1
|
Li X, He Y, Wang Y, Lin K, Lin X. CHARMM36 All-Atom Gas Model for Lipid Nanobubble Simulation. J Chem Inf Model 2024; 64:7503-7512. [PMID: 39262130 DOI: 10.1021/acs.jcim.4c01027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Lipid nanobubbles with different gas cores may integrate the biocompatibility of lipids, powerful physicochemical properties of nanobubbles, and therapeutic effects of gas molecules, which thus promote enormous biomedical applications such as ultrasound molecular imaging, gene/drug delivery, and gas therapy. In order for further more precise applications, the exact molecular mechanisms for the interactions between lipid nanobubbles and biological systems should be studied. Molecular dynamics (MD) simulation provides a powerful computational tool for this purpose. However, previous state-of-the-art MD simulations of free gas nanobubble/lipid nanobubble employed the vacuum as their gas cores, which is not suitable for studying the interactions between functional lipid nanobubbles and biological systems and revealing the biological roles of gas molecules. Hence, in this work, we developed and optimized the CHARMM36 all-atom gas parameters for six gases including N2, O2, H2, CO, CO2, and SO2, which accurately reproduced the gas density at different pressures as well as the spontaneous formation of gas nanobubbles. Subsequent applications of these gas parameters for lipid nanobubble simulations also reproduced the self-assembly process of the lipid nanobubble. We further developed a Python script to generate all-atom lipid nanobubble simulation systems, which was proven to be efficient for all-atom MD simulations of lipid nanobubbles and to be able to capture the exact dynamics of gas molecules at the gas-lipid and lipid-water interfaces of the lipid nanobubble. In summary, the all-atom gas models proposed in this work are suitable for simulating free gas nanobubbles and lipid nanobubbles, which are supposed to overcome the shortcomings of previous state-of-the-art MD simulations with the vacuum replacing the gas core and play key roles in revealing the molecular-level interactions between lipid nanobubbles and biological systems.
Collapse
Affiliation(s)
- Xiu Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yuan He
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yuxuan Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Kaidong Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
2
|
Van Court B, Ciccaglione M, Neupert B, Knitz MW, Maroney SP, Nguyen D, Abdelazeem KNM, Exner AA, Saviola AJ, Benninger RKP, Karam SD. Heterogeneous Kinetics of Nanobubble Ultrasound Contrast Agent and Angiogenic Signaling in Head and Neck Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614362. [PMID: 39386624 PMCID: PMC11463497 DOI: 10.1101/2024.09.22.614362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Recently developed nanobubble ultrasound contrast agents are a promising tool for imaging and drug delivery in tumors. To better understand their unusual kinetics, we implemented a novel pixel clustering analysis, which provides unique information by accounting for spatial heterogeneity. By combining ultrasound results with proteomics of the imaged tumors, we show that this analysis is highly predictive of protein expression and that specific types of nanobubble time-intensity curve are associated with upregulation of different metabolic pathways. We applied this method to study the effects of two proteins, EphB4 and ephrinB2, which control tumor angiogenesis through bidirectional juxtacrine signaling, in mouse models of head and neck cancer. We show that ephrinB2 expression by endothelial cells and EphB4 expression by cancer cells have similar effects on tumor vasculature, despite sometimes opposite effects on tumor growth. This implicates a cancer-cell-intrinsic effect of EphB4 forward signaling and not angiogenesis in EphB4's action as a tumor suppressor.
Collapse
|
3
|
Na L, Fan F. Advances in nanobubbles for cancer theranostics: Delivery, imaging and therapy. Biochem Pharmacol 2024; 226:116341. [PMID: 38848778 DOI: 10.1016/j.bcp.2024.116341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Maximizing treatment efficacy and forecasting patient prognosis in cancer necessitates the strategic use of targeted therapy, coupled with the prompt precise detection of malignant tumors. Theutilizationof gaseous systems as an adaptable platform for creating nanobubbles (NBs) has garnered significant attention as theranostics, which involve combining contrast chemicals typically used for imaging with pharmaceuticals to diagnose and treattumorssynergistically in apersonalizedmanner for each patient. This review specifically examines the utilization of oxygen NBsplatforms as a theranostic weapon in the field of oncology. We thoroughly examine the key factors that impact the effectiveness of NBs preparations and the consequences of these treatment methods. This review extensively examines recent advancements in composition schemes, advanced developments in pre-clinical phases, and other groundbreaking inventions in the area of NBs. Moreover, this review offers a thorough examination of the optimistic future possibilities, addressing prospective methods for improvement and incorporation into widely accepted therapeutic practices. As we explore the ever-changing field of cancer theranostics, the incorporation of oxygen NBs appears as a promising development, providing new opportunities for precision medicine and marking a revolutionary age in cancer research and therapy.
Collapse
Affiliation(s)
- Liu Na
- Ultrasound Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Fan Fan
- School of Automation, Xi'an University of Posts and Telecommunications, Xi'an 710121, China.
| |
Collapse
|
4
|
Wegierak D, Cooley MB, Perera R, Wulftange WJ, Gurkan UA, Kolios MC, Exner AA. Decorrelation Time Mapping as an Analysis Tool for Nanobubble-Based Contrast Enhanced Ultrasound Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2370-2380. [PMID: 38329864 PMCID: PMC11234354 DOI: 10.1109/tmi.2024.3364076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Nanobubbles (NBs; ~100-500 nm diameter) are preclinical ultrasound (US) contrast agents that expand applications of contrast enhanced US (CEUS). Due to their sub-micron size, high particle density, and deformable shell, NBs in pathological states of heightened vascular permeability (e.g. in tumors) extravasate, enabling applications not possible with microbubbles (~1000-10,000 nm diameter). A method that can separate intravascular versus extravascular NB signal is needed as an imaging biomarker for improved tumor detection. We present a demonstration of decorrelation time (DT) mapping for enhanced tumor NB-CEUS imaging. In vitro models validated the sensitivity of DT to agent motion. Prostate cancer mouse models validated in vivo imaging potential and sensitivity to cancerous tissue. Our findings show that DT is inversely related to NB motion, offering enhanced detail of NB dynamics in tumors, and highlighting the heterogeneity of the tumor environment. Average DT was high in tumor regions (~9 s) compared to surrounding normal tissue (~1 s) with higher sensitivity to tumor tissue compared to other mapping techniques. Molecular NB targeting to tumors further extended DT (11 s) over non-targeted NBs (6 s), demonstrating sensitivity to NB adherence. From DT mapping of in vivo NB dynamics we demonstrate the heterogeneity of tumor tissue while quantifying extravascular NB kinetics and delineating intra-tumoral vasculature. This new NB-CEUS-based biomarker can be powerful in molecular US imaging, with improved sensitivity and specificity to diseased tissue and potential for use as an estimator of vascular permeability and the enhanced permeability and retention (EPR) effect in tumors.
Collapse
|
5
|
Wang J, Wang Y, Zhong L, Yan F, Zheng H. Nanoscale contrast agents: A promising tool for ultrasound imaging and therapy. Adv Drug Deliv Rev 2024; 207:115200. [PMID: 38364906 DOI: 10.1016/j.addr.2024.115200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/31/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Nanoscale contrast agents have emerged as a versatile platform in the field of biomedical research, offering great potential for ultrasound imaging and therapy. Various kinds of nanoscale contrast agents have been extensively investigated in preclinical experiments to satisfy diverse biomedical applications. This paper provides a comprehensive review of the structure and composition of various nanoscale contrast agents, as well as their preparation and functionalization, encompassing both chemosynthetic and biosynthetic strategies. Subsequently, we delve into recent advances in the utilization of nanoscale contrast agents in various biomedical applications, including ultrasound molecular imaging, ultrasound-mediated drug delivery, and cell acoustic manipulation. Finally, the challenges and prospects of nanoscale contrast agents are also discussed to promote the development of this innovative nanoplatform in the field of biomedicine.
Collapse
Affiliation(s)
- Jieqiong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 201206, China
| | - Yuanyuan Wang
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lin Zhong
- School of public health, Nanchang University, Nanchang, Jiangxi, 330019, China
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Hairong Zheng
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Chen C, Perera R, Mischi M, Kolios M, Exner A, Turco S. Quantification of extravasation and binding of PSMA-targeted nanobubbles by modelling the second-wave phenomenon. Mol Imaging Biol 2024; 26:253-263. [PMID: 38151581 DOI: 10.1007/s11307-023-01891-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023]
Abstract
PURPOSE With about ten-fold smaller diameter than MBs, nanobubbles (NBs) were developed as new-generation ultrasound contrast agents (UCA) able to extravasate and target specific receptors expressed on extravascular cancer cells, such as the prostate-specific membrane antigen (PSMA). It has been shown that PSMA-targeted NBs (PSMA-NBs) can bind to specific prostate cancer (PCa) cells and exhibit a prolonged retention effect (PRE), observable by NB-based CEUS (NB-CEUS). However, previous analyses of PRE were mainly limited to the semi-quantitative assessment of the time-intensity curve (TIC) in an entire tumor ROI, possibly losing information on tumor spatial heterogeneity and local characteristics. When analyzing the pixel-level TICs of free NB-based CEUS, we observed a unique second-wave phenomenon: The first pass of the NB wave (bolus) is usually accompanied by a second wave in the time range of 3 to 15 min after the bolus injection. Such a phenomenon was shown to be potentially valuable in supporting the diagnostics of cancerous lesions. PROCEDURES Seven male athymic nude mice were included and implanted with a tumor expressing PSMA (PSMA+) and tumors not expressing PSMA (PSMA-) on two flanks. Using either free NBs or PSMA-NBs, the characteristics of pixel-level TICs were estimated by a specialized model accounting for the two-wave phenomenon, compared with a conventional model describing only one wave. The estimated parameters by the two models were presented as parametric maps to visualize the PRE of PSMA-NBs in a dual-tumor mouse model. The effectiveness of the two models were also assessed by comparing the estimated parameters in the PSMA+ and PSMA- tumors through Mann-Whitney U test and quartile difference. RESULTS Two parameters, the peak time and residual factor of the second wave, by the second-wave model were significantly different between PSMA+ and PSMA- tumors when using PSMA-NBs. Compared with the TICs of free NBs, TICs of PSMA-NBs present higher peak intensity and a more delayed second wave, especially in the PSMA+ tumor. CONCLUSIONS The estimation of parametric maps allows the estimation and visualization of specific binding of PSMA-NBs in PCa. The incorporation of the second-wave phenomenon enrich our understanding of NB kinetics in vivo and can possibly contribute to improved diagnostics of PCa in the future.
Collapse
Affiliation(s)
- Chuan Chen
- Southeast University, Nanjing, People's Republic of China.
- Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | - Massimo Mischi
- Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | - Agata Exner
- Case Western Reserve University, Cleveland, OH, USA
| | - Simona Turco
- Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
7
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Nittayacharn P, Abenojar E, Cooley MB, Berg FM, Counil C, Sojahrood AJ, Khan MS, Yang C, Berndl E, Golczak M, Kolios MC, Exner AA. Efficient ultrasound-mediated drug delivery to orthotopic liver tumors - Direct comparison of doxorubicin-loaded nanobubbles and microbubbles. J Control Release 2024; 367:135-147. [PMID: 38237687 DOI: 10.1016/j.jconrel.2024.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
Liver metastasis is a major obstacle in treating aggressive cancers, and current therapeutic options often prove insufficient. To overcome these challenges, there has been growing interest in ultrasound-mediated drug delivery using lipid-shelled microbubbles (MBs) and nanobubbles (NBs) as promising strategies for enhancing drug delivery to tumors. Our previous work demonstrated the potential of Doxorubicin-loaded C3F8 NBs (hDox-NB, 280 ± 123 nm) in improving cancer treatment in vitro using low-frequency unfocused therapeutic ultrasound (TUS). In this study, we investigated the pharmacokinetics and biodistribution of sonicated hDox-NBs in orthotopic rat liver tumors. We compared their delivery and therapeutic efficiency with size-isolated MBs (hDox-MB, 1104 ± 373 nm) made from identical shell material and core gas. Results showed a similar accumulation of hDox in tumors treated with hDox-MBs and unfocused therapeutic ultrasound (hDox-MB + TUS) and hDox-NB + TUS. However, significantly increased apoptotic cell death in the tumor and fewer off-target apoptotic cells in the normal liver were found upon the treatment with hDox-NB + TUS. The tumor-to-liver apoptotic ratio was elevated 9.4-fold following treatment with hDox-NB + TUS compared to hDox-MB + TUS, suggesting that the therapeutic efficacy and specificity are significantly increased when using hDox-NB + TUS. These findings highlight the potential of this approach as a viable treatment modality for liver tumors. By elucidating the behavior of drug-loaded bubbles in vivo, we aim to contribute to developing more effective liver cancer treatments that could ultimately improve patient outcomes and decrease off-target side effects.
Collapse
Affiliation(s)
- Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Puttamonthon, Nakorn Pathom, Thailand
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Felipe M Berg
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Claire Counil
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Amin Jafari Sojahrood
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Muhammad Saad Khan
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Celina Yang
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Elizabeth Berndl
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital, a site of Unity Health Toronto and Toronto Metropolitan University, Toronto, Canada
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
9
|
Paknahad AA, Zalloum IO, Karshafian R, Kolios MC, Tsai SSH. High throughput microfluidic nanobubble generation by microporous membrane integration and controlled bubble shrinkage. J Colloid Interface Sci 2024; 653:277-284. [PMID: 37716307 DOI: 10.1016/j.jcis.2023.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/30/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
Microfluidics has recently been proposed as a viable method for producing bulk nanobubbles for use in various applications. The portability, compact size, and capacity to precisely control fluids on a small scale are a few of the benefits of microfluidics that may be exploited to create customized bulk nanobubbles. However, despite the potential of microfluidic nanobubble generation, low throughput and limited nanobubble concentration remain challenging for microfluidics. Here, we integrate a microporous silicon membrane into a polydimethylsiloxane microfluidic chip to generate bulk nanobubbles in the 100-140 nm diameter range with a concentration of up to 108 mL-1. We investigate the nanobubble size and morphology using several characterisation techniques, including transmission electron microscopy, resonance mass measurement, dynamic light scattering, and the Tyndall effect. This new nanobubble generation technique can increase nanobubble concentration by ∼ 23 times compared to earlier microfluidic nanobubble generation platforms, which should increase the feasibility of translation to medical applications.
Collapse
Affiliation(s)
- Ali A Paknahad
- Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Intesar O Zalloum
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada; Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Raffi Karshafian
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada; Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Michael C Kolios
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada; Department of Physics, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada.
| | - Scott S H Tsai
- Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada; Graduate Program in Biomedical Engineering, Toronto Metropolitan University, Toronto M5B 2K3, Canada.
| |
Collapse
|
10
|
Shah R, Phatak N, Choudhary A, Gadewar S, Ajazuddin, Bhattacharya S. Exploring the Theranostic Applications and Prospects of Nanobubbles. Curr Pharm Biotechnol 2024; 25:1167-1181. [PMID: 37861011 DOI: 10.2174/0113892010248189231010085827] [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: 02/08/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023]
Abstract
Anticancer medications as well as additional therapeutic compounds, have poor clinical effectiveness due to their diverse distribution, non-selectivity for malignant cells, and undesirable off-target side effects. As a result, ultrasound-based targeted delivery of therapeutic compounds carried in sophisticated nanocarriers has grown in favor of cancer therapy and control. Nanobubbles are nanoscale bubbles that exhibit unique physiochemical properties in both their inner core and outer shell. Manufacturing nanobubbles primarily aims to enhance therapeutic agents' bioavailability, stability, and targeted delivery. The small size of nanobubbles allows for their extravasation from blood vessels into surrounding tissues and site-specific release through ultrasound targeting. Ultrasound technology is widely utilized for therapy due to its speed, safety, and cost-effectiveness, and micro/nanobubbles, as ultrasound contrast agents, have numerous potential applications in disease treatment. Thus, combining ultrasound applications with NBs has recently demonstrated increased localization of anticancer molecules in tumor tissues with triggered release behavior. Consequently, an effective therapeutic concentration of drugs/genes is achieved in target tumor tissues with ultimately increased therapeutic efficacy and minimal side effects on other non-cancerous tissues. This paper provides a brief overview of the production processes for nanobubbles, along with their key characteristics and potential therapeutic uses.
Collapse
Affiliation(s)
- Rahul Shah
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Niraj Phatak
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Ashok Choudhary
- Department of Quality Assurance, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Sakshi Gadewar
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences & Research, Khoka-Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| |
Collapse
|
11
|
Ling B, Ko JH, Stordy B, Zhang Y, Didden TF, Malounda D, Swift MB, Chan WCW, Shapiro MG. Gas Vesicle-Blood Interactions Enhance Ultrasound Imaging Contrast. NANO LETTERS 2023; 23:10748-10757. [PMID: 37983479 PMCID: PMC10722532 DOI: 10.1021/acs.nanolett.3c02780] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Gas vesicles (GVs) are genetically encoded, air-filled protein nanostructures of broad interest for biomedical research and clinical applications, acting as imaging and therapeutic agents for ultrasound, magnetic resonance, and optical techniques. However, the biomedical applications of GVs as systemically injectable nanomaterials have been hindered by a lack of understanding of GVs' interactions with blood components, which can significantly impact in vivo behavior. Here, we investigate the dynamics of GVs in the bloodstream using a combination of ultrasound and optical imaging, surface functionalization, flow cytometry, and mass spectrometry. We find that erythrocytes and serum proteins bind to GVs and shape their acoustic response, circulation time, and immunogenicity. We show that by modifying the GV surface we can alter these interactions and thereby modify GVs' in vivo performance. These results provide critical insights for the development of GVs as agents for nanomedicine.
Collapse
Affiliation(s)
- Bill Ling
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Jeong Hoon Ko
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Benjamin Stordy
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Terrence
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S
3E1, Canada
| | - Yuwei Zhang
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Terrence
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S
3E1, Canada
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Tighe F. Didden
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Dina Malounda
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Margaret B. Swift
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
| | - Warren C. W. Chan
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, ON M5S 3G9, Canada
- Terrence
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON M5S
3E1, Canada
- Department
of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Mikhail G. Shapiro
- Division
of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, California 91125, United States
- Division
of Engineering and Applied Science, California
Institute of Technology, Pasadena, California 91125, United States
- Howard Hughes
Medical Institute, California Institute
of Technology, Pasadena, California 91125, United States
| |
Collapse
|
12
|
Zhang Y, Dang Y, Huang M, Ma Y, Zhang D, Wang X. Development of bioactive and ultrasound-responsive microdroplets for preventing ovariectomy (OVX)-induced osteoporosis. J Mater Chem B 2023; 11:11344-11356. [PMID: 37990947 DOI: 10.1039/d3tb01726e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
As a common bone disease in the elderly population, osteoporosis-related bone loss and bone structure deterioration represent a major public health problem. Therapeutic strategies targeting excessive osteoclast formation are frequently used for osteoporosis treatment; however, potential side effects have been recorded. Here, we have developed a novel therapeutic strategy using microdroplets (MDs) encapsulated with NFATc1-siRNA and investigated the role of bioactive MDs-NFATc1 biocompatibility in RAW 264.7 macrophages and human mesenchymal stem cells (hBMSCs), respectively. Its role in regulating osteoclast differentiation and formation was also investigated in vitro. We first fabricated MDs with spherical morphology along with a well-defined core-shell structure. The ultrasound-responsive study demonstrated time-dependent responsive structural changes following ultrasound stimulation. The internalization study into unstimulated macrophages, inflammatory macrophages, and hBMSCs indicated good delivery efficiency. Furthermore, the results from the MTT assay, the live/dead assay, and the cellular morphological analysis further indicated good biocompatibility of our bioactive MDs-NFATc1. Following MDs-NFATc1 treatment, the number of osteoclasts was greatly reduced, indicating their inhibitory effect on osteoclastogenesis and osteoclast formation. Subsequently, osteoporotic rats that underwent ovariectomy (OVX) were used for the in vivo studies. The rats treated with MDs-NFATc1 exhibited significant resistance to bone loss induced by OVX. In conclusion, our results demonstrate that MDs-NFATc1 could become an important regulator in osteoclast differentiation and functions, thus having potential applications in osteoclast-related bone diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Hygiene Toxicology, Zunyi Medical University, Zunyi, 563000 Guizhou, China
- Key Laboratory of Maternal & Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Yi Dang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou, China.
| | - Maodi Huang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou, China.
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou, China.
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou, China.
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003 Guizhou, China.
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, 563000 Guizhou, China
| |
Collapse
|
13
|
Cooley MB, Wegierak D, Perera R, Abenojar EC, Nittayacharn PA, Berg FM, Kim Y, Kolios MC, Exner AA. Assessing Tumor Microenvironment Characteristics and Stratifying EPR with a Nanobubble Companion Nanoparticle via Contrast-Enhanced Ultrasound Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567934. [PMID: 38045236 PMCID: PMC10690218 DOI: 10.1101/2023.11.20.567934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The tumor microenvironment is characterized by dysfunctional endothelial cells, resulting in heightened vascular permeability. Many nanoparticle-based drug delivery systems attempt to use this enhanced permeability combined with impaired lymphatic drainage (a concept known as the 'enhanced permeability and retention effect' or EPR effect) as the primary strategy for drug delivery, but this has not proven to be as clinically effective as anticipated. The specific mechanisms behind the inconsistent clinical outcomes of nanotherapeutics have not been clearly articulated, and the field has been hampered by a lack of accessible tools to study EPR-associated phenomena in clinically relevant scenarios. While medical imaging has tremendous potential to contribute to this area, it has not been broadly explored. This work examines, for the first time, the use of multiparametric dynamic contrast-enhanced ultrasound (CEUS) with a novel nanoscale contrast agent to examine tumor microenvironment characteristics noninvasively and in real-time. We demonstrate that CEUS imaging can: (1) evaluate tumor microenvironment features and (2) be used to help predict the distribution of doxorubicin-loaded liposomes in the tumor parenchyma. CEUS using nanobubbles (NBs) was carried out in two tumor types of high (LS174T) and low (U87) vascular permeability, and time-intensity curve (TIC) parameters were evaluated in both models prior to injection of doxorubicin liposomes. Consistently, LS174T tumors showed significantly different TIC parameters, including area under the rising curve (2.7x), time to peak intensity (1.9x) and decorrelation time (DT, 1.9x) compared to U87 tumors. Importantly, the DT parameter successfully predicted tumoral nanoparticle distribution (r = 0.86 ± 0.13). Ultimately, substantial differences in NB-CEUS generated parameters between LS174T and U87 tumors suggest that this method may be useful in determining tumor vascular permeability and could be used as a biomarker for identifying tumor characteristics and predicting sensitivity to nanoparticle-based therapies. These findings could ultimately be applied to predicting treatment efficacy and to evaluating EPR in other diseases with pathologically permeable vasculature.
Collapse
|
14
|
Nittayacharn P, Abenojar E, Cooley M, Berg F, Counil C, Sojahrood AJ, Khan MS, Yang C, Berndl E, Golczak M, Kolios MC, Exner AA. Efficient ultrasound-mediated drug delivery to orthotopic liver tumors - Direct comparison of doxorubicin-loaded nanobubbles and microbubbles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555196. [PMID: 37732235 PMCID: PMC10508722 DOI: 10.1101/2023.09.01.555196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Liver metastasis is a major obstacle in treating aggressive cancers, and current therapeutic options often prove insufficient. To overcome these challenges, there has been growing interest in ultrasound-mediated drug delivery using lipid-shelled microbubbles (MBs) and nanobubbles (NBs) as promising strategies for enhancing drug delivery to tumors. Our previous work demonstrated the potential of Doxorubicin-loaded C3F8 NBs (hDox-NB, 280 ± 123 nm) in improving cancer treatment in vitro using low-frequency ultrasound. In this study, we investigated the pharmacokinetics and biodistribution of sonicated hDox-NBs in orthotopic rat liver tumors. We compared their delivery and therapeutic efficiency with size-isolated MBs (hDox-MB, 1104 ± 373 nm). Results showed a similar accumulation of hDox in tumors treated with hDox-MBs and unfocused therapeutic ultrasound (hDox-MB+TUS) and hDox-NB+TUS. However, significantly increased apoptotic cell death in the tumor and fewer off-target apoptotic cells in the normal liver were found upon the treatment with hDox-NB+TUS. The tumor-to-liver apoptotic ratio was elevated 9.4-fold following treatment with hDox-NB+TUS compared to hDox-MB+TUS, suggesting that the therapeutic efficacy and specificity are significantly increased when using hDox-NB+TUS. These findings highlight the potential of this approach as a viable treatment modality for liver tumors. By elucidating the behavior of drug-loaded bubbles in vivo, we aim to contribute to developing more effective liver cancer treatments that could ultimately improve patient outcomes and decrease off-target side effects.
Collapse
Affiliation(s)
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Michaela Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Felipe Berg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Claire Counil
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Celina Yang
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Elizabeth Berndl
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Marcin Golczak
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Michael C. Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Canada
| | - Agata A. Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
15
|
Baroni S, Argenziano M, La Cava F, Soster M, Garello F, Lembo D, Cavalli R, Terreno E. Hard-Shelled Glycol Chitosan Nanoparticles for Dual MRI/US Detection of Drug Delivery/Release: A Proof-of-Concept Study. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2227. [PMID: 37570545 PMCID: PMC10420971 DOI: 10.3390/nano13152227] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
Abstract
This paper describes a novel nanoformulation for dual MRI/US in vivo monitoring of drug delivery/release. The nanosystem was made of a perfluoropentane core coated with phospholipids stabilized by glycol chitosan crosslinked with triphosphate ions, and it was co-loaded with the prodrug prednisolone phosphate (PLP) and the structurally similar MRI agent Gd-DTPAMA-CHOL. Importantly, the in vitro release of PLP and Gd-DTPAMA-CHOL from the nanocarrier showed similar profiles, validating the potential impact of the MRI agent as an imaging reporter for the drug release. On the other hand, the nanobubbles were also detectable by US imaging both in vitro and in vivo. Therefore, the temporal evolution of both MRI and US contrast after the administration of the proposed nanosystem could report on the delivery and the release kinetics of the transported drug in a given lesion.
Collapse
Affiliation(s)
- Simona Baroni
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (S.B.); (F.L.C.); (F.G.)
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, 10125 Torino, Italy; (M.A.); (M.S.)
| | - Francesca La Cava
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (S.B.); (F.L.C.); (F.G.)
| | - Marco Soster
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, 10125 Torino, Italy; (M.A.); (M.S.)
| | - Francesca Garello
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (S.B.); (F.L.C.); (F.G.)
| | - David Lembo
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Regione Gonzole, 10, 10043 Orbassano, Italy;
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, 10125 Torino, Italy; (M.A.); (M.S.)
| | - Enzo Terreno
- Molecular and Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (S.B.); (F.L.C.); (F.G.)
| |
Collapse
|
16
|
Cooley MB, Wulftange WJ, Wegierak D, Goreke U, Abenojar EC, Gurkan UA, Exner AA. Real-time imaging of nanobubble ultrasound contrast agent flow, extravasation, and diffusion through an extracellular matrix using a microfluidic model. LAB ON A CHIP 2023; 23:3453-3466. [PMID: 37424286 DOI: 10.1039/d3lc00514c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Lipid shell-stabilized nanoparticles with a perfluorocarbon gas-core, or nanobubbles, have recently attracted attention as a new contrast agent for molecular ultrasound imaging and image-guided therapy. Due to their small size (∼275 nm diameter) and flexible shell, nanobubbles have been shown to extravasate through hyperpermeable vasculature (e.g., in tumors). However, little is known about the dynamics and depth of extravasation of intact, acoustically active nanobubbles. Accordingly, in this work, we developed a microfluidic chip with a lumen and extracellular matrix (ECM) and imaging method that allows real-time imaging and characterization of the extravasation process with high-frequency ultrasound. The microfluidic device has a lumen and is surrounded by an extracellular matrix with tunable porosity. The combination of ultrasound imaging and the microfluidic chip advantageously produces real-time images of the entire length and depth of the matrix. This captures the matrix heterogeneity, offering advantages over other imaging techniques with smaller fields of view. Results from this study show that nanobubbles diffuse through a 1.3 μm pore size (2 mg mL-1) collagen I matrix 25× faster with a penetration depth that was 0.19 mm deeper than a 3.7 μm (4 mg mL-1) matrix. In the 3.7 μm pore size matrix, nanobubbles diffused 92× faster than large nanobubbles (∼875 nm diameter). Decorrelation time analysis was successfully used to differentiate flowing and extra-luminally diffusing nanobubbles. In this work, we show for the first time that combination of an ultrasound-capable microfluidic chip and real-time imaging provided valuable insight into spatiotemporal nanoparticle movement through a heterogeneous extracellular matrix. This work could help accurately predict parameters (e.g., injection dosage) that improve translation of nanoparticles from in vitro to in vivo environments.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - William J Wulftange
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Utku Goreke
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Eric C Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Umut A Gurkan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|
17
|
Dehariya D, Eswar K, Tarafdar A, Balusamy S, Rengan AK. Recent Advances of Nanobubble-based systems in Cancer Therapeutics: A Review. BIOMEDICAL ENGINEERING ADVANCES 2023. [DOI: 10.1016/j.bea.2023.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023] Open
|
18
|
Cooley MB, Abenojar EC, Wegierak D, Sen Gupta A, Kolios MC, Exner AA. Characterization of the interaction of nanobubble ultrasound contrast agents with human blood components. Bioact Mater 2023; 19:642-652. [PMID: 35600972 PMCID: PMC9109121 DOI: 10.1016/j.bioactmat.2022.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/24/2022] [Accepted: 05/01/2022] [Indexed: 02/06/2023] Open
Abstract
Nanoscale ultrasound contrast agents, or nanobubbles, are being explored in preclinical applications ranging from vascular and cardiac imaging to targeted drug delivery in cancer. These sub-micron particles are approximately 10x smaller than clinically available microbubbles. This allows them to effectively traverse compromised physiological barriers and circulate for extended periods of time. While various aspects of nanobubble behavior have been previously examined, their behavior in human whole blood has not yet been explored. Accordingly, herein we examined, for the first time, the short and long-term effects of blood components on nanobubble acoustic response. We observed differences in the kinetics of backscatter from nanobubble suspensions in whole blood compared to bubbles in phosphate buffered saline (PBS), plasma, or red blood cell solutions (RBCs). Specifically, after introducing nanobubbles to fresh human whole blood, signal enhancement, or the magnitude of nonlinear ultrasound signal, gradually increased by 22.8 ± 13.1% throughout our experiment, with peak intensity reached within 145 s. In contrast, nanobubbles in PBS had a stable signal with negligible change in intensity (−1.7 ± 3.2%) over 8 min. Under the same conditions, microbubbles made with the same lipid formulation showed a −56.8 ± 6.1% decrease in enhancement in whole blood. Subsequent confocal, fluorescent, and scanning electron microscopy analysis revealed attachment of the nanobubbles to the surface of RBCs, suggesting that direct interactions, or hitchhiking, of nanobubbles on RBCs in the presence of plasma may be a possible mechanism for the observed effects. This phenomenon could be key to extending nanobubble circulation time and has broad implications in drug delivery, where RBC interaction with nanoparticles could be exploited to improve delivery efficiency. Show distinct signal enhancement curve of nanobubbles (NB) in human whole blood Compare signal enhancement over time in blood with NBs and microbubbles (MBs) Examine the effect of blood components on NB and MB movement over time Demonstrate, with microscopy, that NBs localize to the RBC surface in whole blood
Collapse
|
19
|
Su C, Ren X, Yang F, Li B, Wu H, Li H, Nie F. Ultrasound-sensitive siRNA-loaded nanobubbles fabrication and antagonism in drug resistance for NSCLC. Drug Deliv 2022; 29:99-110. [PMID: 34964410 PMCID: PMC8725955 DOI: 10.1080/10717544.2021.2021321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 10/26/2022] Open
Abstract
Due to the lack of safe, effective, and gene-targeted delivery technology. In this study, we have prepared nanobubbles loaded PDLIM5 siRNA (PDLIM5siRNA-NBs) to investigate the transfection efficiency and their antagonism in drug resistance in combination with ultrasound irradiation for non-small-cell lung cancer (NSCLC). Research results show that the PDLIM5 siRNA are effectively bound to the shell of NBs with a mean diameter of 191.6 ± 0.50 nm and a Zeta potential of 11.8 ± 0.68 mV. And the ultrasonic imaging indicated that the PDLIM5 siRNA NBs maintain the same signals as the microbubbles (SonoVue). Under the optimized conditions of 0.5 W/m2 ultrasound intensity and 1 min irradiation duration, the highest transfection efficiency of PC9GR cells was 90.23 ± 1.45%, which resulted in the inhibition of PDLIM5 mRNA and protein expression. More importantly, the anti-tumor effect of fabricated PDLIM5siRNA-NBs with the help of ultrasound irradiation has been demonstrated to significantly inhibit tumor cell growth and promote apoptosis. Therefore, NBs carrying PDLIM5siRNA may have the potential to act as gene vectors combined with ultrasound irradiation to antagonize drug resistance for NSCLC.
Collapse
Affiliation(s)
- Chunhong Su
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, China
- Department of Pain, Lanzhou University Second Hospital, Lanzhou, China
| | - XiaoJun Ren
- Department of Pediatric Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Bin Li
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Hao Wu
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, China
| | - Hui Li
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, China
| | - Fang Nie
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
20
|
Batchelor DB, Armistead FJ, Ingram N, Peyman SA, McLaughlan JR, Coletta PL, Evans SD. The Influence of Nanobubble Size and Stability on Ultrasound Enhanced Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:13943-13954. [PMID: 36322191 PMCID: PMC9671049 DOI: 10.1021/acs.langmuir.2c02303] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/17/2022] [Indexed: 06/16/2023]
Abstract
Lipid-shelled nanobubbles (NBs) are emerging as potential dual diagnostic and therapeutic agents. Similar to their micron-scale counterparts, microbubbles (1-10 μm), they can act as ultrasound contrast agents as well as locally enhance therapeutic uptake. Recently, it has been shown that the reduced size of NBs (<1 μm) promotes increased uptake and accumulation in tumor interstitial space, which can enhance their diagnostic and therapeutic performance. However, accurate characterization of NB size and concentration is challenging and may limit their translation into clinical use. Their submicron nature limits accuracy of conventional microscopy techniques, while common light scattering techniques fail to distinguish between subpopulations present in NB samples (i.e., bubbles and liposomes). Due to the difficulty in the characterization of NBs, relatively little is known about the influence of size on their therapeutic performance. In this study, we describe a novel method of using a commercially available nanoparticle tracking analysis system, to distinguish between NBs and liposomes based on their differing optical properties. We used this technique to characterize three NB populations of varying size, isolated via centrifugation, and subsequently used this to assess their potential for enhancing localized delivery. Confocal fluorescence microscopy and image analysis were used to quantify the ultrasound enhanced uptake of fluorescent dextran into live colorectal cancer cells. Our results showed that the amount of localized uptake did not follow the expected trends, in which larger NB populations out-perform smaller NBs, at matched concentration. To understand this observed behavior, the stability of each NB population was assessed. It was found that dilution of the NB samples from their stock concentration influences their stability, and it is hypothesized that both the total free lipid and interbubble distance play a role in NB lifetime, in agreement with previously proposed theories and models.
Collapse
Affiliation(s)
- Damien
V. B. Batchelor
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - Fern J. Armistead
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - Nicola Ingram
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, LeedsLS9 7TF, United Kingdom
- Faculty
of Electronic and Electrical Engineering, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - Sally A. Peyman
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - James R. McLaughlan
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, LeedsLS9 7TF, United Kingdom
- Faculty
of Electronic and Electrical Engineering, University of Leeds, LeedsLS2 9JT, United Kingdom
| | - P. Louise Coletta
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, LeedsLS9 7TF, United Kingdom
| | - Stephen D. Evans
- Molecular
and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, LeedsLS2 9JT, United Kingdom
| |
Collapse
|
21
|
Xie F, Yan L, Li YM, Lan Y, Xiao J, Zhang MB, Jin Z, Zhang Y, Tian XQ, Zhu YQ, Li ZP, Luo YK. Targeting Diagnosis of High-Risk Papillary Thyroid Carcinoma Using Ultrasound Contrast Agent With the BRAF V600E Mutation: An Experimental Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:2789-2802. [PMID: 35229905 DOI: 10.1002/jum.15967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/14/2022] [Accepted: 02/02/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE High-risk papillary thyroid carcinoma (PTC) patients with BRAF mutation have lymph node and distant metastases and poor prognosis. Therefore, this study aims to develop a targeted ultrasound contrast agent for the BRAFV600E mutation to screen high-risk PTC at early stage. METHODS The targeted lipid nanobubbles carrying BRAFV600E antibody were prepared using thin film hydration-sonication and avidin-biotin binding methods. The physicochemical properties and stability of the targeted nanobubbles were detected by transmission electron microscopy, atomic force microscopy, and confocal laser scanning microscopy. The target binding abilities of the targeted nanobubbles in the PTC cells (B-CPAP) overexpressed mutant BRAFV600E were evaluated by immunofluorescence staining, quantitative real-time polymerase chain reaction, western blot, and fluorescence microscopy. After PTC tumor models overexpressed mutant BRAFV600E were established, the enhanced images of targeted lipid nanobubbles and untargeted lipid nanobubbles on PTC tumors in nude mice were observed using contrast-enhanced ultrasound imaging. RESULTS The targeted lipid nanobubbles revealed uniform, round morphology, and good stability with a nanoscale size. Besides, BRAFV600E monoclonal antibody was observed to be combined on the surface of lipid nanobubbles. Furthermore, the targeted nanobubbles had a good targeting diagnosis ability in PTC cells with BRAFV600E overexpression. Moreover, the targeted nanobubbles had better ultrasound enhancement and peak intensity of the time-intensity curve (P < .001) in PTC tumors with BRAFV600E overexpression as compared to the untargeted lipid nanobubbles. CONCLUSION The targeted lipid nanobubbles carrying BRAFV600E antibody could be regarded as a potential targeted ultrasound contrast agent for the diagnosis of high-risk PTC.
Collapse
Affiliation(s)
- Fang Xie
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Lin Yan
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Yi-Ming Li
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Yu Lan
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Jing Xiao
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Ming-Bo Zhang
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Zhuang Jin
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Ying Zhang
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Xiao-Qi Tian
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Ya-Qiong Zhu
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| | - Zhi-Ping Li
- Pharmacology Research Department, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yu-Kun Luo
- Department of Ultrasound, The First Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
22
|
Fundamentals and applications of nanobubbles: A review. Chem Eng Res Des 2022. [DOI: 10.1016/j.cherd.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
23
|
Ultrastable shelled PFC nanobubbles: A platform for ultrasound-assisted diagnostics, and therapy. NANOMEDICINE: NANOTECHNOLOGY, BIOLOGY AND MEDICINE 2022; 46:102611. [DOI: 10.1016/j.nano.2022.102611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/06/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022]
|
24
|
Chen C, Perera R, Kolios MC, Wijkstra H, Mischi M, Exner AA, Turco S. Pharmacokinetic modeling of PSMA-targeted nanobubbles for quantification of extravasation and binding in mice models of prostate cancer. Med Phys 2022; 49:6547-6559. [PMID: 36049109 PMCID: PMC9588563 DOI: 10.1002/mp.15962] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 08/19/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Contrast-enhanced ultrasound (CEUS) by injection of microbubbles (MBs) has shown promise as a cost-effective imaging modality for prostate cancer (PCa) detection. More recently, nanobubbles (NBs) have been proposed as novel ultrasound contrast agents. Unlike MBs, which are intravascular ultrasound contrast agents, the smaller diameter of NBs allows them to cross the vessel wall and target specific receptors on cancer cells such as the prostate-specific membrane antigen (PSMA). It has been demonstrated that PSMA-targeted NBs can bind to the receptors of PCa cells and show a prolonged retention effect in dual-tumor mice models. However, the analysis of the prolonged retention effect has so far been limited to qualitative or semi-quantitative approaches. METHODS This work introduces two pharmacokinetics models for quantitative analysis of time-intensity curves (TICs) obtained from the CEUS loops. The first model is based on describing the vascular input by the modified local density random walk (mLDRW) model and independently interprets TICs from each tumor lesion. Differently, the second model is based on the reference-tissue model, previously proposed in the context of nuclear imaging, and describes the binding kinetics of an indicator in a target tissue by using a reference tissue where binding does not occur. RESULTS Our results show that four estimated parameters, β,β / λ $\beta /\lambda $ ,β + / β - ${\beta }_ + /{\beta }_ - $ , for the mLDRW-input model, and γ for the reference-based model, were significantly different (p-value <0.05) between free NBs and PSMA-NBs. These parameters estimated by the two models demonstrate different behaviors between PSMA-targeted and free NBs. CONCLUSIONS These promising results encourage further quantitative analysis of targeted NBs for improved cancer diagnostics and characterization.
Collapse
Affiliation(s)
- Chuan Chen
- Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Reshani Perera
- Case Western Reserve University, Cleveland, Ohio, United States
| | | | - Hessel Wijkstra
- Eindhoven University of Technology, Eindhoven, the Netherlands
- Universtity Medical Center Amsterdam, Amsterdam, the Netherlands
| | - Massimo Mischi
- Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Agata A. Exner
- Case Western Reserve University, Cleveland, Ohio, United States
| | - Simona Turco
- Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
25
|
Bismuth M, Katz S, Mano T, Aronovich R, Hershkovitz D, Exner AA, Ilovitsh T. Low frequency nanobubble-enhanced ultrasound mechanotherapy for noninvasive cancer surgery. NANOSCALE 2022; 14:13614-13627. [PMID: 36070492 DOI: 10.1039/d2nr01367c] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Scaling down the size of microbubble contrast agents to the nanometer level holds the promise for noninvasive cancer therapy. However, the small size of nanobubbles limits the obtained bioeffects as a result of ultrasound cavitation, when operating near the nanobubble resonance frequency. Here we show that coupled with low energy insonation at a frequency of 80 kHz, well below the resonance frequency of these agents, nanobubbles serve as noninvasive therapeutic warheads that trigger potent mechanical effects in tumors following a systemic injection. We demonstrate these capabilities in tissue mimicking phantoms, where a comparison of the acoustic response of micro- and nano-bubbles after insonation at a frequency of 250 or 80 kHz revealed that higher pressures were needed to implode the nanobubbles compared to microbubbles. Complete nanobubble destruction was achieved at a mechanical index of 2.6 for the 250 kHz insonation vs. 1.2 for the 80 kHz frequency. Thus, the 80 kHz insonation complies with safety regulations that recommend operation below a mechanical index of 1.9. In vitro in breast cancer tumor cells, the cell viability was reduced to 17.3 ± 1.7% of live cells. In vivo, in a breast cancer tumor mouse model, nanobubble tumor distribution and accumulation were evaluated by high frequency ultrasound imaging. Finally, nanobubble-mediated low frequency insonation of breast cancer tumors resulted in effective mechanical tumor ablation and tumor tissue fractionation. This approach provides a unique theranostic platform for safe, noninvasive and low energy tumor mechanotherapy.
Collapse
Affiliation(s)
- Mike Bismuth
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Sharon Katz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tamar Mano
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Ramona Aronovich
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
| | - Dov Hershkovitz
- Department of Pathology, Tel-Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 6997800, Israel
| | - Agata A Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel.
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
26
|
Ishida H, Naganuma H. New horizon of contrast-enhanced sonography in the differential diagnosis of periampullary mass: A random forest nomogram. JOURNAL OF CLINICAL ULTRASOUND : JCU 2022; 50:929-930. [PMID: 36069464 DOI: 10.1002/jcu.23259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Hideaki Ishida
- Center of Diagnostic Ultrasound, Akita Red Cross Hospital, Akita city, Japan
| | - Hiroko Naganuma
- Department of Gastroenterology, Yokote Municipal Hospital, Yokote, Japan
| |
Collapse
|
27
|
Role of Multiparametric Intestinal Ultrasound in the Evaluation of Response to Biologic Therapy in Adults with Crohn’s Disease. Diagnostics (Basel) 2022; 12:diagnostics12081991. [PMID: 36010341 PMCID: PMC9407413 DOI: 10.3390/diagnostics12081991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
Crohn’s disease is one of the two most common types of inflammatory bowel disease. Current medical therapies are based on the use of glucocorticoids, exclusive enteral nutrition, immunosuppressors such as azathioprine and methotrexate, and biological agents such as infliximab, adalimumab, vedolizumab, or ustekinumab. International guidelines suggest regular disease assessment and surveillance through objective instruments to adjust and personalize the therapy, reducing the overall rates of hospitalization and surgery. Although endoscopy represents the gold-standard for surveillance, its frequent use is strongly bordered by associated risks and costs. Consequently, alternative non-invasive tools to objectify disease activity and rule active inflammation out are emerging. Alongside laboratory exams and computed tomography or magnetic resonance enterography, intestinal ultrasonography (IUS) shows to be a valid choice to assess transmural inflammation and to detect transmural healing, defined as bowel wall thickness normalization, no hypervascularization, normal stratification, and no creeping fat. Compared to magnetic resonance imaging (MRI) or computed tomography, CT scan, IUS is cheaper and more widespread, with very similar accuracy. Furthermore, share wave elastography, color Doppler, and contrast-enhanced ultrasonography (CEUS) succeed in amplifying the capacity to determine the disease location, disease activity, and complications. This review aimed to discuss the role of standard and novel ultrasound techniques such as CEUS, SICUS, or share wave elastography in adults with Crohn’s disease, mainly for therapeutic monitoring and follow-up.
Collapse
|
28
|
The unique second wave phenomenon in contrast enhanced ultrasound imaging with nanobubbles. Sci Rep 2022; 12:13619. [PMID: 35948582 PMCID: PMC9365822 DOI: 10.1038/s41598-022-17756-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/30/2022] [Indexed: 12/19/2022] Open
Abstract
Investigation of nanobubble (NB) pharmacokinetics in contrast-enhanced ultrasound (CEUS) at the pixel level shows a unique phenomenon where the first pass of the contrast agent bolus is accompanied by a second wave. This effect has not been previously observed in CEUS with microbubbles. The objective of this study was to investigate this second-wave phenomenon and its potential clinical applications. Seven mice with a total of fourteen subcutaneously-implanted tumors were included in the experiments. After injecting a bolus of NBs, the NB-CEUS images were acquired to record the time-intensity curves (TICs) at each pixel. These TICs are fitted to a pharmacokinetic model which we designed to describe the observed second-wave phenomenon. The estimated model parameters are presented as parametric maps to visualize the characteristics of tumor lesions. Histological analysis was also conducted in one mouse to compare the molecular features of tumor tissue with the obtained parametric maps. The second-wave phenomenon is evidently shown in a series of pixel-based TICs extracted from either tumor or tissues. The value of two model parameters, the ratio of the peak intensities of the second over the first wave, and the decay rate of the wash-out process present large differences between malignant tumor and normal tissue (0.04 < Jessen-Shannon divergence < 0.08). The occurrence of a second wave is a unique phenomenon that we have observed in NB-CEUS imaging of both mouse tumor and tissue. As the characteristics of the second wave are different between tumor and tissue, this phenomenon has the potential to support the diagnosis of cancerous lesions.
Collapse
|
29
|
Kida H, Feril LB, Irie Y, Endo H, Itaka K, Tachibana K. Influence of Nanobubble Size Distribution on Ultrasound-Mediated Plasmid DNA and Messenger RNA Gene Delivery. Front Pharmacol 2022; 13:855495. [PMID: 35721213 PMCID: PMC9198282 DOI: 10.3389/fphar.2022.855495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/17/2022] [Indexed: 12/13/2022] Open
Abstract
The use of nanobubbles (NBs) for ultrasound-mediated gene therapy has recently attracted much attention. However, few studies have evaluated the effect of different NB size distribution to the efficiency of gene delivery into cells. In this study, various size of albumin stabilized sub-micron bubbles were examined in an in vitro ultrasound (1 MHz) irradiation setup in the aim to compare and optimize gene transfer efficiency. Results with pDNA showed that gene transfer efficiency in the presence of NB size of 254.7 ± 3.8 nm was 2.5 fold greater than those with 187.3 ± 4.8 nm. Similarly, carrier-free mRNA transfer efficiency increased in the same conditions. It is suggested that NB size greater than 200 nm contributed more to the delivery of genes into the cytoplasm with ultrasound. Although further experiments are needed to understand the underlying mechanism for this phenomenon, the present results offer valuable information in optimizing of NB for future ultrasound-mediate gene therapy.
Collapse
Affiliation(s)
- Hiroshi Kida
- Department of Anatomy, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Loreto B Feril
- Department of Anatomy, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yutaka Irie
- Department of Anatomy, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hitomi Endo
- Department of Anatomy, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Katsuro Tachibana
- Department of Anatomy, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
30
|
Li CH, Chang YC, Hsiao M, Chan MH. Ultrasound and Nanomedicine for Cancer-Targeted Drug Delivery: Screening, Cellular Mechanisms and Therapeutic Opportunities. Pharmaceutics 2022; 14:1282. [PMID: 35745854 PMCID: PMC9229768 DOI: 10.3390/pharmaceutics14061282] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer is a disease characterized by abnormal cell growth. According to a report published by the World Health Organization (WHO), cancer is the second leading cause of death globally, responsible for an estimated 9.6 million deaths in 2018. It should be noted that ultrasound is already widely used as a diagnostic procedure for detecting tumorigenesis. In addition, ultrasound energy can also be utilized effectively for treating cancer. By filling the interior of lipospheres with gas molecules, these particles can serve both as contrast agents for ultrasonic imaging and as delivery systems for drugs such as microbubbles and nanobubbles. Therefore, this review aims to describe the nanoparticle-assisted drug delivery system and how it can enhance image analysis and biomedicine. The formation characteristics of nanoparticles indicate that they will accumulate at the tumor site upon ultrasonic imaging, in accordance with their modification characteristics. As a result of changing the accumulation of materials, it is possible to examine the results by comparing images of other tumor cell lines. It is also possible to investigate ultrasound images for evidence of cellular effects. In combination with a precision ultrasound imaging system, drug-carrying lipospheres can precisely track tumor tissue and deliver drugs to tumor cells to enhance the ability of this nanocomposite to treat cancer.
Collapse
Affiliation(s)
- Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hsien Chan
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| |
Collapse
|
31
|
Lin C, Chen YZ, Wu B, Yang MT, Liu CQ, Zhao Y. Advances and prospects of ultrasound targeted drug delivery systems using biomaterial-modified micro/nanobubbles for tumor therapy. Curr Med Chem 2022; 29:5062-5075. [PMID: 35362371 DOI: 10.2174/0929867329666220331110315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 11/22/2022]
Abstract
The incidence of malignant tumors is rising rapidly and tends to be in the younger, which has been one of the most important factors endangering the safety of human life. Ultrasound micro/nanobubbles, as a noninvasive and highly specific antitumor strategy, can reach and destroy tumor tissue through their effects of cavitation and acoustic perforation under the guidance of ultrasound. Meanwhile, micro/nanobubbles are now used as a novel drug carrier, releasing drugs at a target region, especially on the prospects of biomaterial-modified micro/nanobubbles as a dual modality for drug delivery and therapeutic monitoring. and successful evaluation of the sonoporation mechanism(s), ultrasound parameters, drug type and dose will need to be addressed before translating this technology for clinical use. Therefore, this paper collects the literature on the experimental and clinical studies of ultrasound biomaterial-modified micro/nanobubbles therapy in vitro and in vivo in recent years.
Collapse
Affiliation(s)
- Chen Lin
- Medical College of China three Gorges University;Yichang; China
| | - Ye-Zi Chen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy,China Three Gorges University; Yichang; China
| | - Bo Wu
- Medical College of China three Gorges University;Yichang; China
| | - Meng-Ting Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy,China Three Gorges University; Yichang; China
| | - Chao-Qi Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy,China Three Gorges University; Yichang; China
| | - Yun Zhao
- Medical College of China three Gorges University;Yichang; China
| |
Collapse
|
32
|
Henderson E, Huynh G, Wilson K, Plebanski M, Corrie S. The Development of Nanoparticles for the Detection and Imaging of Ovarian Cancers. Biomedicines 2021; 9:1554. [PMID: 34829783 PMCID: PMC8615601 DOI: 10.3390/biomedicines9111554] [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: 10/01/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/27/2022] Open
Abstract
Ovarian cancer remains as one of the most lethal gynecological cancers to date, with major challenges associated with screening, diagnosis and treatment of the disease and an urgent need for new technologies that can meet these challenges. Nanomaterials provide new opportunities in diagnosis and therapeutic management of many different types of cancers. In this review, we highlight recent promising developments of nanoparticles designed specifically for the detection or imaging of ovarian cancer that have reached the preclinical stage of development. This includes contrast agents, molecular imaging agents and intraoperative aids that have been designed for integration into standard imaging procedures. While numerous nanoparticle systems have been developed for ovarian cancer detection and imaging, specific design criteria governing nanomaterial targeting, biodistribution and clearance from the peritoneal cavity remain key challenges that need to be overcome before these promising tools can accomplish significant breakthroughs into the clinical setting.
Collapse
Affiliation(s)
- Edward Henderson
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; (E.H.); (G.H.)
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (K.W.); (M.P.)
| | - Gabriel Huynh
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; (E.H.); (G.H.)
| | - Kirsty Wilson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (K.W.); (M.P.)
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (K.W.); (M.P.)
| | - Simon Corrie
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; (E.H.); (G.H.)
- ARC Training Center for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
33
|
Detecting insulitis in type 1 diabetes with ultrasound phase-change contrast agents. Proc Natl Acad Sci U S A 2021; 118:2022523118. [PMID: 34607942 DOI: 10.1073/pnas.2022523118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 11/18/2022] Open
Abstract
Type 1 diabetes (T1D) results from immune infiltration and destruction of insulin-producing β cells within the pancreatic islets of Langerhans (insulitis). Early diagnosis during presymptomatic T1D would allow for therapeutic intervention prior to substantial β-cell loss at onset. There are limited methods to track the progression of insulitis and β-cell mass decline. During insulitis, the islet microvasculature increases permeability, such that submicron-sized particles can extravasate and accumulate within the islet microenvironment. Ultrasound is a widely deployable and cost-effective clinical imaging modality. However, conventional microbubble contrast agents are restricted to the vasculature. Submicron nanodroplet (ND) phase-change agents can be vaporized into micron-sized bubbles, serving as a microbubble precursor. We tested whether NDs extravasate into the immune-infiltrated islet microenvironment. We performed ultrasound contrast-imaging following ND infusion in nonobese diabetic (NOD) mice and NOD;Rag1ko controls and tracked diabetes development. We measured the biodistribution of fluorescently labeled NDs, with histological analysis of insulitis. Ultrasound contrast signal was elevated in the pancreas of 10-wk-old NOD mice following ND infusion and vaporization but was absent in both the noninfiltrated kidney of NOD mice and the pancreas of Rag1ko controls. High-contrast elevation also correlated with rapid diabetes onset. Elevated contrast was also observed as early as 4 wk, prior to mouse insulin autoantibody detection. In the pancreata of NOD mice, infiltrated islets and nearby exocrine tissue were selectively labeled with fluorescent NDs. Thus, contrast ultrasound imaging with ND phase-change agents can detect insulitis prior to diabetes onset. This will be important for monitoring disease progression, to guide and assess preventative therapeutic interventions for T1D.
Collapse
|
34
|
Paknahad AA, Kerr L, Wong DA, Kolios MC, Tsai SSH. Biomedical nanobubbles and opportunities for microfluidics. RSC Adv 2021; 11:32750-32774. [PMID: 35493576 PMCID: PMC9042222 DOI: 10.1039/d1ra04890b] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/19/2021] [Indexed: 12/17/2022] Open
Abstract
The use of bulk nanobubbles in biomedicine is increasing in recent years, which is attributable to the array of therapeutic and diagnostic tools promised by developing bulk nanobubble technologies. From cancer drug delivery and ultrasound contrast enhancement to malaria detection and the diagnosis of acute donor tissue rejection, the potential applications of bulk nanobubbles are broad and diverse. Developing these technologies to the point of clinical use may significantly impact the quality of patient care. This review compiles and summarizes a representative collection of the current applications, fabrication techniques, and characterization methods of bulk nanobubbles in biomedicine. Current state-of-the-art generation methods are not designed to create nanobubbles of high concentration and low polydispersity, both characteristics of which are important for several bulk nanobubble applications. To date, microfluidics has not been widely considered as a tool for generating nanobubbles, even though the small-scale precision and real-time control offered by microfluidics may overcome the challenges mentioned above. We suggest possible uses of microfluidics for improving the quality of bulk nanobubble populations and propose ways of leveraging existing microfluidic technologies, such as organ-on-a-chip platforms, to expand the experimental toolbox of researchers working to develop biomedical nanobubbles. The use of bulk nanobubbles in biomedicine is increasing in recent years. This translates into new opportunities for microfluidics, which may enable the generation of higher quality nanobubbles that lead to advances in diagnostics and therapeutics.![]()
Collapse
Affiliation(s)
- Ali A Paknahad
- Department of Mechanical and Industrial Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada .,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada
| | - Liam Kerr
- Department of Mechanical and Industrial Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada .,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada
| | - Daniel A Wong
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada.,Department of Electrical, Computer, and Biomedical Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada
| | - Michael C Kolios
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada.,Department of Physics, Ryerson University Toronto Ontario M5B 2K3 Canada
| | - Scott S H Tsai
- Department of Mechanical and Industrial Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada .,Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Ryerson University and St. Michael's Hospital 209 Victoria Street Toronto Ontario M5B 1T8 Canada.,Keenan Research Centre for Biomedical Science, Unity Health Toronto 209 Victoria Street Toronto Ontario M5B 1W8 Canada.,Graduate Program in Biomedical Engineering, Ryerson University 350 Victoria Street Toronto Ontario M5B 2K3 Canada
| |
Collapse
|
35
|
|
36
|
Abstract
Low-intensity ultrasound-triggered sonodynamic therapy (SDT) is a promising noninvasive therapeutic modality due to its strong tissue penetration ability. In recent years, with the development of nanotechnology, nanoparticle-based sonosensitizer-mediated SDT has been widely investigated. With the increasing demand for precise and personalized treatment, abundant novel sonosensitizers with imaging capabilities have been developed for determining the optimal therapeutic window, thus significantly enhancing treatment efficacy. In this review, we focus on the molecular imaging-guided SDT. The prevalent mechanisms of SDT are discussed, including ultrasonic cavitation, sonoluminescence, reactive oxygen species, and mechanical damage. In addition, we introduce the major molecular imaging techniques and the design principles based on nanoparticles to achieve efficient imaging. Furthermore, the imaging-guided SDT for the treatment of cancer, bacterial infections, and vascular diseases is summarized. The ultimate goal is to design more effective imaging-guided SDT modalities and provide novel ideas for clinical translation of SDT.
Collapse
Affiliation(s)
- Juan Guo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Xueting Pan
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Chaohui Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| |
Collapse
|
37
|
Wang Y, Cong H, Wang S, Yu B, Shen Y. Development and application of ultrasound contrast agents in biomedicine. J Mater Chem B 2021; 9:7633-7661. [PMID: 34586124 DOI: 10.1039/d1tb00850a] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the rapid development of molecular imaging, ultrasound (US) medicine has evolved from traditional imaging diagnosis to integrated diagnosis and treatment at the molecular level. Ultrasound contrast agents (UCAs) play a crucial role in the integration of US diagnosis and treatment. As the micro-bubbles (MBs) in UCAs can enhance the cavitation effect and promote the biological effect of US, UCAs have also been studied in the fields of US thrombolysis, mediated gene transfer, drug delivery, and high intensity focused US. The application range of UCAs is expanding, and the value of their applications is improving. This paper reviews the development and application of UCAs in biomedicine in recent years, and the existing problems and prospects are pointed out.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China.
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China.
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Building D, Science Park, Qingdao 266071, China. .,Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
38
|
Zhang Y, Fowlkes JB. Liposomes-based nanoplatform enlarges ultrasound-related diagnostic and therapeutic precision. Curr Med Chem 2021; 29:1331-1341. [PMID: 34348609 DOI: 10.2174/0929867328666210804092624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/07/2022]
Abstract
Ultrasound (US) is notable in the medical field as a safe and effective imaging modality due to its lack of ionizing radiation, non-invasive approach, and real-time monitoring capability. Accompanying recent progress in nanomedicine, US has been providing hope of theranostic capability not only for imaging-based diagnosis but also for US-based therapy by taking advantage of the bioeffects induced by US. Cavitation, sonoporation, thermal effects, and other cascade effects stimulated by acoustic energy conversion have contributed to medical problem-solving in the past decades although to varying degrees of efficacy in comparisons to other methods. Recently, the usage of liposomes-based nanoplatform fuels the development of nanomedicine and provides novel clinical strategies for antitumor, thrombolysis, and controlled drug release. Merging of novel liposome-based nanoplatforms and US-induced reactions has promise for a new blueprint for future medicine. In the present review article, the value of liposome-based nanoplatforms in US-related diagnosis and therapy will be discussed and summarized along with potential future directions for further investigations.
Collapse
Affiliation(s)
- Ying Zhang
- Dept. Radiology, University of Michigan, Ann Arbor, Michigan, 48109. United States
| | - J Brian Fowlkes
- Dept. Radiology, University of Michigan, Ann Arbor, Michigan, 48109. United States
| |
Collapse
|
39
|
Exner AA, Kolios MC. Bursting Microbubbles: How Nanobubble Contrast Agents Can Enable the Future of Medical Ultrasound Molecular Imaging and Image-Guided Therapy. Curr Opin Colloid Interface Sci 2021; 54:101463. [PMID: 34393610 PMCID: PMC8356903 DOI: 10.1016/j.cocis.2021.101463] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The field of medical ultrasound has undergone a significant evolution since the development of microbubbles as contrast agents. However, due to their size, microbubbles remain in the vasculature, and therefore have limited clinical applications. Building a better - and smaller - bubble can expand the applications of contrast-enhanced ultrasound by allowing bubbles to extravasate from blood vessels - creating new opportunities. In this review, we summarize recent research on the formulation and use of NBs as imaging agents and as therapeutic vehicles. We discuss the ongoing debates in the field and reluctance to accepting NBs as an acoustically active construct and a potentially impactful clinical tool that can help shape the future of medical ultrasound. We hope that the overview of key experimental and theoretical findings in the NB field presented in this paper provides a fundamental framework that will help clarify NB-ultrasound interactions and inspire engagement in the field.
Collapse
Affiliation(s)
- Agata A. Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
40
|
Batchelor DV, Armistead FJ, Ingram N, Peyman SA, Mclaughlan JR, Coletta PL, Evans SD. Nanobubbles for therapeutic delivery: Production, stability and current prospects. Curr Opin Colloid Interface Sci 2021. [DOI: 10.1016/j.cocis.2021.101456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Abou-Saleh RH, Armistead FJ, Batchelor DVB, Johnson BRG, Peyman SA, Evans SD. Horizon: Microfluidic platform for the production of therapeutic microbubbles and nanobubbles. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2021; 92:074105. [PMID: 34340422 DOI: 10.1063/5.0040213] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 06/17/2021] [Indexed: 06/13/2023]
Abstract
Microbubbles (MBs) have a multitude of applications including as contrast agents in ultrasound imaging and as therapeutic drug delivery vehicles, with further scope for combining their diagnostic and therapeutic properties (known as theranostics). MBs used clinically are commonly made by mechanical agitation or sonication methods, which offer little control over population size and dispersity. Furthermore, clinically used MBs are yet to be used therapeutically and further research is needed to develop these theranostic agents. In this paper, we present our MB production instrument "Horizon," which is a robust, portable, and user-friendly instrument, integrating the key components for producing MBs using microfluidic flow-focusing devices. In addition, we present the system design and specifications of Horizon and the optimized protocols that have so far been used to produce MBs with specific properties. These include MBs with tailored size and low dispersity (monodisperse); MBs with a diameter of ∼2 μm, which are more disperse but also produced in higher concentration; nanobubbles with diameters of 100-600 nm; and therapeutic MBs with drug payloads for targeted delivery. Multiplexed chips were able to improve production rates up to 16-fold while maintaining production stability. This work shows that Horizon is a versatile instrument with potential for mass production and use across many research facilities, which could begin to bridge the gap between therapeutic MB research and clinical use.
Collapse
Affiliation(s)
- Radwa H Abou-Saleh
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Fern J Armistead
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Damien V B Batchelor
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Benjamin R G Johnson
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sally A Peyman
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Stephen D Evans
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
42
|
Johansen ML, Perera R, Abenojar E, Wang X, Vincent J, Exner AA, Brady-Kalnay SM. Ultrasound-Based Molecular Imaging of Tumors with PTPmu Biomarker-Targeted Nanobubble Contrast Agents. Int J Mol Sci 2021; 22:1983. [PMID: 33671448 PMCID: PMC7922223 DOI: 10.3390/ijms22041983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 11/16/2022] Open
Abstract
Ultrasound imaging is a widely used, readily accessible and safe imaging modality. Molecularly-targeted microbubble- and nanobubble-based contrast agents used in conjunction with ultrasound imaging expand the utility of this modality by specifically targeting and detecting biomarkers associated with different pathologies including cancer. In this study, nanobubbles directed to a cancer biomarker derived from the Receptor Protein Tyrosine Phosphatase mu, PTPmu, were evaluated alongside non-targeted nanobubbles using contrast enhanced ultrasound both in vitro and in vivo in mice. In vitro resonant mass and clinical ultrasound measurements showed gas-core, lipid-shelled nanobubbles conjugated to either a PTPmu-directed peptide or a Scrambled control peptide were equivalent. Mice with heterotopic human tumors expressing the PTPmu-biomarker were injected with PTPmu-targeted or control nanobubbles and dynamic contrast-enhanced ultrasound was performed. Tumor enhancement was more rapid and greater with PTPmu-targeted nanobubbles compared to the non-targeted control nanobubbles. Peak tumor enhancement by the PTPmu-targeted nanobubbles occurred within five minutes of contrast injection and was more than 35% higher than the Scrambled nanobubble signal for the subsequent two minutes. At later time points, the signal in tumors remained higher with PTPmu-targeted nanobubbles demonstrating that PTPmu-targeted nanobubbles recognize tumors using molecular ultrasound imaging and may be useful for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Mette L. Johansen
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106-4960, USA; (M.L.J.); (J.V.)
| | - Reshani Perera
- Department of Radiology, Case Western Reserve University, Cleveland, OH 44106-4960, USA; (R.P.); (E.A.)
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH 44106-4960, USA; (R.P.); (E.A.)
| | - Xinning Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106-7207, USA;
| | - Jason Vincent
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106-4960, USA; (M.L.J.); (J.V.)
| | - Agata A. Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH 44106-4960, USA; (R.P.); (E.A.)
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106-7207, USA;
| | - Susann M. Brady-Kalnay
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106-4960, USA; (M.L.J.); (J.V.)
| |
Collapse
|
43
|
Xiaoting ZBS, Zhifei DP. Micro/Nanobubbles Driven Multimodal Imaging and Theragnostics of Cancer. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2021. [DOI: 10.37015/audt.2021.200053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
44
|
Su C, Ren X, Nie F, Li T, Lv W, Li H, Zhang Y. Current advances in ultrasound-combined nanobubbles for cancer-targeted therapy: a review of the current status and future perspectives. RSC Adv 2021; 11:12915-12928. [PMID: 35423829 PMCID: PMC8697319 DOI: 10.1039/d0ra08727k] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
The non-specific distribution, non-selectivity towards cancerous cells, and adverse off-target side effects of anticancer drugs and other therapeutic molecules lead to their inferior clinical efficacy. Accordingly, ultrasound-based targeted delivery of therapeutic molecules loaded in smart nanocarriers is currently gaining wider acceptance for the treatment and management of cancer. Nanobubbles (NBs) are nanosize carriers, which are currently used as effective drug/gene delivery systems because they can deliver drugs/genes selectively to target sites. Thus, combining the applications of ultrasound with NBs has recently demonstrated increased localization of anticancer molecules in tumor tissues with triggered release behavior. Consequently, an effective therapeutic concentration of drugs/genes is achieved in target tumor tissues with ultimately increased therapeutic efficacy and minimal side-effects on other non-cancerous tissues. This review illustrates present developments in the field of ultrasound-nanobubble combined strategies for targeted cancer treatment. The first part of this review discusses the composition and the formulation parameters of NBs. Next, we illustrate the interactions and biological effects of combining NBs and ultrasound. Subsequently, we explain the potential of NBs combined with US for targeted cancer therapeutics. Finally, the present and future directions for the improvement of current methods are proposed. NBs combined with ultrasound demonstrated the ability to enhance the targeting of anticancer agents and improve the efficacy.![]()
Collapse
Affiliation(s)
- Chunhong Su
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
- Department of Pain, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - XiaoJun Ren
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Fang Nie
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Tiangang Li
- Department of Ultrasound Diagnosis, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou, 730030, Gansu Province, China
| | - Wenhao Lv
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Hui Li
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
- Department of Pneumology, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| | - Yao Zhang
- Department of Ultrasound Diagnosis, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
- Department of Emergency, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu Province, China
| |
Collapse
|
45
|
Zhu Y, Sun Y, Liu W, Guan W, Liu H, Duan Y, Chen Y. Magnetic polymeric nanobubbles with optimized core size for MRI/ultrasound bimodal molecular imaging of prostate cancer. Nanomedicine (Lond) 2020; 15:2901-2916. [PMID: 33300812 DOI: 10.2217/nnm-2020-0188] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim: To design MRI/ultrasound (US) dual modality imaging probes with optimized size for prostate cancer imaging by targeting prostate-specific membrane antigen (PSMA). Materials & methods: The PSMA-targeting polypeptide-nanobubbles (PP-NBs) with core size of 400 and 700 nm were fabricated and evaluated. Results: With excellent physical property and specificity, PP-NBs of both core size could image PSMA expression in prostate cancer xenografts. Particularly, 400 nm PP-NBs generated higher PSMA-specific MRI/US dual modality contrast enhancement than 700 nm PP-NBs in correlation with histopathologic findings. Conclusion: Benefit from the smaller core size, 400 nm PP-NBs had higher permeability and specificity than 700 nm PP-NBs, hence producing better PSMA-specific MRI/US dual modality imaging.
Collapse
Affiliation(s)
- Yunkai Zhu
- Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Weiyong Liu
- Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Wenbin Guan
- Department of Pathology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Yaqing Chen
- Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| |
Collapse
|
46
|
Hysi E, Fadhel MN, Wang Y, Sebastian JA, Giles A, Czarnota GJ, Exner AA, Kolios MC. Photoacoustic imaging biomarkers for monitoring biophysical changes during nanobubble-mediated radiation treatment. PHOTOACOUSTICS 2020; 20:100201. [PMID: 32775198 PMCID: PMC7393572 DOI: 10.1016/j.pacs.2020.100201] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/24/2020] [Accepted: 07/22/2020] [Indexed: 05/04/2023]
Abstract
The development of novel anticancer therapies warrants the parallel development of biomarkers that can quantify their effectiveness. Photoacoustic imaging has the potential to measure changes in tumor vasculature during treatment. Establishing the accuracy of imaging biomarkers requires direct comparisons with gold histological standards. In this work, we explore whether a new class of submicron, vascular disrupting, ultrasonically stimulated nanobubbles enhance radiation therapy. In vivo experiments were conducted on mice bearing prostate cancer tumors. Combined nanobubble plus radiation treatments were compared against conventional microbubbles and radiation alone (single 8 Gy fraction). Acoustic resolution photoacoustic imaging was used to monitor the effects of the treatments 2- and 24-hs post-administration. Histological examination provided metrics of tumor vascularity and tumoral cell death, both of which were compared to photoacoustic-derived biomarkers. Photoacoustic metrics of oxygen saturation reveal a 20 % decrease in oxygenation within 24 h post-treatment. The spectral slope metric could separate the response of the nanobubble treatments from the microbubble counterparts. This study shows that histopathological assessment correlated well with photoacoustic biomarkers of treatment response.
Collapse
Affiliation(s)
- Eno Hysi
- Department of Physics, Ryerson University, Toronto, Canada
- Insitute for Biomedical Engineering, Science and Technology, St. Michael’s Hospital, Toronto, Canada
| | - Muhannad N. Fadhel
- Department of Physics, Ryerson University, Toronto, Canada
- Insitute for Biomedical Engineering, Science and Technology, St. Michael’s Hospital, Toronto, Canada
| | - Yanjie Wang
- Department of Physics, Ryerson University, Toronto, Canada
- Insitute for Biomedical Engineering, Science and Technology, St. Michael’s Hospital, Toronto, Canada
| | - Joseph A. Sebastian
- Department of Physics, Ryerson University, Toronto, Canada
- Insitute for Biomedical Engineering, Science and Technology, St. Michael’s Hospital, Toronto, Canada
| | - Anoja Giles
- Deparment of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Gregory J. Czarnota
- Deparment of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
- Deparment of Medical Biophysics, University of Toronto, Canada
- Department of Radiation Oncology, University of Toronto, Canada
| | - Agata A. Exner
- Department of Radiology, Case Western Reserve University, Cleveland, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, United States
| | - Michael C. Kolios
- Department of Physics, Ryerson University, Toronto, Canada
- Insitute for Biomedical Engineering, Science and Technology, St. Michael’s Hospital, Toronto, Canada
| |
Collapse
|
47
|
Pellow C, Abenojar EC, Exner AA, Zheng G, Goertz DE. Concurrent visual and acoustic tracking of passive and active delivery of nanobubbles to tumors. Am J Cancer Res 2020; 10:11690-11706. [PMID: 33052241 PMCID: PMC7545999 DOI: 10.7150/thno.51316] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background: There has been growing interest in nanobubbles for their potential to extend bubble-mediated ultrasound approaches beyond that of their larger microbubble counterparts. In particular, the smaller scale of nanobubbles may enable them to access the tumor extravascular compartment for imaging and therapy in closer proximity to cancer cells. Compelling preliminary demonstrations of the imaging and therapeutic abilities of nanobubbles have thus emerged, with emphasis on their ability to extravasate. However, studies to date rely on indirect histologic evidence that cannot confirm whether the structures remain intact beyond the vasculature - leaving their extravascular potential largely untapped. Methods: Nanobubble acoustic scattering was assessed using a recently reported ultra-stable formulation at low concentration (106 mL-1) and frequency (1 MHz), over a range of pressures (100-1500 kPa) in a channel phantom. The pressure-dependent response was utilized as a basis for in vivo experiments where ultrasound transmitters and receivers were integrated into a window chamber for simultaneous intravital multiphoton microscopy and acoustic monitoring in tumor-affected microcirculation. Microscopy and acoustic data were utilized to assess passive and active delivery of nanobubbles and determine whether they remained intact beyond the vasculature. Results: Nanobubbles exhibit pressure-dependent nonlinear acoustic scattering. Nanobubbles are also found to have prolonged acoustic vascular pharmacokinetics, and passively extravasate intact into tumors. Ultrasound stimulation of nanobubbles is shown to actively enhance the delivery of both intact nanobubbles and shell material, increasing their spatial bioavailability deeper into the extravascular space. A range of acute vascular effects were also observed. Conclusion: This study presents the first direct evidence that nanobubbles passively and actively extravasate intact in tumor tissue, and is the first to directly capture acute vascular events from ultrasound-stimulation of nanobubbles. The insights gained here demonstrate an important step towards unlocking the potential of nanobubbles and extending ultrasound-based applications.
Collapse
|
48
|
Batchelor DVB, Abou-Saleh RH, Coletta PL, McLaughlan JR, Peyman SA, Evans SD. Nested Nanobubbles for Ultrasound-Triggered Drug Release. ACS APPLIED MATERIALS & INTERFACES 2020; 12:29085-29093. [PMID: 32501014 PMCID: PMC7333229 DOI: 10.1021/acsami.0c07022] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Because of their size (1-10 μm), microbubble-based drug delivery agents suffer from confinement to the vasculature, limiting tumor penetration and potentially reducing the drug efficacy. Nanobubbles (NBs) have emerged as promising candidates for ultrasound-triggered drug delivery because of their small size, allowing drug delivery complexes to take advantage of the enhanced permeability and retention effect. In this study, we describe a simple method for production of nested-nanobubbles (Nested-NBs) by encapsulation of NBs (∼100 nm) within drug-loaded liposomes. This method combines the efficient and well-established drug-loading capabilities of liposomes while utilizing NBs as an acoustic trigger for drug release. Encapsulation was characterized using transmission electron microscopy with an encapsulation efficiency of 22 ± 2%. Nested-NBs demonstrated echogenicity using diagnostic B-mode imaging, and acoustic emissions were monitored during high-intensity focused ultrasound (HIFU) in addition to monitoring of model drug release. Results showed that although the encapsulated NBs were destroyed by pulsed HIFU [peak negative pressure (PNP) 1.54-4.83 MPa], signified by loss of echogenicity and detection of inertial cavitation, no model drug release was observed. Changing modality to continuous wave (CW) HIFU produced release across a range of PNPs (2.01-3.90 MPa), likely because of a synergistic effect of mechanical and increased thermal stimuli. Because of this, we predict that our NBs contain a mixed population of both gaseous and liquid core particles, which upon CW HIFU undergo rapid phase conversion, triggering liposomal drug release. This hypothesis was investigated using previously described models to predict the existence of droplets and their phase change potential and the ability of this phase change to induce liposomal drug release.
Collapse
Affiliation(s)
| | - Radwa H. Abou-Saleh
- Department of Physics
and Astronomy, University of Leeds, Leeds, U.K.
- Department
of Physics, Mansoura University, Mansoura, Egypt
| | - P. Louise Coletta
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St. James’s University Hospital, Leeds, U.K.
| | - James. R. McLaughlan
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St. James’s University Hospital, Leeds, U.K.
- School
of Electronic and Electrical Engineering, University of Leeds, Leeds, U.K.
| | - Sally A. Peyman
- Department of Physics
and Astronomy, University of Leeds, Leeds, U.K.
- Leeds
Institute of Medical Research, Wellcome Trust Brenner Building, St. James’s University Hospital, Leeds, U.K.
| | - Stephen D. Evans
- Department of Physics
and Astronomy, University of Leeds, Leeds, U.K.
- . Phone/Fax: (+44) (0)113 343 3852
| |
Collapse
|
49
|
Ramirez DG, Abenojar E, Hernandez C, Lorberbaum DS, Papazian LA, Passman S, Pham V, Exner AA, Benninger RKP. Contrast-enhanced ultrasound with sub-micron sized contrast agents detects insulitis in mouse models of type1 diabetes. Nat Commun 2020; 11:2238. [PMID: 32382089 PMCID: PMC7206014 DOI: 10.1038/s41467-020-15957-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
In type1 diabetes (T1D) autoreactive T-cells infiltrate the islets of Langerhans, depleting insulin-secreting β-cells (insulitis). Insulitis arises during an asymptomatic phase, prior to clinical diagnosis of T1D. Methods to diagnose insulitis and β-cell mass changes during this asymptomatic phase are limited, precluding early therapeutic intervention. During T1D the islet microvasculature increases permeability, allowing nanoparticles to access the microenvironment. Contrast enhanced ultrasound (CEUS) uses shell-stabilized gas bubbles to provide acoustic backscatter in vasculature. Here, we report that sub-micron sized 'nanobubble' ultrasound contrast agents can be used to measure increased islet microvasculature permeability and indicate asymptomatic T1D. Through CEUS and histological analysis, pre-clinical models of T1D show accumulation of nanobubbles specifically within pancreatic islets, correlating with insulitis. Importantly, accumulation is detected early in disease progression and decreases with successful therapeutic intervention. Thus, sub-micron sized nanobubble ultrasound contrast agents provide a predicative marker for disease progression and therapeutic reversal early in asymptomatic T1D.
Collapse
Affiliation(s)
- David G Ramirez
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric Abenojar
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher Hernandez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - David S Lorberbaum
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lucine A Papazian
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Samantha Passman
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Vinh Pham
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Agata A Exner
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
50
|
Kida H, Nishimura K, Ogawa K, Watanabe A, Feril LB, Irie Y, Endo H, Kawakami S, Tachibana K. Nanobubble Mediated Gene Delivery in Conjunction With a Hand-Held Ultrasound Scanner. Front Pharmacol 2020; 11:363. [PMID: 32300298 PMCID: PMC7145407 DOI: 10.3389/fphar.2020.00363] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Recent research has revealed that nanobubbles (NBs) can be an effective tool for gene transfection in conjunction with therapeutic ultrasound (US). However, an approach to apply commercially available hand-held diagnostic US scanners for this purpose has not been evaluated as of now. In the present study, we first compared in vitro, the efficiency of gene transfer (pCMV-Luciferase) with lipid-based and albumin-based NBs irradiated by therapeutic US (1MHz, 5.0 W/cm2) in oral squamous carcinoma cell line HSC-2. Secondly, we similarly examined if gene transfer in mice is possible using a clinical hand-held US scanner (2.3MHz, MI 1.0). Results showed that lipid-based NBs induced more gene transfection compared to albumin-based NBs, in vitro. Furthermore, significant gene transfer was also obtained in mice liver with lipid-based NBs. Sub-micro sized bubbles proved to be a powerful gene transfer reagent in combination with conventional hand-held ultrasonic diagnostic device.
Collapse
Affiliation(s)
- Hiroshi Kida
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Koyo Nishimura
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Akiko Watanabe
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Loreto B Feril
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yutaka Irie
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Hitomi Endo
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuro Tachibana
- Department of Anatomy, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|