s)
- Mihaela Kancheva
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Lauren Aronson
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Tara Pattilachan
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Francesco Sautto
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Benjamin Daines
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Donald Thommes
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Angela Shar
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
6
|
Wang H, Guo Y, Hu Y, Zhou Y, Chen Y, Huang X, Chen J, Deng Q, Cao S, Hu B, Jiang R, Pan J, Tan T, Wang Y, Chen Y, Dong Q, Chen P, Zhou Q. Ultrasound-controlled nano oxygen carriers enhancing cell viability in 3D GelMA hydrogel for the treatment of myocardial infarction. Int J Biol Macromol 2023:125139. [PMID: 37268076 DOI: 10.1016/j.ijbiomac.2023.125139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
Heart failure is a critical and ultimate phase of cardiovascular ailment that leads to a considerable incidence of disability and mortality. Among various factors contributing to heart failure, myocardial infarction is one of the most frequent and significant causes, which is still difficult to manage effectively. An innovative therapeutic strategy, namely a 3D bio-printed cardiac patch, has recently emerged as a promising approach to substitute damaged cardiomyocytes in a localized infarct region. Nevertheless, the efficacy of this treatment primarily relies on the long-term viability of the transplanted cells. In this study, we aimed to construct acoustically sensitive nano oxygen carriers to improve cell survival inside the bio-3D printed patch. In this study, we initially created nanodroplets capable of phase transition triggered by ultrasound and integrated them into GelMA (Gelatin Methacryloyl) hydrogels, which were then employed for 3D bioprinting. After adding nanodroplets and ultrasonic irradiation, numerous pores appeared inside the hydrogel with improved permeability. We further encapsulated hemoglobin into nanodroplets (ND-Hb) to construct oxygen carriers. Results of in vitro experiments showed the highest cell survival within the patch of ND-Hb irradiated by the low-intensity pulsed ultrasound (LIPUS) group. The genomic analysis discovered that the increased survival of seeded cells within the patch might be related to the protection of mitochondrial function owing to the improved hypoxic state. Eventually, in vivo studies revealed that the LIPUS+ND-Hb group had improved cardiac function and increased revascularization after myocardial infarction. To summarize, our study successfully improved the permeability of the hydrogel in a non-invasive and efficient manner, facilitating the exchange of substances in the cardiac patch. Moreover, ultrasound-controlled oxygen release augmented the viability of the transplanted cells and expedited the repair of infarcted tissues.
Collapse
Affiliation(s)
- Hao Wang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yuxin Guo
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yugang Hu
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yanxiang Zhou
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yueying Chen
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xin Huang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jinling Chen
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Qing Deng
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Sheng Cao
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Bo Hu
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Riyue Jiang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Juhong Pan
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Tuantuan Tan
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yijia Wang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yun Chen
- Wuhan University School of Basic Medical Science, 430060 Wuhan, China
| | - Qi Dong
- Wuhan University School of Basic Medical Science, 430060 Wuhan, China
| | - Pu Chen
- Wuhan University School of Basic Medical Science, 430060 Wuhan, China
| | - Qing Zhou
- Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
7
|
Navarro-Becerra JA, Borden MA. Targeted Microbubbles for Drug, Gene, and Cell Delivery in Therapy and Immunotherapy. Pharmaceutics 2023; 15:1625. [PMID: 37376072 DOI: 10.3390/pharmaceutics15061625] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Microbubbles are 1-10 μm diameter gas-filled acoustically-active particles, typically stabilized by a phospholipid monolayer shell. Microbubbles can be engineered through bioconjugation of a ligand, drug and/or cell. Since their inception a few decades ago, several targeted microbubble (tMB) formulations have been developed as ultrasound imaging probes and ultrasound-responsive carriers to promote the local delivery and uptake of a wide variety of drugs, genes, and cells in different therapeutic applications. The aim of this review is to summarize the state-of-the-art of current tMB formulations and their ultrasound-targeted delivery applications. We provide an overview of different carriers used to increase drug loading capacity and different targeting strategies that can be used to enhance local delivery, potentiate therapeutic efficacy, and minimize side effects. Additionally, future directions are proposed to improve the tMB performance in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, CO 80309, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
8
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
9
|
Piorecka K, Kurjata J, Stanczyk WA. Acriflavine, an Acridine Derivative for Biomedical Application: Current State of the Art. J Med Chem 2022; 65:11415-11432. [PMID: 36018000 PMCID: PMC9469206 DOI: 10.1021/acs.jmedchem.2c00573] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Acriflavine (ACF) has been known for years as an antibacterial
drug. The identification of key oncogenic mechanisms has brought,
in recent years, a significant increase in studies on ACF as a multipurpose
drug that would improve the prognosis for cancer patients. ACF interferes
with the expression of the hypoxia inducible factor, thus acting on
metastatic niches of tumors and significantly enhancing the effects
of other anticancer therapies. It has been recognized as the most
potent HIF-1 inhibitor out of the 336 drugs approved by the FDA. This
work presents up-to-date knowledge about the mechanisms of action
of ACF and its related prodrug systems in the context of anticancer
and SARS-CoV-2 inhibitory properties. It explains the multitask nature
of this drug and suggests mechanisms of ACF’s action on the
coronavirus. Other recent reports on ACF-based systems as potential
antibacterial and antiviral drugs are also described.
Collapse
Affiliation(s)
- Kinga Piorecka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Jan Kurjata
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Wlodzimierz A Stanczyk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
10
|
Overcoming Hypoxia-Induced Drug Resistance via Promotion of Drug Uptake and Reoxygenation by Acousto–Mechanical Oxygen Delivery. Pharmaceutics 2022; 14:pharmaceutics14050902. [PMID: 35631488 PMCID: PMC9144555 DOI: 10.3390/pharmaceutics14050902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
Hypoxia-induced drug resistance (HDR) is a critical issue in cancer therapy. The presence of hypoxic tumor cells impedes drug uptake and reduces the cytotoxicity of chemotherapeutic drugs, leading to HDR and increasing the probability of tumor recurrence and metastasis. Microbubbles, which are used as an ultrasound contrast agent and drug/gas carrier, can locally deliver drugs/gas and produce an acousto–mechanical effect to enhance cell permeability under ultrasound sonication. The present study applied oxygen-loaded microbubbles (OMBs) to evaluate the mechanisms of overcoming HDR via promotion of drug uptake and reoxygenation. A hypoxic mouse prostate tumor cell model was established by hypoxic incubation for 4 h. After OMB treatment, the permeability of HDR cells was enhanced by 23 ± 5% and doxorubicin uptake was increased by 11 ± 7%. The 61 ± 14% reoxygenation of HDR cells increased the cytotoxicity of doxorubicin from 18 ± 4% to 58 ± 6%. In combination treatment with OMB and doxorubicin, the relative contributions of uptake promotion and reoxygenation towards overcoming HDR were 11 ± 7% and 28 ± 10%, respectively. Our study demonstrated that reoxygenation of hypoxic conditions is a critical mechanism in the inhibition of HDR and enhancing the outcome of OMB treatment.
Collapse
|
11
|
Evaluation of Liposome-Loaded Microbubbles as a Theranostic Tool in a Murine Collagen-Induced Arthritis Model. Sci Pharm 2022. [DOI: 10.3390/scipharm90010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe inflammation of the synovial tissue. Here, we assess the feasibility of liposome-loaded microbubbles as theranostic agents in a murine arthritis model. First, contrast-enhanced ultrasound (CEUS) was used to quantify neovascularization in this model since CEUS is well-established for RA diagnosis in humans. Next, the potential of liposome-loaded microbubbles and ultrasound (US) to selectively enhance liposome delivery to the synovium was evaluated with in vivo fluorescence imaging. This procedure is made very challenging by the presence of hard joints and by the limited lifetime of the microbubbles. The inflamed knee joints were exposed to therapeutic US after intravenous injection of liposome-loaded microbubbles. Loaded microbubbles were found to be quickly captured by the liver. This resulted in fast clearance of attached liposomes while free and long-circulating liposomes were able to accumulate over time in the inflamed joints. Our observations show that murine arthritis models are not well-suited for evaluating the potential of microbubble-mediated drug delivery in joints given: (i) restricted microbubble passage in murine synovial vasculature and (ii) limited control over the exact ultrasound conditions in situ given the much shorter length scale of the murine joints as compared to the therapeutic wavelength.
Collapse
|
12
|
Highlights in ultrasound-targeted microbubble destruction-mediated gene/drug delivery strategy for treatment of malignancies. Int J Pharm 2021; 613:121412. [PMID: 34942327 DOI: 10.1016/j.ijpharm.2021.121412] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/05/2023]
Abstract
Ultrasound is one of the safest and most advanced medical imaging technologies that is widely used in clinical practice. Ultrasound microbubbles, traditionally used for contrast-enhanced imaging, are increasingly applied in Ultrasound-targeted Microbubble Destruction (UTMD) technology which enhances tissue and cell membrane permeability through cavitation and sonoporation, to result in a promising therapeutic gene/drug delivery strategy. Here, we review recent developments in the application of UTMD-mediated gene and drug delivery in the diagnosis and treatment of tumors, including the concept, mechanism of action, clinical application status, and advantages of UTMD. Furthermore, the future perspectives that should be paid more attention to in this field are prospected.
Collapse
|
13
|
Sun Y, Zhou Z, Yang S, Yang H. Modulating hypoxia inducible factor-1 by nanomaterials for effective cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1766. [PMID: 34713633 DOI: 10.1002/wnan.1766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
Hypoxia, which is induced by abnormal tumor growth when it outstrips its oxygen supply, is a major character of cancer. The reaction of cells against hypoxia is mainly concentrated on the hypoxia-induced transcription factors (HIFs), especially HIF-1, which remain stabilized during hypoxia. Additionally, the oxygen-independent mechanism of regulating HIF-1 acts a vital part in different stages of tumor progression as well as chemo-/radio-/PDT resistance, resulting in poor curative effects and prognosis. In this review, we will outline the up-to-date information about how HIF-1 interferes with tumor metastasis and therapy resistance, followed by a detailed introduction of motivating techniques based on various nanomaterials to interfere with HIF signaling for effective cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yun Sun
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Zhiguo Zhou
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Shiping Yang
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Hong Yang
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| |
Collapse
|
14
|
Krafft MP, Riess JG. Therapeutic oxygen delivery by perfluorocarbon-based colloids. Adv Colloid Interface Sci 2021; 294:102407. [PMID: 34120037 DOI: 10.1016/j.cis.2021.102407] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
After the protocol-related indecisive clinical trial of Oxygent, a perfluorooctylbromide/phospholipid nanoemulsion, in cardiac surgery, that often unduly assigned the observed untoward effects to the product, the development of perfluorocarbon (PFC)-based O2 nanoemulsions ("blood substitutes") has come to a low. Yet, significant further demonstrations of PFC O2-delivery efficacy have continuously been reported, such as relief of hypoxia after myocardial infarction or stroke; protection of vital organs during surgery; potentiation of O2-dependent cancer therapies, including radio-, photodynamic-, chemo- and immunotherapies; regeneration of damaged nerve, bone or cartilage; preservation of organ grafts destined for transplantation; and control of gas supply in tissue engineering and biotechnological productions. PFC colloids capable of augmenting O2 delivery include primarily injectable PFC nanoemulsions, microbubbles and phase-shift nanoemulsions. Careful selection of PFC and other colloid components is critical. The basics of O2 delivery by PFC nanoemulsions will be briefly reminded. Improved knowledge of O2 delivery mechanisms has been acquired. Advanced, size-adjustable O2-delivering nanoemulsions have been designed that have extended room-temperature shelf-stability. Alternate O2 delivery options are being investigated that rely on injectable PFC-stabilized microbubbles or phase-shift PFC nanoemulsions. The latter combine prolonged circulation in the vasculature, capacity for penetrating tumor tissues, and acute responsiveness to ultrasound and other external stimuli. Progress in microbubble and phase-shift emulsion engineering, control of phase-shift activation (vaporization), understanding and control of bubble/ultrasound/tissue interactions is discussed. Control of the phase-shift event and of microbubble size require utmost attention. Further PFC-based colloidal systems, including polymeric micelles, PFC-loaded organic or inorganic nanoparticles and scaffolds, have been devised that also carry substantial amounts of O2. Local, on-demand O2 delivery can be triggered by external stimuli, including focused ultrasound irradiation or tumor microenvironment. PFC colloid functionalization and targeting can help adjust their properties for specific indications, augment their efficacy, improve safety profiles, and expand the range of their indications. Many new medical and biotechnological applications involving fluorinated colloids are being assessed, including in the clinic. Further uses of PFC-based colloidal nanotherapeutics will be briefly mentioned that concern contrast diagnostic imaging, including molecular imaging and immune cell tracking; controlled delivery of therapeutic energy, as for noninvasive surgical ablation and sonothrombolysis; and delivery of drugs and genes, including across the blood-brain barrier. Even when the fluorinated colloids investigated are designed for other purposes than O2 supply, they will inevitably also carry and deliver a certain amount of O2, and may thus be considered for O2 delivery or co-delivery applications. Conversely, O2-carrying PFC nanoemulsions possess by nature a unique aptitude for 19F MR imaging, and hence, cell tracking, while PFC-stabilized microbubbles are ideal resonators for ultrasound contrast imaging and can undergo precise manipulation and on-demand destruction by ultrasound waves, thereby opening multiple theranostic opportunities.
Collapse
Affiliation(s)
- Marie Pierre Krafft
- University of Strasbourg, Institut Charles Sadron (CNRS), 23 rue du Loess, 67034 Strasbourg, France.
| | - Jean G Riess
- Harangoutte Institute, 68160 Ste Croix-aux-Mines, France
| |
Collapse
|
15
|
Jangjou A, Meisami AH, Jamali K, Niakan MH, Abbasi M, Shafiee M, Salehi M, Hosseinzadeh A, Amani AM, Vaez A. The promising shadow of microbubble over medical sciences: from fighting wide scope of prevalence disease to cancer eradication. J Biomed Sci 2021; 28:49. [PMID: 34154581 PMCID: PMC8215828 DOI: 10.1186/s12929-021-00744-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Microbubbles are typically 0.5-10 μm in size. Their size tends to make it easier for medication delivery mechanisms to navigate the body by allowing them to be swallowed more easily. The gas included in the microbubble is surrounded by a membrane that may consist of biocompatible biopolymers, polymers, surfactants, proteins, lipids, or a combination thereof. One of the most effective implementation techniques for tiny bubbles is to apply them as a drug carrier that has the potential to activate ultrasound (US); this allows the drug to be released by US. Microbubbles are often designed to preserve and secure medicines or substances before they have reached a certain area of concern and, finally, US is used to disintegrate microbubbles, triggering site-specific leakage/release of biologically active drugs. They have excellent therapeutic potential in a wide range of common diseases. In this article, we discussed microbubbles and their advantageous medicinal uses in the treatment of certain prevalent disorders, including Parkinson's disease, Alzheimer's disease, cardiovascular disease, diabetic condition, renal defects, and finally, their use in the treatment of various forms of cancer as well as their incorporation with nanoparticles. Using microbubble technology as a novel carrier, the ability to prevent and eradicate prevalent diseases has strengthened the promise of effective care to improve patient well-being and life expectancy.
Collapse
Affiliation(s)
- Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Hossein Meisami
- Department of Emergency Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kazem Jamali
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Niakan
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Shafiee
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahmad Hosseinzadeh
- Thoracic and Vascular Surgery Research Center, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
17
|
Chen L, Chai Y, Luo J, Wang J, Liu X, Wang T, Xu X, Zhou G, Feng X. Apoptotic changes and myofibrils degradation in post-mortem chicken muscles by ultrasonic processing. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.110985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Li C, Hu S, Yue Y. Ultrasound Microbubble-Mediated VHL Regulates the Biological Behavior of Ovarian Cancer Cells. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:723-732. [PMID: 33261909 DOI: 10.1016/j.ultrasmedbio.2020.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 06/12/2023]
Abstract
According to the literature, the von Hippel-Lindau (VHL) gene has a certain correlation with ovarian cancer. In this study, we investigated the effect and mechanism of ultrasound microbubble-mediated VHL on the biological function of ovarian cancer cells. Non-targeting lipid microbubbles and targeted lipid microbubbles were prepared. OVCAR-3 cells were treated with VHL mediated by ultrasound and microbubbles alone or together. Expressions of VHL, Akt, epithelial-mesenchymal-transition-related proteins and apoptosis-related proteins were detected by Western blot and quantitative real-time polymerase chain reaction as needed. The effect of ultrasound microbubble-mediated VHL on the proliferation, apoptosis, cell cycle, migration and invasion of OVCAR-3 cells was examined by Cell Counting Kit-8, flow cytometry, wound-healing assay and Transwell. Compared with other treatment methods, ultrasound microbubble mediation enhanced VHL expression in OVCAR-3 cells. Overexpression of liposome-mediated VHL inhibited the proliferation and migration; caused cell-cycle arrest; promoted apoptosis: downregulated the expressions of MMP2, MMP9, E-cadherin, Akt and Bcl-2; and upregulated the expressions of VHL and BCL2-associated X protein (BAX) in OVCAR-3 cells. The effect of microbubble-treated VHL was similar to liposome-mediated regulation, while ultrasound treatment enhanced the effect of VHL on OVCAR-3 cells. More interestingly, ultrasound microbubble-mediated VHL had the most obvious regulatory effect on OVCAR-3 cells. Ultrasound microbubble technology increases the transfection efficiency of VHL into OVCAR-3 cells and enhances the effect of VHL gene on the biological function of OVCAR-3 cells.
Collapse
Affiliation(s)
- Cong Li
- Ultrasonography Department, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Suling Hu
- Functional Department, Baoding Infectious Disease Hospital, Baoding, Hebei Province, China
| | - Yan Yue
- Department of Gynaecology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
19
|
Recent advances in ultrasound-triggered drug delivery through lipid-based nanomaterials. Drug Discov Today 2020; 25:2182-2200. [PMID: 33010479 DOI: 10.1016/j.drudis.2020.09.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/24/2020] [Accepted: 09/22/2020] [Indexed: 12/28/2022]
Abstract
The high prescribed dose of anticancer drugs and their resulting adverse effects on healthy tissue are significant drawbacks to conventional chemotherapy (CTP). Ideally, drugs should have the lowest possible degree of interaction with healthy cells, which would diminish any adverse effects. Therefore, an ideal scenario to bring about improvements in CTP is the use of technological strategies to ensure the efficient, specific, and selective transport and/or release of drugs to the target site. One practical and feasible solution to promote the efficiency of conventional CTP is the use of ultrasound (US). In this review, we highlight the potential role of US in combination with lipid-based carriers to achieve a targeted CTP strategy in engineered smart drug delivery systems.
Collapse
|
20
|
Al-Jawadi S, Thakur SS. Ultrasound-responsive lipid microbubbles for drug delivery: A review of preparation techniques to optimise formulation size, stability and drug loading. Int J Pharm 2020; 585:119559. [PMID: 32574685 DOI: 10.1016/j.ijpharm.2020.119559] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023]
Abstract
Lipid-shelled microbubbles have received extensive interest to enhance ultrasound-responsive drug delivery outcomes due to their high biocompatibility. While therapeutic effectiveness of microbubbles is well established, there remain limitations in sample homogeneity, stability profile and drug loading properties which restrict these formulations from seeing widespread use in the clinical setting. In this review, we evaluate and discuss the most encouraging leads in lipid microbubble design and optimisation. We examine current applications in drug delivery for the systems and subsequently detail shell compositions and preparation strategies that improve monodispersity while retaining ultrasound responsiveness. We review how excipients and storage techniques help maximise stability and introduce different characterisation and drug loading techniques and evaluate their impact on formulation performance. The review concludes with current quality control measures in place to ensure lipid microbubbles can be reproducibly used in drug delivery.
Collapse
Affiliation(s)
- Sana Al-Jawadi
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Sachin S Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
21
|
Chattaraj R, Hwang M, Zemerov SD, Dmochowski IJ, Hammer DA, Lee D, Sehgal CM. Ultrasound Responsive Noble Gas Microbubbles for Applications in Image-Guided Gas Delivery. Adv Healthc Mater 2020; 9:e1901721. [PMID: 32207250 PMCID: PMC7457952 DOI: 10.1002/adhm.201901721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 12/18/2022]
Abstract
Noble gases, especially xenon (Xe), have been shown to have antiapoptotic effects in treating hypoxia ischemia related injuries. Currently, in vivo gas delivery is systemic and performed through inhalation, leading to reduced efficacy at the injury site. This report provides a first demonstration of the encapsulation of pure Xe, Ar, or He in phospholipid-coated sub-10 µm microbubbles, without the necessity of stabilizing perfluorocarbon additives. Optimization of shell compositions and preparation techniques show that distearoylphosphatidylcholine (DSPC) with DSPE-PEG5000 can produce stable microbubbles upon shaking, while dibehenoylphosphatidylcholine (DBPC) blended with either DSPE-PEG2000 or DSPE-PEG5000 produces a high yield of microbubbles via a sonication/centrifugation method. Xe and Ar concentrations released into the microbubble suspension headspace are measured using GC-MS, while Xe released directly in solution is detected by the fluorescence quenching of a Xe-sensitive cryptophane molecule. Bubble production is found to be amenable to scale-up while maintaining their size distribution and stability. Excellent ultrasound contrast is observed in a phantom for several minutes under physiological conditions, while an intravenous administration of a bolus of pure Xe microbubbles provides significant contrast in a mouse in pre- and post-lung settings (heart and kidney, respectively), paving the way for image-guided, localized gas delivery for theranostic applications.
Collapse
Affiliation(s)
- Rajarshi Chattaraj
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Misun Hwang
- Department of Radiology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, United States; Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Serge D. Zemerov
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ivan J Dmochowski
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Daniel A. Hammer
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Daeyeon Lee
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Chandra M. Sehgal
- Department of Radiology, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
22
|
Nimmagadda S, Penet MF. Ovarian Cancer Targeted Theranostics. Front Oncol 2020; 9:1537. [PMID: 32039018 PMCID: PMC6985364 DOI: 10.3389/fonc.2019.01537] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/19/2019] [Indexed: 11/20/2022] Open
Abstract
Ovarian cancer is a leading cause of death from gynecological malignancies. Although the prognosis is quite favorable if detected at an early stage, the vast majority of cases are diagnosed at an advanced stage, when 5-year survival rates are only 30–40%. Most recurrent ovarian tumors are resistant to traditional therapies underscoring the need for new therapeutic options. Theranostic agents, that combine diagnostic and therapeutic capabilities, are being explored to better detect, diagnose and treat ovarian cancer. To minimize morbidity, improve survival rates, and eventually cure patients, new strategies are needed for early detection and for delivering specifically anticancer therapies to tumor sites. In this review we will discuss various molecular imaging modalities and targets that can be used for imaging, therapeutic and theranostic agent development for improved diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Sridhar Nimmagadda
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marie-France Penet
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
23
|
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
|
24
|
Mérida F, Rinaldi C, Juan EJ, Torres-Lugo M. In vitro Ultrasonic Potentiation of 2-Phenylethynesulfonamide/Magnetic Fluid Hyperthermia Combination Treatments for Ovarian Cancer. Int J Nanomedicine 2020; 15:419-432. [PMID: 32021188 PMCID: PMC6982443 DOI: 10.2147/ijn.s217870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/25/2019] [Indexed: 01/15/2023] Open
Abstract
Background Magnetic Fluid Hyperthermia (MFH) is a promising adjuvant for chemotherapy, potentiating the action of anticancer agents. However, drug delivery to cancer cells must be optimized to improve the overall therapeutic effect of drug/MFH combination treatments. Purpose The aim of this work was to demonstrate the potentiation of 2-phenylethynesulfonamide (PES) at various combination treatments with MFH, using low-intensity ultrasound as an intracellular delivery enhancer. Methods The effect of ultrasound (US), MFH, and PES was first evaluated individually and then as combination treatments. Definity® microbubbles and polyethylene glycol (PEG)-coated iron oxide nanoparticles were used to induce cell sonoporation and MFH, respectively. Assessment of cell membrane permeabilization was evaluated via fluorescence microscopy, iron uptake by cells was quantified by UV-Vis spectroscopy, and cell viability was determined using automatic cell counting. Results Notable reductions in cancer cell viability were observed when ultrasound was incorporated. For example, the treatment US+PES reduced cell viability by 37% compared to the non-toxic effect of the drug. Similarly, the treatment US+MFH using mild hyperthermia (41°C), reduced cell viability by an additional 18% when compared to the effect of MH alone. Significant improvements were observed for the combination of US+PES+MFH with cell viability reduced by an additional 26% compared to the PES+MFH group. The improved cytotoxicity was attributed to enhanced drug/nanoparticle intracellular delivery, with iron uptake values nearly twice those achieved without ultrasound. Various treatment schedules were examined, and all of them showed substantial cell death, indicating that the time elapsed between sonoporation and magnetic field exposure was not significant. Conclusion Superior cancer cell-killing patterns took place when ultrasound was incorporated thus demonstrating the in vitro ultrasonic potentiation of PES and mild MFH. This work demonstrated that ultrasound is a promising non-invasive enhancer of PES/MFH combination treatments, aiming to establish a sono-thermo-chemotherapy in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Fernando Mérida
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| | - Carlos Rinaldi
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA.,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Eduardo J Juan
- Department of Electrical and Computer Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| | - Madeline Torres-Lugo
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| |
Collapse
|
25
|
Integration of cascade delivery and tumor hypoxia modulating capacities in core-releasable satellite nanovehicles to enhance tumor chemotherapy. Biomaterials 2019; 223:119465. [DOI: 10.1016/j.biomaterials.2019.119465] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/19/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022]
|
26
|
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
|
27
|
Ho YJ, Chu SW, Liao EC, Fan CH, Chan HL, Wei KC, Yeh CK. Normalization of Tumor Vasculature by Oxygen Microbubbles with Ultrasound. Am J Cancer Res 2019; 9:7370-7383. [PMID: 31695774 PMCID: PMC6831304 DOI: 10.7150/thno.37750] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/30/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor microenvironment influences the efficacy of anti-cancer therapies. The dysfunctional tumor vasculature limits the efficiency of oxygenation and drug delivery to reduce treatment outcome. A concept of tumor vascular normalization (VN), which inhibits angiogenesis to improve vessel maturity, blood perfusion, and oxygenation, has been demonstrated under the anti-angiogenic therapy. The efficiency of drug delivery and penetration is increased by enhancing perfusion and reducing interstitial fluid pressure during the time window of VN. However, anti-angiogenic agents only induce transient VN and then prune vessels to aggravate tumor hypoxia. To repair tumor vessels without altering vessel density, we proposed to induce tumor VN by local oxygen release via oxygen microbubbles with ultrasound. With tumor perfusion enhancement under ultrasound contrast imaging tracing, the time window of VN was defined as 2-8 days after a single oxygen microbubble treatment. The enhanced tumor oxygenation after oxygen microbubble treatment inhibited hypoxia inducible factor-1 alpha (HIF-1α)/vascular endothelial growth factor (VEGF) pathway to improve the morphology and function of tumor vasculature. The pericyte coverage and Hoechst penetration of tumor vessels increased without any changes to the vessel density. Finally, the intratumoral accumulation of anti-cancer drug doxorubicin could be increased 3-4 folds during tumor VN. These findings demonstrate that regulating tumor oxygenation by oxygen microbubbles could normalize dysfunctional vessels to enhance vascular maturity, blood perfusion, and drug penetration. Furthermore, ultrasound perfusion imaging provides a simple and non-invasive way to detect the VN time window, which increases the feasibility of VN in clinical cancer applications.
Collapse
|
28
|
Abenojar EC, Nittayacharn P, de Leon AC, Perera R, Wang Y, Bederman I, Exner AA. Effect of Bubble Concentration on the in Vitro and in Vivo Performance of Highly Stable Lipid Shell-Stabilized Micro- and Nanoscale Ultrasound Contrast Agents. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10192-10202. [PMID: 30913884 DOI: 10.1021/acs.langmuir.9b00462] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Ultrasound (US) is a widely used diagnostic imaging tool because it is inexpensive, safe, portable, and broadly accessible. Ultrasound contrast agents (UCAs) are employed to enhance backscatter echo and improve imaging contrast. The most frequently utilized UCAs are echogenic bubbles made with a phospholipid or protein-stabilized hydrophobic gas core. While clinically utilized, applications of UCAs are often limited by rapid signal decay (<5 min) in vivo under typical ultrasound imaging protocols. Here, we report on a formulation of lipid shell-stabilized perfluoropropane (C3F8) microbubbles and nanobubbles with a significantly prolonged in vivo stability. Microbubbles (875 ± 280 nm) of the target size were prepared by utilizing a multiple-step centrifugation cycle, while nanobubbles (299 ± 189 nm) were isolated from the activated vial using a single centrifugation step. To provide in-depth acoustic characterization of the new construct we evaluated the effect of size and concentration on their in vitro and in vivo performance. In vitro and in vivo characterization were carried out for a range of bubble concentrations normalized by total gas volume quantified via headspace gas chromatography/mass spectrometry (GC/MS). In vitro characterization revealed that nanobubbles at different concentrations are more consistently stable over time with the highest and lowest dilutions (50-fold decrease) only differing in US signal after 8 min exposure by 10.34%, while for microbubbles the difference was 86.46%. As expected, due to the difference in hydrodynamic diameter and scattering cross section difference, nanobubbles showed lower overall initial signal intensity. In vivo experiments showed that both microbubbles and nanobubbles with similar initial peak signal intensity are comparably stable over time with 66.8% and 60.6% remaining signal after 30 min, respectively. This study demonstrates that bubble concentration has significant effects on the persistence of both microbubbles and nanobubbles in vitro and in vivo, but the effects are more pronounced in larger bubbles. These effects should be taken into account when selecting the appropriate bubble parameters for future imaging applications.
Collapse
Affiliation(s)
- Eric C Abenojar
- Department of Radiology , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Pinunta Nittayacharn
- Department of Biomedical Engineering , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Al Christopher de Leon
- Department of Radiology , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Reshani Perera
- Department of Radiology , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Yu Wang
- Department of Radiology , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Ilya Bederman
- Department of Pediatrics, School of Medicine , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Agata A Exner
- Department of Radiology , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
- Department of Biomedical Engineering , Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| |
Collapse
|
29
|
Zhou Q, Zeng Y, Xiong Q, Zhong S, Li P, Ran H, Yin Y, Reutelingsperger C, Prinze FW, Ling Z. Construction of CNA35 Collagen-Targeted Phase-Changeable Nanoagents for Low-Intensity Focused Ultrasound-Triggered Ultrasound Molecular Imaging of Myocardial Fibrosis in Rabbits. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23006-23017. [PMID: 31136144 DOI: 10.1021/acsami.9b05999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Myocardial fibrosis plays an important role in the development of heart failure and malignant arrhythmia, which potentially increases the incidence of sudden cardiac death. Therefore, early detection of myocardial fibrosis is of great significance for evaluating the prognosis of patients and formulating appropriate treatment strategies. Late gadolinium-enhanced magnetic resonance imaging is considered as the currently effective strategy for noninvasive detection of myocardial fibrosis, but it still suffers from some critical issues. In this work, multifunctional CNA35-labeled perfluoropentane nanoparticles (CNA35-PFP NPs) have been elaborately designed and constructed for molecular imaging of fibrotic myocardium based on ultrasound imaging. These as-constructed CNA35-PFP NPs are intravenously infused into rabbit circulation with an animal model of myocardial infarction. Especially, these targeted CNA35-PFP NPs with nanoscale size could efficiently pass through the endothelial cell gap and adhere to the surface of fibroblasts in the fibrotic myocardium. Importantly, followed by low-intensity focused ultrasound irradiation on the myocardium, these intriguing CNA35-PFP NPs could transform from liquid into gaseous microbubbles, which further significantly enhanced the ultrasound contrast in the fibrotic area, facilitating the detection by diagnostic ultrasound imaging. Therefore, this work provides a desirable noninvasive, economical, and real-time imaging technique for the assessment of cardiac fibrosis with diagnostic ultrasound based on the rational design of liquid-to-gas phase-changeable nanoplatforms.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yalin Zeng
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Qingsong Xiong
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Shigen Zhong
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Pan Li
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Haitao Ran
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yuehui Yin
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Chris Reutelingsperger
- Department of Physiology, Cardiovascular Research Institute Maastricht , Maastricht University , P.O. Box 616 , 6200 MD , Maastricht , The Netherlands
| | - Frits W Prinze
- Department of Biochemistry, Cardiovascular Research Institute Maastricht , University of Maastricht , P.O. Box 616 , 6200 MD , Maastricht , The Netherlands
| | - Zhiyu Ling
- Department of Cardiology , Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Institute of Ultrasound Imaging, Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| |
Collapse
|
30
|
Han S, Dwivedi P, Mangrio FA, Dwivedi M, Khatik R, Cohn DE, Si T, Xu RX. Sustained release paclitaxel-loaded core-shell-structured solid lipid microparticles for intraperitoneal chemotherapy of ovarian cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:957-967. [DOI: 10.1080/21691401.2019.1576705] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Shuya Han
- Department of Precision Machinery and Precision Instrumentation, School of Engineering Science, University of Science and Technology of China, Hefei, P.R.China
| | - Pankaj Dwivedi
- Department of Precision Machinery and Precision Instrumentation, School of Engineering Science, University of Science and Technology of China, Hefei, P.R.China
| | - Farhana Akbar Mangrio
- Department of Precision Machinery and Precision Instrumentation, School of Engineering Science, University of Science and Technology of China, Hefei, P.R.China
| | - Monika Dwivedi
- Department of Precision Machinery and Precision Instrumentation, School of Engineering Science, University of Science and Technology of China, Hefei, P.R.China
| | - Renuka Khatik
- Department of Chemistry, Laboratory of Nanomaterials for Energy Conversion (LNEC), University of Science and Technology of China, Hefei, Anhui, PR China
| | - David E. Cohn
- Division of Gynecologic Oncology, Ohio State University College of Medicine, Columbus, OH, USA
| | - Ting Si
- Department of Precision Machinery and Precision Instrumentation, School of Engineering Science, University of Science and Technology of China, Hefei, P.R.China
| | - Ronald X. Xu
- Department of Precision Machinery and Precision Instrumentation, School of Engineering Science, University of Science and Technology of China, Hefei, P.R.China
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
31
|
Gonet M, Epel B, Elas M. Data processing of 3D and 4D in-vivo electron paramagnetic resonance imaging co-registered with ultrasound. 3D printing as a registration tool. COMPUTERS & ELECTRICAL ENGINEERING : AN INTERNATIONAL JOURNAL 2019; 74:130-137. [PMID: 30820068 PMCID: PMC6388699 DOI: 10.1016/j.compeleceng.2019.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We present the concept of image registration using ultrasound (US) and electron paramagnetic resonance (EPR) imaging and discuss the benefits of this solution, as well as its limitations. Both phantoms and murine tumors were used to test US and EPR image co-registration. Comparison of dental molding cast immobilization and predesigned cradle revealed that the latter approach is more effective in stabilizing the fiducial position. In vivo imaging of mouse tumors, image registration and comparison of fiducials system for 3D spatial as well as 4D spatial-spectral EPR imaging supported by 3D US were demonstrated. Ultrasound may provide a convenient alternative to other anatomical imaging methods for image registration in preclinical research. Of particular interest is a fusion of US tissue structure, doppler vascular function and EPR oxygen or redox imaging.
Collapse
Affiliation(s)
- M Gonet
- 1. Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- 3. Novilet, Poznan, Poland
| | - B Epel
- 2. Department of Radiation and Cellular Oncology, and Center for EPR Imaging In Vivo Physiology, University of Chicago, Chicago, IL 60637, United States
| | - M Elas
- 1. Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
32
|
Upadhyay A, Dalvi SV. Microbubble Formulations: Synthesis, Stability, Modeling and Biomedical Applications. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:301-343. [PMID: 30527395 DOI: 10.1016/j.ultrasmedbio.2018.09.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 05/12/2023]
Abstract
Microbubbles are increasingly being used in biomedical applications such as ultrasonic imaging and targeted drug delivery. Microbubbles typically range from 0.1 to 10 µm in size and consist of a protective shell made of lipids or proteins. The shell encapsulates a gaseous core containing gases such as oxygen, sulfur hexafluoride or perfluorocarbons. This review is a consolidated account of information available in the literature on research related to microbubbles. Efforts have been made to present an overview of microbubble synthesis techniques; microbubble stability; microbubbles as contrast agents in ultrasonic imaging and drug delivery vehicles; and side effects related to microbubble administration in humans. Developments related to the modeling of microbubble dissolution and stability are also discussed.
Collapse
Affiliation(s)
- Awaneesh Upadhyay
- Chemical Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, India
| | - Sameer V Dalvi
- Chemical Engineering, Indian Institute of Technology Gandhinagar, Gandhinagar, India.
| |
Collapse
|
33
|
Dong F, Zhang J, Wang K, Liu Z, Guo J, Zhang J. Cold plasma gas loaded microbubbles as a novel ultrasound contrast agent. NANOSCALE 2019; 11:1123-1130. [PMID: 30574971 DOI: 10.1039/c8nr08451c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nowadays, cold atmospheric plasma (CAP) that contains lots of active free radicals has tremendous potential applications in biomedical engineering, and target delivery of a controllable dose of plasma gas is highly desired in clinical use. In this conceptual study, we developed a novel microbubble loaded by plasma gas and proposed an ultrasound-triggered strategy for the ultrasound-triggered release of free radicals from the microbubbles. The plasma microbubbles (PMBs) were fabricated by mixing plasma gas in the core of the surfactant microbubbles by a modified emulsification process. The resulting PMBs with an average size of 2.54 ± 2.28 μm were successfully fabricated using the proposed approach and the experimental result showed that PMBs exhibited a satisfactory ability to meet the requirement of ultrasound contrast-enhanced imaging. Furthermore, we depicted that ultrasound induced PMB destruction to release the plasma gas and PMBs with ultrasound stimulation could significantly improve the concentration of nitric oxide and hydrogen peroxide compared with the control group. In addition, Dil acting as a model drug was loaded into the PMBs and an in vitro cell experiment showed that Dil and plasma gas could be released from PMBs and internalized by PIEC cells with ultrasound mediation. Our experimental results showed that ultrasound induced PMB destruction could successfully release many active free radicals in plasma gas, including nitric oxide and hydrogen peroxide. The developed novel microbubbles demonstrated the technical potential of plasma gas loaded MBs for disease diagnostics and therapy with ultrasound imaging guidance.
Collapse
Affiliation(s)
- Feihong Dong
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | | | | | | | | | | |
Collapse
|
34
|
Khan MS, Hwang J, Lee K, Choi Y, Kim K, Koo HJ, Hong JW, Choi J. Oxygen-Carrying Micro/Nanobubbles: Composition, Synthesis Techniques and Potential Prospects in Photo-Triggered Theranostics. Molecules 2018; 23:E2210. [PMID: 30200336 PMCID: PMC6225314 DOI: 10.3390/molecules23092210] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/27/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022] Open
Abstract
Microbubbles and nanobubbles (MNBs) can be prepared using various shells, such as phospholipids, polymers, proteins, and surfactants. MNBs contain gas cores due to which they are echogenic and can be used as contrast agents for ultrasonic and photoacoustic imaging. These bubbles can be engineered in various sizes as vehicles for gas and drug delivery applications with novel properties and flexible structures. Hypoxic areas in tumors develop owing to an imbalance of oxygen supply and demand. In tumors, hypoxic regions have shown more resistance to chemotherapy, radiotherapy, and photodynamic therapies. The efficacy of photodynamic therapy depends on the effective accumulation of photosensitizer drug in tumors and the availability of oxygen in the tumor to generate reactive oxygen species. MNBs have been shown to reverse hypoxic conditions, degradation of hypoxia inducible factor 1α protein, and increase tissue oxygen levels. This review summarizes the synthesis methods and shell compositions of micro/nanobubbles and methods deployed for oxygen delivery. Methods of functionalization of MNBs, their ability to deliver oxygen and drugs, incorporation of photosensitizers and potential application of photo-triggered theranostics, have also been discussed.
Collapse
Affiliation(s)
- Muhammad Saad Khan
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Jangsun Hwang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Kyungwoo Lee
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Kyobum Kim
- Division of Bioengineering, Incheon National University, Incheon 22012, Korea.
| | - Hyung-Jun Koo
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Korea.
| | - Jong Wook Hong
- Department of Bionano Technology, Hanyang University, Seoul 04763, Korea.
- Department of Bionano Engingeering, Hanyang University, Ansan 15588, Korea.
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| |
Collapse
|
35
|
Meng L, Cheng Y, Tong X, Gan S, Ding Y, Zhang Y, Wang C, Xu L, Zhu Y, Wu J, Hu Y, Yuan A. Tumor Oxygenation and Hypoxia Inducible Factor-1 Functional Inhibition via a Reactive Oxygen Species Responsive Nanoplatform for Enhancing Radiation Therapy and Abscopal Effects. ACS NANO 2018; 12:8308-8322. [PMID: 30102510 DOI: 10.1021/acsnano.8b03590] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Hypoxia, and hypoxia inducible factor-1 (HIF-1), can induce tumor resistance to radiation therapy. To overcome hypoxia-induced radiation resistance, recent studies have described nanosystems to improve tumor oxygenation for immobilizing DNA damage and simultaneously initiate oxygen-dependent HIF-1α degradation. However, HIF-1α degradation is incomplete during tumor oxygenation treatment alone. Therefore, tumor oxygenation combined with residual HIF-1 functional inhibition is crucial to optimizing therapeutic outcomes of radiotherapy. Here, a reactive oxygen species (ROS) responsive nanoplatform is reported to successfully add up tumor oxygenation and HIF-1 functional inhibition. This ROS responsive nanoplatform, based on manganese dioxide (MnO2) nanoparticles, delivers the HIF-1 inhibitor acriflavine and other hydrophilic cationic drugs to tumor tissues. After reacting with overexpressed hydrogen peroxide (H2O2) within tumor tissues, Mn2+ and oxygen molecules are released for magnetic resonance imaging and tumor oxygenation, respectively. Cooperating with the HIF-1 functional inhibition, the expression of tumor invasion-related signaling molecules (VEGF, MMP-9) is obviously decreased to reduce the risk of metastasis. Furthermore, the nanoplatform could relieve T-cell exhaustion via downregulation of PD-L1, whose effects are similar to the checkpoint inhibitor PD-L1 antibody, and subsequently activates tumor-specific immune responses against abscopal tumors. These therapeutic benefits including increased X-ray-induced damage, downregulated resistance, and T-cell exhaustion related proteins expression achieved synergistically the optimal inhibition of tumor growth. Overall, this designed ROS responsive nanoplatform is of great potential in the sensitization of radiation for combating primary and metastatic tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lei Xu
- College of Biotechnology and Pharmaceutical Engineering , Nanjing Tech University , Nanjing 211800 , China
| | - Yishen Zhu
- College of Biotechnology and Pharmaceutical Engineering , Nanjing Tech University , Nanjing 211800 , China
| | - Jinhui Wu
- Institute of Drug R&D , Medical School of Nanjing University , Nanjing 210093 , China
| | - Yiqiao Hu
- Institute of Drug R&D , Medical School of Nanjing University , Nanjing 210093 , China
| | - Ahu Yuan
- Institute of Drug R&D , Medical School of Nanjing University , Nanjing 210093 , China
| |
Collapse
|
36
|
Calcitriol and Calcidiol Can Sensitize Melanoma Cells to Low⁻LET Proton Beam Irradiation. Int J Mol Sci 2018; 19:ijms19082236. [PMID: 30065179 PMCID: PMC6122082 DOI: 10.3390/ijms19082236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/19/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022] Open
Abstract
Proton beam irradiation promises therapeutic utility in the management of uveal melanoma. Calcitriol (1,25(OH)2D3)—the biologically active metabolite of vitamin D3—and its precursor, calcidiol (25(OH)D3), exert pleiotropic effects on melanoma cells. The aim of the study was to evaluate the effect of both calcitriol and calcidiol on melanoma cell proliferation and their response to proton beam irradiation. Three melanoma cell lines (human SKMEL-188 and hamster BHM Ma and BHM Ab), pre-treated with 1,25(OH)2D3 or 25(OH)D3 at graded concentrations (0, 10, 100 nM), were irradiated with 0–5 Gy and then cultured in vitro. Growth curves were determined by counting the cell number every 24 h up to 120 h, which was used to calculate surviving fractions. The obtained survival curves were analysed using two standard models: linear-quadratic and multi-target single hit. Calcitriol inhibited human melanoma proliferation at 10 nM, while only calcidiol inhibited proliferation of hamster lines at 10 and 100 nM doses. Treatment with either 1,25(OH)2D3 or 25(OH)D3 radio sensitized melanoma cells to low doses of proton beam radiation. The strength of the effect increased with the concentration of vitamin D3. Our data suggest that vitamin D3 may be an adjuvant that modifies proton beam efficiency during melanoma therapy.
Collapse
|
37
|
Chen Y, Sun L, Guo D, Wu Z, Chen W. Co-delivery of hypoxia inducible factor-1α small interfering RNA and 5-fluorouracil to overcome drug resistance in gastric cancer SGC-7901 cells. J Gene Med 2017; 19. [PMID: 29106062 DOI: 10.1002/jgm.2998] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/21/2017] [Accepted: 10/22/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Drug resistance cancer cells have become a major problem in chemotherapy. To solve this problem, the co-delivery of small interefering RNA (siRNA) and 5-fluorouracil chitosan nanoparticles was employed, aiming to reverse the multidrug resistance of gastric cancer SGC-7901 cells in vitro. METHODS Chitosan nanoparticles were prepared using an ionic gel method. siRNA nanoparticles were characterized by gel retardation assays. Particle size and zeta potential were measured to confirm nanoparticle formation. The transfection efficiency of siRNA was determined by flow cytometry and high-content screening. Western blotting and a quantitative real-time-polymerase chain reaction were used to assess the silencing efficiency of siRNA. Accumulation and efflux experiments for rhodamine-123, cell migration experiments, cell sensitivity analyses and cell apoptosis assays were used to determine whether siRNA could reverse multidrug resistance. A systemic toxicity assay was used to evaluate the safety of nanoparticles. RESULTS Compared to naked siRNA, the co-delivery system demonstrated a higher transfection efficiency and gene silencing efficiency by inhibiting the efflux of P-glycoprotein and cell migration. Moreover, the combination treatment with siRNA and 5-fluorouracil co-delivered by chitosan nanoparticles can increase the sensitivity of drug resistance cells and cell apoptosis. Finally, the safety of nanoparticles was evaluated in vivo and the results obtained suggested that nanoparticles did not have any obvious toxicity. CONCLUSIONS Co-delivery of siRNA and 5-fluorouracil chitosan nanoparticles is an attractive strategy for overcoming multidrug resistance.
Collapse
Affiliation(s)
- Yunna Chen
- The College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Academy of Chinese Medicine, Anhui Hefei, China.,Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, China
| | - Li Sun
- The College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Dongdong Guo
- The College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Academy of Chinese Medicine, Anhui Hefei, China.,Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, China
| | - Ziteng Wu
- The College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Academy of Chinese Medicine, Anhui Hefei, China.,Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, China
| | - Weidong Chen
- The College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Anhui Academy of Chinese Medicine, Anhui Hefei, China.,Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
38
|
Ma R, Wu Q, Si T, Chang S, Xu RX. Oxygen and Indocyanine Green loaded microparticles for dual-mode imaging and sonodynamic treatment of cancer cells. ULTRASONICS SONOCHEMISTRY 2017; 39:197-207. [PMID: 28732936 DOI: 10.1016/j.ultsonch.2017.03.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 06/07/2023]
Abstract
Oxygen and Indocyanine Green (ICG) loaded microparticles (OI-MPs) were fabricated by a gas-driven coaxial flow focusing (CFF) process for dual-mode imaging and sonodynamic therapy (SDT). The produced OI-MPs agent showed stable optical properties, superior imaging depth in near infrared (NIR) fluorescence imaging, and enhanced acoustic contrast after ultrasound mediation. We hypothesized that encapsulating ICG and oxygen in microparticles would enhance reactive oxygen species (ROS) production in SDT. This hypothesis was validated in a cell-free environment. We further hypothesized that ultrasound mediated fragmentation of the OI-MPs would induce cytotoxicity and apoptosis of cancer cells. This hypothesis was validated in SKOV3 ovarian cancer cells. Our research demonstrated that OI-MPs can be potentially used as a dual-mode theranostic agent for image guided SDT with enhanced efficacy. Further study is needed to delineate the mechanism of ROS-induced cell apoptosis and optimize the OI-MPs formulation for the maximal anti-cancer potency.
Collapse
Affiliation(s)
- Rong Ma
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Qiang Wu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ting Si
- Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Ronald X Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230027, China; Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
39
|
Ma X, Huang B, Wang G, Fu X, Qiu S. Numerical simulation of the red blood cell aggregation and deformation behaviors in ultrasonic field. ULTRASONICS SONOCHEMISTRY 2017; 38:604-613. [PMID: 27590752 DOI: 10.1016/j.ultsonch.2016.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/05/2016] [Accepted: 08/15/2016] [Indexed: 06/06/2023]
Abstract
The objective of this paper is to propose an immersed boundary lattice Boltzmann method (IB-LBM) considering the ultrasonic effect to simulate red blood cell (RBC) aggregation and deformation in ultrasonic field. Numerical examples involving the typical streamline, normalized out-of-plane vorticity contours and vector fields in pure plasma under three different ultrasound intensities are presented. Meanwhile, the corresponding transient aggregation behavior of RBCs, with special emphasis on the detailed process of RBC deformation, is shown. The numerical results reveal that the ultrasound wave acted on the pure plasma can lead to recirculation flow, which contributes to the RBCs aggregation and deformation in microvessel. Furthermore, increasing the intensity of the ultrasound wave can significantly enhance the aggregation and deformation of the RBCs. And the formation of the RBCs aggregation leads to the fluctuated and dropped vorticity value of plasma in return.
Collapse
Affiliation(s)
- Xiaojian Ma
- School of Mechanical and Vehicular Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Biao Huang
- School of Mechanical and Vehicular Engineering, Beijing Institute of Technology, Beijing 100081, China.
| | - Guoyu Wang
- School of Mechanical and Vehicular Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoying Fu
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University, Chengdu 610065, China
| | - Sicong Qiu
- School of Mechanical and Vehicular Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
40
|
Xu J, Yuan S, Tian J, Martin KA, Song J, Li C, Wang Z, Lin J, Si T, Xu RX. Ultrasound mediated delivery of oxygen and LLL12 loaded stimuli responsive microdroplets for the treatment of hypoxic cancer cells. Sci Rep 2017; 7:44908. [PMID: 28322306 PMCID: PMC5359616 DOI: 10.1038/srep44908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/14/2017] [Indexed: 01/05/2023] Open
Abstract
LLL12 exhibits high specificity for inhibiting STAT3 phosphorylation and dimerization, and inducing apoptosis to constitutively activated STAT3 cancer cells without cytotoxicity to normal cells with dormant STAT3. However, clinical deployment of LLL12 in cancer treatment is hindered by its low bioavailability and hypoxia-induced resistance. To overcome these limitations, we encapsulate both oxygen and LLL12 in stimuli responsive microdroplets (SRMs) by a gas-driven coaxial flow focusing (CFF) process for ultrasound mediated treatment of hypoxic cancer cells. Our benchtop experiments demonstrate that the CFF process is able to produce SRMs with uniform size distribution, large oxygen loading capacity, high LLL12 encapsulation efficiency, well protection of bioactivity, and steadily long shelf time. The in vitro therapeutic studies in pancreatic cancer cells (PANC-1 and CAPAN-1) demonstrate the immediate release of oxygen and LLL12 in exposure to therapeutic ultrasound pulses as well as the improved anticancer effects under hypoxic conditions. The findings suggest that the proposed oxygen and LLL12 loaded SRMs provide a promising drug delivery strategy for more effective treatment of hypoxic cancer cells.
Collapse
Affiliation(s)
- Jinshun Xu
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Shuai Yuan
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Jilai Tian
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Kyle A. Martin
- Department of Mechanical Engineering, The Ohio State University, Columbus, OH, 43210 USA
| | - Jinhua Song
- Division of Medicinal Chemistry and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210 USA
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210 USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jiayuh Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Ting Si
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Ronald X. Xu
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
41
|
Luo T, Sun J, Zhu S, He J, Hao L, Xiao L, Zhu Y, Wang Q, Pan X, Wang Z, Chang S. Ultrasound-mediated destruction of oxygen and paclitaxel loaded dual-targeting microbubbles for intraperitoneal treatment of ovarian cancer xenografts. Cancer Lett 2016; 391:1-11. [PMID: 28043912 DOI: 10.1016/j.canlet.2016.12.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022]
Abstract
Folate receptor (FR) is overexpressed in many epithelial cancers and tumor-associated macrophages (TAMs), which enable it to function as an appropriate target for cancer treatment. We have successfully synthesized multifunctional folate-targeted and oxygen/paclitaxel loaded microbubbles (TOPLMBs) for ultrasound (US) mediated delivery for combination therapy in an intraperitoneal ovarian cancer xenograft model. The TOPLMBs target both ovarian cancer cells and TAMs and provide a promising drug delivery strategy for the combination treatment of ovarian cancer and tumor microenvironment. Microscopic imaging and flow cytometric analysis showed that TOPLMBs significantly penetrated into ovarian cancer cells and tumor-associated macrophages (TAMs) within tumor ascites fluid and the tumor nodules. Immunohistochemical analyses of dissected tumor tissue confirmed the increased tumor apoptosis, the reduced expression of vascular endothelial growth factor (VEGF) and microvascular density (MVD), and the reduced expression of CD68 after treatment (P < 0.05). Our experiment results suggest that intraperitoneal injection of dual-targeting TOPLMBs followed by US mediation provide a promising drug delivery strategy for combination treatment of ovarian cancer and tumor microenvironment with the therapeutic outcome superior to that of conventional therapeutic options.
Collapse
Affiliation(s)
- Tingting Luo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China; Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shenyin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Juan He
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Lan Hao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linlin Xiao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qianqian Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xin Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|