51
|
Songtanin B, Brittan K, Sanchez S, Le M, Schmidt C, Ingviya T, Manatsathit W. Diagnostic performance of contrast-enhanced ultrasound in diagnosing hepatic artery occlusion after liver transplantation: A systematic review and meta-analysis. Clin Transplant 2023; 37:e15070. [PMID: 37398993 DOI: 10.1111/ctr.15070] [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: 03/02/2023] [Revised: 06/03/2023] [Accepted: 06/25/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Hepatic artery occlusion (HAO) is a significant complication post-liver transplantation. Doppler ultrasound (DUS) has been widely used as an initial screening test for detecting HAO; however, its performance is often not sufficient. Although other diagnostic tests such as computed tomography angiography (CTA), magnetic resonance angiography (MRA), and angiogram are more accurate, they are invasive and have several limitations. Contrast-enhanced ultrasound (CEUS) is an emerging tool for detecting HAO; however, the results from previous studies were limited due to a small number of patients. Therefore, we aimed to evaluate its performance by performing a meta-analysis. METHOD We performed a systemic review and meta-analysis of studies evaluating the performance of CEUS for the detection of HAO in an adult population. A literature search of EMBASE, Scopus, CINAHL, and Medline was conducted through March 2022. Pooled sensitivity, specificity, log diagnostic odd ratio (LDOR), and area under summary receiver operating curve (AUC) were calculated. Publication bias was assessed by Deeks' funnel plot. RESULT Eight studies were included, with 434 CEUS performed. Using a combination of CTA, MRA, angiography, clinical follow-up, and surgery as the gold standard, the sensitivity, specificity, and LDOR of CEUS for detection of HAO were .969 (.938, .996), .991 (.981, 1.001), and 5.732 (4.539, 6.926), respectively. AUC was .959. The heterogeneity between studies appeared universally low, and no significant publication bias was found (p = .44). CONCLUSION CEUS appeared to have an excellent performance for the detection of HAO and could be considered as an alternative when DUS is non-diagnostic or when CTA, MRA, and angiogram are not feasible.
Collapse
Affiliation(s)
- Busara Songtanin
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kevin Brittan
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sebastian Sanchez
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Michelle Le
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Cynthia Schmidt
- McGoogan Library of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Thammasin Ingviya
- Department of Family and Preventive Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Medical Data Center for Research and Innovation, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Wuttiporn Manatsathit
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
52
|
Jia W, Zhou Z, Zhan W. Musculoskeletal Biomaterials: Stimulated and Synergized with Low Intensity Pulsed Ultrasound. J Funct Biomater 2023; 14:504. [PMID: 37888169 PMCID: PMC10607075 DOI: 10.3390/jfb14100504] [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: 08/11/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Clinical biophysical stimulating strategies, which have significant effects on improving the function of organs or treating diseases by causing the salutary response of body, have shown many advantages, such as non-invasiveness, few side effects, and controllable treatment process. As a critical technique for stimulation, the low intensity pulsed ultrasound (LIPUS) has been explored in regulating osteogenesis, which has presented great promise in bone repair by delivering a combined effect with biomaterials. This review summarizes the musculoskeletal biomaterials that can be synergized with LIPUS for enhanced biomedical application, including bone regeneration, spinal fusion, osteonecrosis/osteolysis, cartilage repair, and nerve regeneration. Different types of biomaterials are categorized for summary and evaluation. In each subtype, the verified biological mechanisms are listed in a table or graphs to prove how LIPUS was effective in improving musculoskeletal tissue regeneration. Meanwhile, the acoustic excitation parameters of LIPUS that were promising to be effective for further musculoskeletal tissue engineering are discussed, as well as their limitations and some perspectives for future research. Overall, coupled with biomimetic scaffolds and platforms, LIPUS may be a powerful therapeutic approach to accelerate musculoskeletal tissue repair and even in other regenerative medicine applications.
Collapse
Affiliation(s)
- Wanru Jia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
53
|
Ganguly A, Chaudhary S, Sirsi SR, Prasad S. H.O.S.T.: Hemoglobin microbubble-based Oxidative stress Sensing Technology. Sci Rep 2023; 13:14942. [PMID: 37696978 PMCID: PMC10495409 DOI: 10.1038/s41598-023-42050-z] [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: 04/03/2023] [Accepted: 09/05/2023] [Indexed: 09/13/2023] Open
Abstract
In this work, we discuss the development of H.O.S.T., a novel hemoglobin microbubble-based electrochemical biosensor for label-free detection of Hydrogen peroxide (H2O2) towards oxidative stress and cancer diagnostic applications. The novelty of the constructed sensor lies in the use of a sonochemically prepared hemoglobin microbubble capture probe, which allowed for an extended dynamic range, lower detection limit, and enhanced resolution compared to the native hemoglobin based H2O2 biosensors. The size of the prepared particles Hemoglobin microbubbles was characterized using Coulter Counter analysis and was found to be 4.4 microns, and the morphology of these spherical microbubbles was shown using Brightfield microscopy. The binding chemistry of the sensor stack elements of HbMbs' and P.A.N.H.S. crosslinker was characterized using Attenuated Total Reflectance Fourier Transform Infrared Spectroscopy and UV-Vis Spectroscopy. The electrochemical biosensor calibration (R2 > 0.95) was done using Electrochemical Impedance Spectroscopy, Cyclic Voltammetry, and Square Wave Voltammetry. The electrochemical biosensor calibration (R2 > 0.95) was done using Electrochemical Impedance Spectroscopy, Cyclic Voltammetry, and Square Wave Voltammetry. The specificity of the sensor for H2O2 was analyzed using cross-reactivity studies using ascorbic acid and glucose as interferents (p < 0.0001 for the highest non-specific dose versus the lowest specific dose). The developed sensor showed good agreement in performance with a commercially available kit for H2O2 detection using Bland Altman Analysis (mean bias = 0.37 for E.I.S. and - 24.26 for CV). The diagnostic potential of the biosensor was further tested in cancerous (N.G.P.) and non-cancerous (H.E.K.) cell lysate for H2O2 detection (p = 0.0064 for E.I.S. and p = 0.0062 for CV). The Michaelis Menten constant calculated from the linear portion of the sensor was found to be [Formula: see text] of 19.44 µM indicating that our biosensor has a higher affinity to Hydrogen peroxide than other available enzymatic sensors, it is attributed to the unique design of the hemoglobin polymers in microbubble.
Collapse
Affiliation(s)
- Antra Ganguly
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Sugandha Chaudhary
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Shalini Prasad
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
54
|
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
|
55
|
Dan Q, Jiang X, Wang R, Dai Z, Sun D. Biogenic Imaging Contrast Agents. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207090. [PMID: 37401173 PMCID: PMC10477908 DOI: 10.1002/advs.202207090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/08/2023] [Indexed: 07/05/2023]
Abstract
Imaging contrast agents are widely investigated in preclinical and clinical studies, among which biogenic imaging contrast agents (BICAs) are developing rapidly and playing an increasingly important role in biomedical research ranging from subcellular level to individual level. The unique properties of BICAs, including expression by cells as reporters and specific genetic modification, facilitate various in vitro and in vivo studies, such as quantification of gene expression, observation of protein interactions, visualization of cellular proliferation, monitoring of metabolism, and detection of dysfunctions. Furthermore, in human body, BICAs are remarkably helpful for disease diagnosis when the dysregulation of these agents occurs and can be detected through imaging techniques. There are various BICAs matched with a set of imaging techniques, including fluorescent proteins for fluorescence imaging, gas vesicles for ultrasound imaging, and ferritin for magnetic resonance imaging. In addition, bimodal and multimodal imaging can be realized through combining the functions of different BICAs, which helps overcome the limitations of monomodal imaging. In this review, the focus is on the properties, mechanisms, applications, and future directions of BICAs.
Collapse
Affiliation(s)
- Qing Dan
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| | - Xinpeng Jiang
- Department of Biomedical EngineeringCollege of Future TechnologyPeking UniversityBeijing100871P. R. China
| | - Run Wang
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| | - Zhifei Dai
- Department of Biomedical EngineeringCollege of Future TechnologyPeking UniversityBeijing100871P. R. China
| | - Desheng Sun
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| |
Collapse
|
56
|
Kancheva M, Aronson L, Pattilachan T, Sautto F, Daines B, Thommes D, Shar A, Razavi M. Bubble-Based Drug Delivery Systems: Next-Generation Diagnosis to Therapy. J Funct Biomater 2023; 14:373. [PMID: 37504868 PMCID: PMC10382061 DOI: 10.3390/jfb14070373] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023] Open
Abstract
Current radiologic and medication administration is systematic and has widespread side effects; however, the administration of microbubbles and nanobubbles (MNBs) has the possibility to provide therapeutic and diagnostic information without the same ramifications. Microbubbles (MBs), for instance, have been used for ultrasound (US) imaging due to their ability to remain in vessels when exposed to ultrasonic waves. On the other hand, nanobubbles (NBs) can be used for further therapeutic benefits, including chronic treatments for osteoporosis and cancer, gene delivery, and treatment for acute conditions, such as brain infections and urinary tract infections (UTIs). Clinical trials are also being conducted for different administrations and utilizations of MNBs. Overall, there are large horizons for the benefits of MNBs in radiology, general medicine, surgery, and many more medical applications. As such, this review aims to evaluate the most recent publications from 2016 to 2022 to report the current uses and innovations for MNBs.
Collapse
Affiliation(s)
- Mihaela Kancheva
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Lauren Aronson
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Tara Pattilachan
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Francesco Sautto
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Benjamin Daines
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Donald Thommes
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Angela Shar
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Mehdi Razavi
- Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
57
|
Karakatsani ME, Ji R, Murillo MF, Kugelman T, Kwon N, Lao YH, Liu K, Pouliopoulos AN, Honig LS, Duff KE, Konofagou EE. Focused ultrasound mitigates pathology and improves spatial memory in Alzheimer's mice and patients. Theranostics 2023; 13:4102-4120. [PMID: 37554284 PMCID: PMC10405840 DOI: 10.7150/thno.79898] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/12/2023] [Indexed: 08/10/2023] Open
Abstract
Rationale: Bilateral sonication with focused ultrasound (FUS) in conjunction with microbubbles has been shown to separately reduce amyloid plaques and hyperphosphorylated tau protein in the hippocampal formation and the entorhinal cortex in different mouse models of Alzheimer's disease (AD) without any therapeutic agents. However, the two pathologies are expressed concurrently in human disease. Therefore, the objective of this study is to investigate the effects of repeated bilateral sonications in the presence of both pathologies. Methods: Herein, we investigate its functional and morphological outcomes on brains bearing both pathologies simultaneously. Eleven transgenic mice of the 3xTg-AD line (14 months old) expressing human amyloid beta and human tau and eleven age-matched wild-type littermates received four weekly bilateral sonications covering the hippocampus followed by working memory testing. Afterwards, immunohistochemistry and immunoassays (western blot and ELISA) were employed to assess any changes in amyloid beta and human tau. Furthermore, we present preliminary data from our clinical trial using a neuronavigation-guided FUS system for sonications in AD patients (NCT04118764). Results: Interestingly, both wild-type and transgenic animals that received FUS experienced improved working memory and spent significantly more time in the escape platform-quadrant, with wild-type animals spending 43.2% (sham: 37.7%) and transgenic animals spending 35.3% (sham: 31.0%) of the trial in the target quadrant. Furthermore, this behavioral amelioration in the transgenic animals correlated with a 58.3% decrease in the neuronal length affected by tau and a 27.2% reduction in total tau levels. Amyloid plaque population, volume and overall load were also reduced overall. Consistently, preliminary data from a clinical trial involving AD patients showed a 1.8% decrease of amyloid PET signal 3-weeks after treatment in the treated hemisphere compared to baseline. Conclusion: For the first time, it is shown that bilateral FUS-induced BBB opening significantly and simultaneously ameliorates both coexistent pathologies, which translated to improvements in spatial memory of transgenic animals with complex AD, the human mimicking phenotype. The level of cognitive improvement was significantly correlated with the volume of BBB opening. Non-transgenic animals were also shown to exhibit similar memory amelioration for the first time, indicating that BBB opening results into benefits in the neuronal function regardless of the existence of AD pathology. A potential mechanism of action for the reduction of the both pathologies investigated was the cholesterol metabolism, specifically the LRP1b receptor, which exhibited increased expression levels in transgenic mice following FUS-induced BBB opening. Initial clinical evidence supported that the beta amyloid reduction shown in rodents could be translatable to humans with significant amyloid reduction shown in the treated hemisphere.
Collapse
Affiliation(s)
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Maria F. Murillo
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Tara Kugelman
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Keyu Liu
- Department of Biomedical Engineering, Columbia University, New York, USA
| | | | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Karen E. Duff
- UK Dementia Research Institute, University College London, London, UK
| | - Elisa E. Konofagou
- Department of Radiology, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
58
|
Zhang W, Metzger H, Vlatakis S, Claxton A, Carbajal MA, Fung LF, Mason J, Chan KLA, Pouliopoulos AN, Fleck RA, Prentice P, Thanou M. Characterising the chemical and physical properties of phase-change nanodroplets. ULTRASONICS SONOCHEMISTRY 2023; 97:106445. [PMID: 37257208 PMCID: PMC10241977 DOI: 10.1016/j.ultsonch.2023.106445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
Phase-change nanodroplets have attracted increasing interest in recent years as ultrasound theranostic nanoparticles. They are smaller compared to microbubbles and they may distribute better in tissues (e.g. in tumours). They are composed of a stabilising shell and a perfluorocarbon core. Nanodroplets can vaporise into echogenic microbubbles forming cavitation nuclei when exposed to ultrasound. Their perfluorocarbon core phase-change is responsible for the acoustic droplet vaporisation. However, methods to quantify the perfluorocarbon core in nanodroplets are lacking. This is an important feature that can help explain nanodroplet phase change characteristics. In this study, we fabricated nanodroplets using lipids shell and perfluorocarbons. To assess the amount of perfluorocarbon in the core we used two methods, 19F NMR and FTIR. To assess the cavitation after vaporisation we used an ultrasound transducer (1.1 MHz) and a high-speed camera. The 19F NMR based method showed that the fluorine signal correlated accurately with the perfluorocarbon concentration. Using this correlation, we were able to quantify the perfluorocarbon core of nanodroplets. This method was used to assess the content of the perfluorocarbon of the nanodroplets in solutions over time. It was found that perfluoropentane nanodroplets lost their content faster and at higher ratio compared to perfluorohexane nanodroplets. The high-speed imaging indicates that the nanodroplets generate cavitation comparable to that from commercial contrast agent microbubbles. Nanodroplet characterisation should include perfluorocarbon concentration assessment as critical information for their development.
Collapse
Affiliation(s)
- Weiqi Zhang
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Hilde Metzger
- School of Engineering, University of Glasgow, United Kingdom
| | - Stavros Vlatakis
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Amelia Claxton
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | | | - Leong Fan Fung
- Department of Surgical & Interventional Engineering, King's College London, United Kingdom
| | - James Mason
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - K L Andrew Chan
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | | | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, United Kingdom
| | - Paul Prentice
- School of Engineering, University of Glasgow, United Kingdom
| | - Maya Thanou
- Institute of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom.
| |
Collapse
|
59
|
Paknahad AA, Zalloum IO, Karshafian R, Kolios MC, Tsai SSH. Microfluidic nanobubbles: observations of a sudden contraction of microbubbles into nanobubbles. SOFT MATTER 2023. [PMID: 37386867 DOI: 10.1039/d3sm00380a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Microfluidic devices are often utilized to generate uniform-size microbubbles. In most microfluidic bubble generation experiments, once the bubbles are formed the gas inside the bubbles begin to dissolve into the surrounding aqueous environment. The bubbles shrink until they attain an equilibrium size dictated by the concentration and type of amphiphilic molecules stabilizing the gas-liquid interface. Here, we exploit this shrinkage mechanism, and control the solution lipid concentration and microfluidic geometry, to make monodisperse bulk nanobubbles. Interestingly, we make the surprising observation of a critical microbubble diameter above and below which the scale of bubble shrinkage dramatically changes. Namely, microbubbles generated with an initial diameter larger than the critical diameter shrinks to a stable diameter that is consistent with previous literature. However, microbubbles that are initially smaller than the critical diameter experience a sudden contraction into nanobubbles whose size is at least an order-of-magnitude below expectations. We apply electron microscopy and resonance mass measurement methods to quantify the size and uniformity of the nanobubbles, and probe the dependence of the critical bubble diameter on the lipid concentration. We anticipate that further analysis of this unexpected microbubble sudden contraction regime can lead to more robust technologies for making monodisperse nanobubbles.
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
|
60
|
Shen Q, Li Z, Meyer MD, De Guzman MT, Lim JC, Bouchard RR, Lu GJ. 50-nm gas-filled protein nanostructures to enable the access of lymphatic cells by ultrasound technologies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546433. [PMID: 37425762 PMCID: PMC10327079 DOI: 10.1101/2023.06.27.546433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Ultrasound imaging and ultrasound-mediated gene and drug delivery are rapidly advancing diagnostic and therapeutic methods; however, their use is often limited by the need of microbubbles, which cannot transverse many biological barriers due to their large size. Here we introduce 50-nm gas-filled protein nanostructures derived from genetically engineered gas vesicles that we referred to as 50nm GVs. These diamond-shaped nanostructures have hydrodynamic diameters smaller than commercially available 50-nm gold nanoparticles and are, to our knowledge, the smallest stable, free-floating bubbles made to date. 50nm GVs can be produced in bacteria, purified through centrifugation, and remain stable for months. Interstitially injected 50nm GVs can extravasate into lymphatic tissues and gain access to critical immune cell populations, and electron microscopy images of lymph node tissues reveal their subcellular location in antigen-presenting cells adjacent to lymphocytes. We anticipate that 50nm GVs can substantially broaden the range of cells accessible to current ultrasound technologies and may generate applications beyond biomedicine as ultrasmall stable gas-filled nanomaterials.
Collapse
|
61
|
Rad IJ, Chapman L, Tupally KR, Veidt M, Al-Sadiq H, Sullivan R, Parekh HS. A systematic review of ultrasound-mediated drug delivery to the eye and critical insights to facilitate a timely path to the clinic. Theranostics 2023; 13:3582-3638. [PMID: 37441595 PMCID: PMC10334839 DOI: 10.7150/thno.82884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/11/2023] [Indexed: 07/15/2023] Open
Abstract
Ultrasound has long been identified as a promising, non-invasive modality for improving ocular drug delivery across a range of indications. Yet, with 20 years of learnings behind us, clinical translation remains limited. To help address this, and in accordance with PRISMA guidelines, the various mechanisms of ultrasound-mediated ocular drug delivery have been appraised, ranging from first principles to emergent applications spanning both ex vivo and in vivo models. The heterogeneity of study methods precluded meta-analysis, however an extensive characterisation of the included studies allowed for semi-quantitative and qualitative assessments. Methods: In this review, we reflected on study quality of reporting, and risk of bias (RoB) using the latest Animal Research: Reporting of In Vivo Experiments (ARRIVE 2.0) guidelines, alongside the Systematic Review Centre for Laboratory animal Experimentation (SYRCLE) RoB tools. Literature studies from 2002 to 2022 were initially characterised according to methods of ultrasound application, ultrasound parameters applied, animal models employed, as well as safety and efficacy assessments. This exercise contributed to developing a comprehensive understanding of the current state of play within ultrasound-mediated ocular drug delivery. The results were then synthesised and processed into a guide to aid future study design, with the goal of improving the reliability of data, and to support efficient and timely translation to the clinic. Results: Key attributes identified as hindering translation included: poor reporting quality and high RoB, skewed use of animals unrepresentative of the human eye, and the over reliance of reductionist safety assessments. Ex vivo modelling studies were often unable to have comprehensive safety assessments performed on them, which are imperative to determining treatment safety, and represent a pre-requisite for clinical translation. Conclusion: With the use of our synthesised guide, and a thorough understanding of the underlying physicochemical interactions between ultrasound and ocular biology provided herein, this review offers a firm foundation on which future studies should ideally be built, such that ultrasound-mediated ocular drug delivery can be translated from concept to the coalface where it can provide immense clinical benefit.
Collapse
Affiliation(s)
- Isaac J Rad
- The University of Queensland, School of Pharmacy, Brisbane, Queensland, Australia
- The University of Queensland, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Luke Chapman
- The University of Queensland, Faculty of Medicine, Brisbane, Queensland, Australia
| | | | - Martin Veidt
- The University of Queensland, School of Mechanical and Mining Engineering, Brisbane, Queensland, Australia
| | - Hussain Al-Sadiq
- Al-Asala University, Department of Industrial Engineering, Dammam, Saudi Arabia
| | - Robert Sullivan
- The University of Queensland, Queensland Brain Institute, Brisbane, Queensland, Australia
| | - Harendra S Parekh
- The University of Queensland, School of Pharmacy, Brisbane, Queensland, Australia
| |
Collapse
|
62
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
63
|
Navarro-Becerra JA, Borden MA. Targeted Microbubbles for Drug, Gene, and Cell Delivery in Therapy and Immunotherapy. Pharmaceutics 2023; 15:1625. [PMID: 37376072 DOI: 10.3390/pharmaceutics15061625] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Microbubbles are 1-10 μm diameter gas-filled acoustically-active particles, typically stabilized by a phospholipid monolayer shell. Microbubbles can be engineered through bioconjugation of a ligand, drug and/or cell. Since their inception a few decades ago, several targeted microbubble (tMB) formulations have been developed as ultrasound imaging probes and ultrasound-responsive carriers to promote the local delivery and uptake of a wide variety of drugs, genes, and cells in different therapeutic applications. The aim of this review is to summarize the state-of-the-art of current tMB formulations and their ultrasound-targeted delivery applications. We provide an overview of different carriers used to increase drug loading capacity and different targeting strategies that can be used to enhance local delivery, potentiate therapeutic efficacy, and minimize side effects. Additionally, future directions are proposed to improve the tMB performance in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
| | - Mark A Borden
- Mechanical Engineering Department, University of Colorado Boulder, Boulder, CO 80309, USA
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
64
|
LuTheryn G, Ho EML, Choi V, Carugo D. Cationic Microbubbles for Non-Selective Binding of Cavitation Nuclei to Bacterial Biofilms. Pharmaceutics 2023; 15:pharmaceutics15051495. [PMID: 37242736 DOI: 10.3390/pharmaceutics15051495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/06/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
The presence of multi-drug resistant biofilms in chronic, persistent infections is a major barrier to successful clinical outcomes of therapy. The production of an extracellular matrix is a characteristic of the biofilm phenotype, intrinsically linked to antimicrobial tolerance. The heterogeneity of the extracellular matrix makes it highly dynamic, with substantial differences in composition between biofilms, even in the same species. This variability poses a major challenge in targeting drug delivery systems to biofilms, as there are few elements both suitably conserved and widely expressed across multiple species. However, the presence of extracellular DNA within the extracellular matrix is ubiquitous across species, which alongside bacterial cell components, gives the biofilm its net negative charge. This research aims to develop a means of targeting biofilms to enhance drug delivery by developing a cationic gas-filled microbubble that non-selectively targets the negatively charged biofilm. Cationic and uncharged microbubbles loaded with different gases were formulated and tested to determine their stability, ability to bind to negatively charged artificial substrates, binding strength, and, subsequently, their ability to adhere to biofilms. It was shown that compared to their uncharged counterparts, cationic microbubbles facilitated a significant increase in the number of microbubbles that could both bind and sustain their interaction with biofilms. This work is the first to demonstrate the utility of charged microbubbles for the non-selective targeting of bacterial biofilms, which could be used to significantly enhance stimuli-mediated drug delivery to the bacterial biofilm.
Collapse
Affiliation(s)
- Gareth LuTheryn
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), The Botnar Research Centre, University of Oxford, Windmill Road, Oxford OX3 7HE, UK
- Faculty of Engineering and Physical Sciences, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Elaine M L Ho
- Faculty of Engineering and Physical Sciences, University of Southampton, University Road, Southampton SO17 1BJ, UK
- Artificial Intelligence and Informatics, The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0QX, UK
| | - Victor Choi
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Parks Road, Oxford OX1 3PJ, UK
| | - Dario Carugo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), The Botnar Research Centre, University of Oxford, Windmill Road, Oxford OX3 7HE, UK
| |
Collapse
|
65
|
Zhao N, Curry D, Evans RE, Isguven S, Freeman T, Eisenbrey JR, Forsberg F, Gilbertie JM, Boorman S, Hilliard R, Dastgheyb SS, Machado P, Stanczak M, Harwood M, Chen AF, Parvizi J, Shapiro IM, Hickok NJ, Schaer TP. Microbubble cavitation restores Staphylococcus aureus antibiotic susceptibility in vitro and in a septic arthritis model. Commun Biol 2023; 6:425. [PMID: 37069337 PMCID: PMC10110534 DOI: 10.1038/s42003-023-04752-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/24/2023] [Indexed: 04/19/2023] Open
Abstract
Treatment failure in joint infections is associated with fibrinous, antibiotic-resistant, floating and tissue-associated Staphylococcus aureus aggregates formed in synovial fluid (SynF). We explore whether antibiotic activity could be increased against Staphylococcus aureus aggregates using ultrasound-triggered microbubble destruction (UTMD), in vitro and in a porcine model of septic arthritis. In vitro, when bacterially laden SynF is diluted, akin to the dilution achieved clinically with lavage and local injection of antibiotics, amikacin and ultrasound application result in increased bacterial metabolism, aggregate permeabilization, and a 4-5 log decrease in colony forming units, independent of microbubble destruction. Without SynF dilution, amikacin + UTMD does not increase antibiotic activity. Importantly, in the porcine model of septic arthritis, no bacteria are recovered from the SynF after treatment with amikacin and UTMD-ultrasound without UTMD is insufficient. Our data suggest that UTMD + antibiotics may serve as an important adjunct for the treatment of septic arthritis.
Collapse
Affiliation(s)
- Neil Zhao
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dylan Curry
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rachel E Evans
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Selin Isguven
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Theresa Freeman
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessica M Gilbertie
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Sophie Boorman
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Rachel Hilliard
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Sana S Dastgheyb
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Priscilla Machado
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Stanczak
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Marc Harwood
- Rothman Orthopaedic Institute, Philadelphia, PA, USA
| | - Antonia F Chen
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Javad Parvizi
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
- Rothman Orthopaedic Institute, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Thomas P Schaer
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA.
| |
Collapse
|
66
|
Kida H, Yamasaki Y, Feril Jr. LB, Endo H, Itaka K, Tachibana K. Efficient mRNA Delivery with Lyophilized Human Serum Albumin-Based Nanobubbles. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1283. [PMID: 37049376 PMCID: PMC10097217 DOI: 10.3390/nano13071283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
In this study, we developed an efficient mRNA delivery vehicle by optimizing a lyophilization method for preserving human serum albumin-based nanobubbles (HSA-NBs), bypassing the need for artificial stabilizers. The morphology of the lyophilized material was verified using scanning electron microscopy, and the concentration, size, and mass of regenerated HSA-NBs were verified using flow cytometry, nanoparticle tracking analysis, and resonance mass measurements, and compared to those before lyophilization. The study also evaluated the response of HSA-NBs to 1 MHz ultrasound irradiation and their ultrasound (US) contrast effect. The functionality of the regenerated HSA-NBs was confirmed by an increased expression of intracellularly transferred Gluc mRNA, with increasing intensity of US irradiation. The results indicated that HSA-NBs retained their structural and functional integrity markedly, post-lyophilization. These findings support the potential of lyophilized HSA-NBs, as efficient imaging, and drug delivery systems for various medical applications.
Collapse
Affiliation(s)
- Hiroshi Kida
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Yutaro Yamasaki
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Loreto B. Feril Jr.
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Hitomi Endo
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Tokyo 101-0062, Japan
| | - Katsuro Tachibana
- Department of Anatomy, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
67
|
Dasgupta A, Sun T, Palomba R, Rama E, Zhang Y, Power C, Moeckel D, Liu M, Sarode A, Weiler M, Motta A, Porte C, Magnuska Z, Said Elshafei A, Barmin R, Graham A, McClelland A, Rommel D, Stickeler E, Kiessling F, Pallares RM, De Laporte L, Decuzzi P, McDannold N, Mitragotri S, Lammers T. Nonspherical ultrasound microbubbles. Proc Natl Acad Sci U S A 2023; 120:e2218847120. [PMID: 36940339 PMCID: PMC10068850 DOI: 10.1073/pnas.2218847120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/22/2023] [Indexed: 03/22/2023] Open
Abstract
Surface tension provides microbubbles (MB) with a perfect spherical shape. Here, we demonstrate that MB can be engineered to be nonspherical, endowing them with unique features for biomedical applications. Anisotropic MB were generated via one-dimensionally stretching spherical poly(butyl cyanoacrylate) MB above their glass transition temperature. Compared to their spherical counterparts, nonspherical polymeric MB displayed superior performance in multiple ways, including i) increased margination behavior in blood vessel-like flow chambers, ii) reduced macrophage uptake in vitro, iii) prolonged circulation time in vivo, and iv) enhanced blood-brain barrier (BBB) permeation in vivo upon combination with transcranial focused ultrasound (FUS). Our studies identify shape as a design parameter in the MB landscape, and they provide a rational and robust framework for further exploring the application of anisotropic MB for ultrasound-enhanced drug delivery and imaging applications.
Collapse
Affiliation(s)
- Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Tao Sun
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, 16163Genova, Italy
| | - Elena Rama
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Yongzhi Zhang
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Chanikarn Power
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Diana Moeckel
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Mengjiao Liu
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Apoorva Sarode
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Marek Weiler
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Alessandro Motta
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Céline Porte
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Zuzanna Magnuska
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Roman Barmin
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Adam Graham
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Arthur McClelland
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Dirk Rommel
- DWI−Leibniz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Polymeric Biomaterials, Rheinisch-Westfälische Technische Hochschule University Aachen, 52074Aachen, Germany
| | - Elmar Stickeler
- Department of Obstetrics and Gynecology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Roger M. Pallares
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Laura De Laporte
- DWI−Leibniz Institute for Interactive Materials, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, Polymeric Biomaterials, Rheinisch-Westfälische Technische Hochschule University Aachen, 52074Aachen, Germany
- Advanced Materials for Biomedicine, Institute of Applied Medical Engineering, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, 16163Genova, Italy
| | - Nathan McDannold
- Focused Ultrasound Laboratory, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| |
Collapse
|
68
|
Edwards IA, De Carlo F, Sitta J, Varner W, Howard CM, Claudio PP. Enhancing Targeted Therapy in Breast Cancer by Ultrasound-Responsive Nanocarriers. Int J Mol Sci 2023; 24:ijms24065474. [PMID: 36982548 PMCID: PMC10053544 DOI: 10.3390/ijms24065474] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Currently, the response to cancer treatments is highly variable, and severe side effects and toxicity are experienced by patients receiving high doses of chemotherapy, such as those diagnosed with triple-negative breast cancer. The main goal of researchers and clinicians is to develop new effective treatments that will be able to specifically target and kill tumor cells by employing the minimum doses of drugs exerting a therapeutic effect. Despite the development of new formulations that overall can increase the drugs’ pharmacokinetics, and that are specifically designed to bind overexpressed molecules on cancer cells and achieve active targeting of the tumor, the desired clinical outcome has not been reached yet. In this review, we will discuss the current classification and standard of care for breast cancer, the application of nanomedicine, and ultrasound-responsive biocompatible carriers (micro/nanobubbles, liposomes, micelles, polymeric nanoparticles, and nanodroplets/nanoemulsions) employed in preclinical studies to target and enhance the delivery of drugs and genes to breast cancer.
Collapse
Affiliation(s)
- Isaiah A. Edwards
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Flavia De Carlo
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Juliana Sitta
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William Varner
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Candace M. Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Pier Paolo Claudio
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Correspondence:
| |
Collapse
|
69
|
Hu Y, Wei J, Shen Y, Chen S, Chen X. Barrier-breaking effects of ultrasonic cavitation for drug delivery and biomarker release. ULTRASONICS SONOCHEMISTRY 2023; 94:106346. [PMID: 36870921 PMCID: PMC10040969 DOI: 10.1016/j.ultsonch.2023.106346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 05/27/2023]
Abstract
Recently, emerging evidence has demonstrated that cavitation actually creates important bidirectional channels on biological barriers for both intratumoral drug delivery and extratumoral biomarker release. To promote the barrier-breaking effects of cavitation for both therapy and diagnosis, we first reviewed recent technical advances of ultrasound and its contrast agents (microbubbles, nanodroplets, and gas-stabilizing nanoparticles) and then reported the newly-revealed cavitation physical details. In particular, we summarized five types of cellular responses of cavitation in breaking the plasma membrane (membrane retraction, sonoporation, endocytosis/exocytosis, blebbing and apoptosis) and compared the vascular cavitation effects of three different types of ultrasound contrast agents in breaking the blood-tumor barrier and tumor microenvironment. Moreover, we highlighted the current achievements of the barrier-breaking effects of cavitation in mediating drug delivery and biomarker release. We emphasized that the precise induction of a specific cavitation effect for barrier-breaking was still challenged by the complex combination of multiple acoustic and non-acoustic cavitation parameters. Therefore, we provided the cutting-edge in-situ cavitation imaging and feedback control methods and suggested the development of an international cavitation quantification standard for the clinical guidance of cavitation-mediated barrier-breaking effects.
Collapse
Affiliation(s)
- Yaxin Hu
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Jianpeng Wei
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Yuanyuan Shen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Siping Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Xin Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China.
| |
Collapse
|
70
|
Microbubbles for human diagnosis and therapy. Biomaterials 2023; 294:122025. [PMID: 36716588 DOI: 10.1016/j.biomaterials.2023.122025] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
Microbubbles (MBs) were observed for the first time in vivo as a curious consequence of quick saline injection during ultrasound (US) imaging of the aortic root, more than 50 years ago. From this serendipitous event, MBs are now widely used as contrast enhancers for US imaging. Their intrinsic properties described in this review, allow a multitude of designs, from shell to gas composition but also from grafting targeting agents to drug payload encapsulation. Indeed, the versatile MBs are deeply studied for their dual potential in imaging and therapy. As presented in this paper, new generations of MBs now opens perspectives for targeted molecular imaging along with the development of new US imaging systems. This review also presents an overview of the different therapeutic strategies with US and MBs for cancer, cardiovascular diseases, and inflammation. The overall aim is to overlap those fields in order to find similarities in the MBs application for treatment enhancement associated with US. To conclude, this review explores the new scales of MBs technologies with nanobubbles development, and along concurrent advances in the US imaging field. This review ends by discussing perspectives for the booming future uses of MBs.
Collapse
|
71
|
Fan CH, Wu N, Yeh CK. Enhanced sonodynamic therapy by carbon dots-shelled microbubbles with focused ultrasound. ULTRASONICS SONOCHEMISTRY 2023; 94:106342. [PMID: 36842213 PMCID: PMC9988694 DOI: 10.1016/j.ultsonch.2023.106342] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 05/28/2023]
Abstract
Sonodynamic therapy involving the non-invasive and local generation of lethal reactive oxygen species (ROS) via ultrasound (US) with sonosensitizers has been proposed as an emerging tumor therapy strategy. However, such therapy is usually associated with inertial cavitation and unnecessary damage to healthy tissue because current sonosensitizers have insufficient sensitivity to US. Here, we report the use of a new proposed sonosensitizer, carbon dots (C-dots), to assemble microbubbles with a gas core (C-dots MBs). As the C-dots were directly integrated into the MB shell, they could effectively absorb the energy of inertial cavitation and transfer it to ROS. Our results revealed the appearance of 1O2, •OH, and H2O2 after US irradiation of C-dots MBs. In in vitro experiments, treatment with C-dots MBs plus US induced lipid peroxidation, elevation of intracellular ROS, and apoptosis in 32.5%, 45.3%, and 50.1% of cells respectively. In an animal solid tumor model, treatment with C-dots MBs plus US resulted in a 3-fold and 2.5-fold increase in the proportion of ROS-damaged cells and apoptotic cells, respectively, compared to C-dots MBs alone. These results will pave the way for the design of novel multifunctional sonosensitizers for SDT tumor therapy.
Collapse
Affiliation(s)
- Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Nan Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
72
|
Yadav AS, Tran DT, Teo AJT, Dai Y, Galogahi FM, Ooi CH, Nguyen NT. Core-Shell Particles: From Fabrication Methods to Diverse Manipulation Techniques. MICROMACHINES 2023; 14:497. [PMID: 36984904 PMCID: PMC10054063 DOI: 10.3390/mi14030497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
Core-shell particles are micro- or nanoparticles with solid, liquid, or gas cores encapsulated by protective solid shells. The unique composition of core and shell materials imparts smart properties on the particles. Core-shell particles are gaining increasing attention as tuneable and versatile carriers for pharmaceutical and biomedical applications including targeted drug delivery, controlled drug release, and biosensing. This review provides an overview of fabrication methods for core-shell particles followed by a brief discussion of their application and a detailed analysis of their manipulation including assembly, sorting, and triggered release. We compile current methodologies employed for manipulation of core-shell particles and demonstrate how existing methods of assembly and sorting micro/nanospheres can be adopted or modified for core-shell particles. Various triggered release approaches for diagnostics and drug delivery are also discussed in detail.
Collapse
Affiliation(s)
- Ajeet Singh Yadav
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Du Tuan Tran
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Adrian J. T. Teo
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University, Singapore 637460, Singapore
| | - Yuchen Dai
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Fariba Malekpour Galogahi
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Chin Hong Ooi
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia
| |
Collapse
|
73
|
Messerschmidt V, Ren W, Tsipursky M, Irudayaraj J. Characterization of Oxygen Nanobubbles and In Vitro Evaluation of Retinal Cells in Hypoxia. Transl Vis Sci Technol 2023; 12:16. [PMID: 36763051 PMCID: PMC9927786 DOI: 10.1167/tvst.12.2.16] [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: 08/23/2022] [Accepted: 01/14/2023] [Indexed: 02/11/2023] Open
Abstract
Purpose Vein or artery occlusion causes a hypoxic environment by preventing oxygen delivery and diffusion to tissues. Diseases such as retinal vein occlusion, central retinal artery occlusion, or diabetic retinopathy create a stroke-type condition that leads to functional blindness in the effected eye. We aim to develop an oxygen delivery system consisting of oxygen nanobubbles (ONBs) that can mitigate retinal ischemia during a severe hypoxic event such as central retinal artery occlusion. Methods ONBs were synthesized to encapsulate oxygen saturated molecular medical grade water. Stability, oxygen release, biocompatibility, reactive oxygen species, superoxide, MTT, and terminal uridine nick-end labeling assays were performed. Cell viability was evaluated, and safety experiments were conducted in rabbits. Results The ONBs were approximately 220 nm in diameter, with a zeta potential of -58.8 mV. Oxygen release studies indicated that 74.06 µg of O2 is released from the ONBs after 12 hours at 37°C. Cell studies indicated that ONBs are safe and cells are viable. There was no significant increase in reactive oxygen species, superoxide, or double-stranded DNA damage after ONB treatment. ONBs preserve mitochondrial function and viability. Histological sections from rabbit eyes indicated that ONBs were not toxic. Conclusions The ONBs proposed have excellent oxygen holding and release properties to mitigate ischemic conditions in the retina. They are sterile, stable, and nontoxic. Translation Relevance ONB technology was evaluated for its physical properties, oxygen release, sterility, stability, and safety. Our results indicate that ONBs could be a viable treatment approach to mitigate hypoxia during ischemic conditions in the eye upon timely administration.
Collapse
Affiliation(s)
- Victoria Messerschmidt
- Biomedical Research Centre (BRC), Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Wen Ren
- Biomedical Research Centre (BRC), Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Michael Tsipursky
- Vitreo-Retinal Surgery, Ophthalmology Department, Carle Foundation Hospital, Champaign, IL, USA
- Carle-Illinois College of Medicine, Champaign, IL, USA
| | - Joseph Irudayaraj
- Biomedical Research Centre (BRC), Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Carle-Illinois College of Medicine, Champaign, IL, USA
- Beckman Institute; Holonyak Micro and Nanotechnology Laboratory; Carl Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
74
|
Pathour T, Chaudhary S, Sirsi SR, Fei B. Hemoglobin Microbubbles and the Prediction of Different Oxygen Levels Using RF Data and Deep Learning. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2023; 12470:124700G. [PMID: 38495411 PMCID: PMC10942652 DOI: 10.1117/12.2655121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Ultrasound contrast agents (UCA) are gas-encapsulated microspheres that oscillate volumetrically when exposed to an ultrasound field producing backscattered signals efficiently, which can be used for improved ultrasound imaging and drug delivery applications. We developed a novel oxygen-sensitive hemoglobin-shell microbubble designed to acoustically detect blood oxygen levels. We hypothesize that structural change in hemoglobin caused due to varying oxygen levels in the body can lead to mechanical changes in the shell of the UCA. This can produce detectable changes in the acoustic response that can be used for measuring oxygen levels in the body. In this study, we have shown that oxygenated hemoglobin microbubbles can be differentiated from deoxygenated hemoglobin microbubbles using a 1D convolutional neural network using radiofrequency (RF) data. We were able to classify RF data from oxygenated and deoxygenated hemoglobin microbubbles into the two classes with a testing accuracy of 90.15%. The results suggest that oxygen content in hemoglobin affects the acoustical response and may be used for determining oxygen levels and thus could open many applications, including evaluating hypoxic regions in tumors and the brain, among other blood-oxygen-level-dependent imaging applications.
Collapse
Affiliation(s)
- Teja Pathour
- Center for Imaging and Surgical Innovation, University of Texas at Dallas, Richardson, TX
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Sugandha Chaudhary
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Shashank R. Sirsi
- Center for Imaging and Surgical Innovation, University of Texas at Dallas, Richardson, TX
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
| | - Baowei Fei
- Center for Imaging and Surgical Innovation, University of Texas at Dallas, Richardson, TX
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX
- Department of Radiology. UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
75
|
Teed C, Hussein H, Kishen A. Synchronized Microbubble Photodynamic Activation to Disinfect Minimally Prepared Root Canals. J Endod 2023; 49:198-204. [PMID: 36509168 DOI: 10.1016/j.joen.2022.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/24/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The purpose of this study was to evaluate the antimicrobial efficacy of a novel irrigation strategy using synchronized microbubble photodynamic activation (SYMPA) in a minimally prepared single canal. METHODS Single-canal mandibular incisors were inoculated with Enterococcus faecalis for 3 weeks and randomly allocated to 4 groups based on the irrigation protocols: (1) control (saline), (2) conventional needle irrigation (CI), (3) ultrasonic-assisted irrigation (UI), and (4) irrigation with SYMPA. The first 3 groups were instrumented to size 25.07v (WaveOne Gold Primary; Dentsply Sirona, Johnson City, TN), and the SYMPA group was minimally prepared to size 20.07v (WaveOne Gold Small, Dentsply Sirona). The apical 5 mm was resected for microbiological assessment using the culture technique (colony-forming unit), adenosine-5'-triphosphate-based viability assay (relative luminescence units), and the percentage of live bacteria using confocal laser scanning microscopy. RESULTS Log colony-forming units from the UI (2.37 ± 0.66) and SYMPA (2.21 ± 0.86) groups showed a reduction compared with the control (5.16 ± 0.75) and CI (4.08 ± 1.19) groups. Relative luminescence unit reduction was significant for UI (619.08 ± 352.78) and SYMPA (415.25 ± 329.51) compared with the control (1213.2 ± 880.03) (P < .05). The percentage of live bacteria was significantly lower in the UI and SYMPA groups compared with the control and CI groups. Although higher microbial reduction was observed in SYMPA compared with UI, there was no statistical significance (P > .05). CONCLUSION SYMPA in minimally prepared canals showed significant antimicrobial efficacy. The novel irrigation strategy using SYMPA could be an effective disinfection strategy for minimally prepared root canals.
Collapse
Affiliation(s)
- Christine Teed
- MSc Endodontics Program, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Hebatullah Hussein
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada; Department of Endodontics, Faculty of Dentistry, Ain-Shams University, Cairo, Egypt
| | - Anil Kishen
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
76
|
Tang Y, Tang S, Huang C, Klippel P, Ma C, Caso N, Chen S, Jing Y, Yao J. High-fidelity deep functional photoacoustic tomography enhanced by virtual point sources. PHOTOACOUSTICS 2023; 29:100450. [PMID: 36685991 PMCID: PMC9852650 DOI: 10.1016/j.pacs.2023.100450] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
Photoacoustic tomography (PAT), a hybrid imaging modality that acoustically detects the optical absorption contrast, is a promising technology for imaging hemodynamic functions in deep tissues far beyond the traditional optical microscopy. However, the most clinically compatible PAT often suffers from the poor image fidelity, mostly due to the limited detection view of the linear ultrasound transducer array. PAT can be improved by employing highly-absorbing contrast agents such as droplets and nanoparticles, which, however, have low clinical translation potential due to safety concerns and regulatory hurdles imposed by these agents. In this work, we have developed a new methodology that can fundamentally improve PAT's image fidelity without hampering any of its functional capability or clinical translation potential. By using clinically-approved microbubbles as virtual point sources that strongly and isotropically scatter the local pressure waves generated by surrounding hemoglobin, we can overcome the limited-detection-view problem and achieve high-fidelity functional PAT in deep tissues, a technology referred to as virtual-point-source PAT (VPS-PAT). We have thoroughly investigated the working principle of VPS-PAT by numerical simulations and in vitro phantom experiments, clearly showing the signal origin of VPSs and the resultant superior image fidelity over traditional PAT. We have also demonstrated in vivo applications of VPT-PAT for functional small-animal studies with physiological challenges. We expect that VPS-PAT can find broad applications in biomedical research and accelerated translation to clinical impact.
Collapse
Affiliation(s)
- Yuqi Tang
- Photoacoustic Imaging Lab, Department of Biomedical Engineering, Duke University, Durham, NC, the United States of America
| | - Shanshan Tang
- Ultrasound Imaging Lab, Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, the United States of America
| | - Chengwu Huang
- Ultrasound Imaging Lab, Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, the United States of America
| | - Paul Klippel
- Graduate Program in Acoustic and Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, the United States of America
| | - Chenshuo Ma
- Photoacoustic Imaging Lab, Department of Biomedical Engineering, Duke University, Durham, NC, the United States of America
| | - Nathan Caso
- Graduate Program in Acoustic and Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, the United States of America
| | - Shigao Chen
- Ultrasound Imaging Lab, Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, the United States of America
| | - Yun Jing
- Graduate Program in Acoustic and Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, the United States of America
| | - Junjie Yao
- Photoacoustic Imaging Lab, Department of Biomedical Engineering, Duke University, Durham, NC, the United States of America
| |
Collapse
|
77
|
Sridharan B, Lim HG. Exosomes and ultrasound: The future of theranostic applications. Mater Today Bio 2023; 19:100556. [PMID: 36756211 PMCID: PMC9900624 DOI: 10.1016/j.mtbio.2023.100556] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Biomaterials and pertaining formulations have been very successful in various diagnostic and therapeutic applications because of its ability to overcome pharmacological limitations. Some of them have gained significant focus in the recent decade for their theranostic properties. Exosomes can be grouped as biomaterials, since they consist of various biological micro/macromolecules and possess all the properties of a stable biomaterial with size in nano range. Significant research has gone into isolation and exploitation of exosomes as potential theranostic agent. However, the limitations in terms of yield, efficacy, and target specificity are continuously being addressed. On the other hand, several nano/microformulations are responsive to physical or chemical alterations and were successfully stimulated by tweaking the physical characteristics of the surrounding environment they are in. Some of them are termed as photodynamic, sonodynamic or thermodynamic therapeutic systems. In this regard, ultrasound and acoustic systems were extensively studied for its ability towards altering the properties of the systems to which they were applied on. In this review, we have detailed about the diagnostic and therapeutic applications of exosomes and ultrasound separately, consisting of their conventional applications, drawbacks, and developments for addressing the challenges. The information were categorized into various sections that provide complete overview of the isolation strategies and theranostic applications of exosomes in various diseases. Then the ultrasound-based disease diagnosis and therapy were elaborated, with special interest towards the use of ultrasound in enhancing the efficacy of nanomedicines and nanodrug delivery systems, Finally, we discussed about the ability of ultrasound in enhancing the diagnostic and therapeutic properties of exosomes, which could be the future of theranostics.
Collapse
Affiliation(s)
| | - Hae Gyun Lim
- Corresponding author. Biomedical Ultrasound Lab, Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
78
|
Kumar M, Kumar D, Chopra S, Mahmood S, Bhatia A. Microbubbles: Revolutionizing Biomedical Applications with Tailored Therapeutic Precision. Curr Pharm Des 2023; 29:3532-3545. [PMID: 38151837 DOI: 10.2174/0113816128282478231219044000] [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: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Over the past ten years, tremendous progress has been made in microbubble-based research for a variety of biological applications. Microbubbles emerged as a compelling and dynamic tool in modern drug delivery systems. They are employed to deliver drugs or genes to targeted regions of interest, and then ultrasound is used to burst the microbubbles, causing site-specific delivery of the bioactive materials. OBJECTIVE The objective of this article is to review the microbubble compositions and physiochemical characteristics in relation to the development of innovative biomedical applications, with a focus on molecular imaging and targeted drug/gene delivery. METHODS The microbubbles are prepared by using various methods, which include cross-linking polymerization, emulsion solvent evaporation, atomization, and reconstitution. In cross-linking polymerization, a fine foam of the polymer is formed, which serves as a bubble coating agent and colloidal stabilizer, resulting from the vigorous stirring of a polymeric solution. In the case of emulsion solvent evaporation, there are two solutions utilized in the production of microbubbles. In atomization and reconstitution, porous spheres are created by atomising a surfactant solution into a hot gas. They are encapsulated in primary modifier gas. After the addition of the second gas or gas osmotic agent, the package is placed into a vial and sealed after reconstituting with sterile saline solution. RESULTS Microbubble-based drug delivery is an innovative approach in the field of drug delivery that utilizes microbubbles, which are tiny gas-filled bubbles, act as carriers for therapeutic agents. These microbubbles can be loaded with drugs, imaging agents, or genes and then guided to specific target sites. CONCLUSION The potential utility of microbubbles in biomedical applications is continually growing as novel formulations and methods. The versatility of microbubbles allows for customization, tailoring the delivery system to various medical applications, including cancer therapy, cardiovascular treatments, and gene therapy.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, Punjab 151001, India
| | - Devesh Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, Punjab 151001, India
| | - Shruti Chopra
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, Punjab 151001, India
| | - Syed Mahmood
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, Punjab 151001, India
| |
Collapse
|
79
|
Wu N, Cao Y, Liu Y, Zhou Y, He H, Tang R, Wan L, Wang C, Xiong X, Zhong L, Li P. Low-intensity focused ultrasound targeted microbubble destruction reduces tumor blood supply and sensitizes anti-PD-L1 immunotherapy. Front Bioeng Biotechnol 2023; 11:1173381. [PMID: 37139047 PMCID: PMC10150078 DOI: 10.3389/fbioe.2023.1173381] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023] Open
Abstract
Immune checkpoint blockade (ICB) typified by anti-PD-1/PD-L1 antibodies as a revolutionary treatment for solid malignancies has been limited to a subset of patients due to poor immunogenicity and inadequate T cell infiltration. Unfortunately, no effective strategies combined with ICB therapy are available to overcome low therapeutic efficiency and severe side effects. Ultrasound-targeted microbubble destruction (UTMD) is an effective and safe technique holding the promise to decrease tumor blood perfusion and activate anti-tumor immune response based on the cavitation effect. Herein, we demonstrated a novel combinatorial therapeutic modality combining low-intensity focused ultrasound-targeted microbubble destruction (LIFU-TMD) with PD-L1 blockade. LIFU-TMD caused the rupture of abnormal blood vessels to deplete tumor blood perfusion and induced the tumor microenvironment (TME) transformation to sensitize anti-PD-L1 immunotherapy, which markedly inhibited 4T1 breast cancer's growth in mice. We discovered immunogenic cell death (ICD) in a portion of cells induced by the cavitation effect from LIFU-TMD, characterized by the increased expression of calreticulin (CRT) on the tumor cell surface. Additionally, flow cytometry revealed substantially higher levels of dendritic cells (DCs) and CD8+ T cells in draining lymph nodes and tumor tissue, as induced by pro-inflammatory molecules like IL-12 and TNF-α. These suggest that LIFU-TMD as a simple, effective, and safe treatment option provides a clinically translatable strategy for enhancing ICB therapy.
Collapse
Affiliation(s)
- Nianhong Wu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuting Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Liu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ultrasound, The Third People’s Hospital of Chengdu City, Chengdu, China
| | - Ying Zhou
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongye He
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Tang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wan
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Can Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xialin Xiong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Linhong Zhong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Pan Li,
| |
Collapse
|
80
|
Sharma D, Tarapacki CM, Kandavel H, Panchalingam M, Kim HC, Cartar H, Kaffas AE, Czarnota GJ. Evaluating the effects of radiation and acoustically-stimulated microbubble therapy in an in vivo breast cancer model. PLoS One 2023; 18:e0277759. [PMID: 37130114 PMCID: PMC10153721 DOI: 10.1371/journal.pone.0277759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/02/2022] [Indexed: 05/03/2023] Open
Abstract
Ultrasound-stimulated microbubbles (USMB) cause localized vascular effects and sensitize tumors to radiation therapy (XRT). We investigated acoustic parameter optimization for combining USMB and XRT. We treated breast cancer xenograft tumors with 500 kHz pulsed ultrasound at varying pressures (570 or 740 kPa), durations (1 to 10 minutes), and microbubble concentrations (0.01 to 1% (v/v)). Radiation therapy (2 Gy) was administered immediately or after a 6-hour delay. Histological staining of tumors 24 hours after treatment detected changes in cell morphology, cell death, and microvascular density. Significant cell death resulted at 570 kPa after a 1-minute exposure with 1% (v/v) microbubbles with or without XRT. However, significant microvascular disruption required higher ultrasound pressure and exposure duration greater than 5 minutes. Introducing a 6-hour delay between treatments (USMB and XRT) showed a similar tumor effect with no further improvement in response as compared to when XRT was delivered immediately after USMB.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | - Harini Kandavel
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | - Holliday Cartar
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ahmed El Kaffas
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
81
|
Mohanto N, Park YJ, Jee JP. Current perspectives of artificial oxygen carriers as red blood cell substitutes: a review of old to cutting-edge technologies using in vitro and in vivo assessments. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2023; 53:153-190. [PMID: 35935469 PMCID: PMC9344254 DOI: 10.1007/s40005-022-00590-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/02/2022] [Indexed: 01/06/2023]
Abstract
Background Several circumstances such as accidents, surgery, traumatic hemorrhagic shock, and other causalities cause major blood loss. Allogenic blood transfusion can be resuscitative for such conditions; however, it has numerous ambivalent effects, including supply shortage, needs for more time, cost for blood grouping, the possibility of spreading an infection, and short shelf-life. Hypoxia or ischemia causes heart failure, neurological problems, and organ damage in many patients. To address this emergent medical need for resuscitation and to treat hypoxic conditions as well as to enhance oxygen transportation, researchers aspire to achieve a robust technology aimed to develop safe and feasible red blood cell substitutes for effective oxygen transport. Area covered This review article provides an overview of the formulation, storage, shelf-life, clinical application, side effects, and current perspectives of artificial oxygen carriers (AOCs) as red blood cell substitutes. Moreover, the pre-clinical (in vitro and in vivo) assessments for the evaluation of the efficacy and safety of oxygen transport through AOCs are key considerations in this study. With the most significant technologies, hemoglobin- and perfluorocarbon-based oxygen carriers as well as other modern technologies, such as synthetically produced porphyrin-based AOCs and oxygen-carrying micro/nanobubbles, have also been elucidated. Expert opinion Both hemoglobin- and perfluorocarbon-based oxygen carriers are significant, despite having the latter acting as safeguards; they are cost-effective, facile formulations which penetrate small blood vessels and remove arterial blockages due to their nano-size. They also show better biocompatibility and longer half-life circulation than other similar technologies.
Collapse
Affiliation(s)
- Nijaya Mohanto
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 61452 Republic of Korea
| | - Young-Joon Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi, Republic of Korea
| | - Jun-Pil Jee
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 61452 Republic of Korea
| |
Collapse
|
82
|
Chapla R, Huynh KT, Schutt CE. Microbubble–Nanoparticle Complexes for Ultrasound-Enhanced Cargo Delivery. Pharmaceutics 2022; 14:pharmaceutics14112396. [PMID: 36365214 PMCID: PMC9698658 DOI: 10.3390/pharmaceutics14112396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble–nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid–polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin–avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble–nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.
Collapse
Affiliation(s)
- Rachel Chapla
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katherine T. Huynh
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
83
|
Bai MY, Chen TH, Wang YC, Lai YJ. Transformation of theranostic alginate-based microbubbles from raspberry-like to core-shell-like microbubbles and in vitro studies. RSC Adv 2022; 12:31943-31949. [PMID: 36380958 PMCID: PMC9641574 DOI: 10.1039/d2ra06298d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
In this study alginate-based microbubbles with a raspberry-like or core-shell-like morphology and with an average particle size of 553.6 ± 69.6 μm were synthesized; this was done through a novel procedure of transforming the structure with a 40 kHz ultrasonication which also stimulated the release of the components inside. Through the use of the electrospray technique in conjunction with agitation processes, components such as shikonin (SHK) and indocyanine green (ICG) were simultaneously encapsulated in alginate microbubbles to produce SHK-ICG alginate microbubbles; these microbubbles had half-maximal inhibitory concentrations of approximately 2.08 and 4.43 μM toward CP70 and SKOV3 ovarian cancer-cell lines, respectively, in an in vitro cell model. Moreover, these SHK-ICG alginate microbubbles enhanced brightness by 2.5 fold in ultrasound imaging relative to CaCl2 medium only. In conclusion, SHK-ICG alginate microbubbles have promise for use in theranostics.
Collapse
Affiliation(s)
- Meng-Yi Bai
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology No. 43, Keelung Rd, Sec. 4, Da'an Dist. Taipei City 10607 Taiwan
- Adjunct Appointment to the Departmnet of Biomedical Engineering, National Defense Medical Centre Taipei 11490 Taiwan
| | - Tsai-Hsuan Chen
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology No. 43, Keelung Rd, Sec. 4, Da'an Dist. Taipei City 10607 Taiwan
| | - Yu-Chi Wang
- Department of Obstetric and Gynecology, Tri-Service General Hospital, National Defense Medical Centre Taiwan
| | - Yu-Ju Lai
- Department of Obstetric and Gynecology, Tri-Service General Hospital, National Defense Medical Centre Taiwan
| |
Collapse
|
84
|
Zalloum IO, Paknahad AA, Kolios MC, Karshafian R, Tsai SSH. Controlled Shrinkage of Microfluidically Generated Microbubbles by Tuning Lipid Concentration. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:13021-13029. [PMID: 36260341 DOI: 10.1021/acs.langmuir.2c01439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Monodisperse microbubbles with diameters less than 10 μm are desirable in several ultrasound imaging and therapeutic delivery applications. However, conventional approaches to synthesize microbubbles, which are usually agitation-based, produce polydisperse bubbles that are less desirable because of their heterogeneous response when exposed to an ultrasound field. Microfluidics technology has the unique advantage of generating size-controlled monodisperse microbubbles, and it is now well established that the diameter of microfluidically made microbubbles can be tuned by varying the liquid flow rate, gas pressure, and dimensions of the microfluidic channel. It is also observed that once the microbubbles form, the bubbles shrink and eventually stabilize to a quasi-equilibrium diameter, and that the rate of stabilization is related to the lipid solution. However, how the lipid solution concentration affects the degree of bubble shrinkage, and the stable size of microbubbles, has not been thoroughly examined. Here, we investigate whether and how the lipid concentration affects the degree of microbubble shrinkage. Namely, we utilize a flow-focusing microfluidic geometry to generate monodisperse bubbles, and observe the effect of gas composition (2.5, 1.42, and 0.17 wt % octafluoropropane in nitrogen) and lipid concentration (1-16 mg/mL) on the degree of microbubble shrinkage. For the lipid system and gas utilized in these experiments, we observe a monotonic increase in the degree of microbubble shrinkage with decreasing lipid concentration, and no dependency on the gas composition. We hypothesize that the degree of shrinkage is related to lipid concentration by the self-assembly of lipids on the gas-liquid interface during bubble generation and subsequent lipid packing on the interface during shrinkage, which is arrested when a maximum packing density is achieved. We anticipate that this approach for creating and tuning the size of monodisperse microbubbles will find utility in biomedical applications, such as contrast-enhanced ultrasound imaging and ultrasound-triggered gene delivery.
Collapse
Affiliation(s)
- Intesar O Zalloum
- Department of Physics, 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, 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
| | - Ali A Paknahad
- Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan 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
| | - Michael C Kolios
- Department of Physics, 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, 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
| | - Raffi Karshafian
- Department of Physics, 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, 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
| | - Scott S H Tsai
- Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), A Partnership Between Toronto Metropolitan 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, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| |
Collapse
|
85
|
Yociss M, Brown K, Bruce M, Hoyt K. Amplitude modulation and baseband delay-multiply-and-sum beamforming for improved vessel visualization with volumetric contrast-enhanced ultrasound. 2022 IEEE INTERNATIONAL ULTRASONICS SYMPOSIUM (IUS) 2022:1-4. [DOI: 10.1109/ius54386.2022.9957183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Megan Yociss
- University of Texas at Dallas,Department of Bioengineering,Richardson,TX,USA
| | - Katherine Brown
- University of Texas at Dallas,Department of Bioengineering,Richardson,TX,USA
| | - Matthew Bruce
- Center for Industrial and Medical Ultrasound, Applied Physics Laboratory, University of Washington,Seattle,Washington,USA
| | - Kenneth Hoyt
- University of Texas at Dallas,Department of Bioengineering,Richardson,TX,USA
| |
Collapse
|
86
|
van den Broek SL, Shalgunov V, Herth MM. Transport of nanomedicines across the blood-brain barrier: Challenges and opportunities for imaging and therapy. BIOMATERIALS ADVANCES 2022; 141:213125. [PMID: 36182833 DOI: 10.1016/j.bioadv.2022.213125] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
The blood-brain barrier (BBB) is a protective and semipermeable border of endothelial cells that prevents toxins and foreign bodies to enter and damage the brain. Unfortunately, the BBB also hampers the development of pharmaceuticals targeting receptors, enzymes, or other proteins that lie beyond this barrier. Especially large molecules, such as monoclonal antibodies (mAbs) or nanoparticles, are prevented to enter the brain. The limited passage of these molecules partly explains why nanomedicines - targeting brain diseases - have not made it into the clinic to a great extent. As nanomedicines can target a wide range of targets including protein isoforms and oligomers or potentially deliver cytotoxic drugs safely to their targets, a pathway to smuggle nanomedicines into the brain would allow to treat brain diseases that are currently considered 'undruggable'. In this review, strategies to transport nanomedicines over the BBB will be discussed. Their challenges and opportunities will be highlighted with respect to their use for molecular imaging or therapies. Several strategies have been explored for this thus far. For example, carrier-mediated and receptor-mediated transcytosis (RMT), techniques to disrupt the BBB, nasal drug delivery or administering nanomedicines directly into the brain have been explored. RMT has been the most widely and successfully explored strategy. Recent work on the use of focused ultrasound based BBB opening has shown great promise. For example, successful delivery of mAbs into the brain has been achieved, even in a clinical setting. As nanomedicines bear the potential to treat incurable brain diseases, drug delivery technologies that can deliver nanomedicines into the brain will play an essential role for future treatment options.
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| |
Collapse
|
87
|
Hwang M, Zhang Z, Katz J, Freeman C, Kilbaugh T. Brain contrast-enhanced ultrasonography and elastography in infants. Ultrasonography 2022; 41:633-649. [PMID: 35879109 PMCID: PMC9532200 DOI: 10.14366/usg.21224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/20/2022] [Accepted: 03/30/2022] [Indexed: 11/19/2022] Open
Abstract
Advanced ultrasound techniques, including brain contrast-enhanced ultrasonography and elastography, are increasingly being explored to better understand infant brain health. While conventional brain ultrasonography provides a convenient, noninvasive means of assessing major intracranial pathologies, its value in revealing functional and physiologic insights into the brain lags behind advanced imaging techniques such as magnetic resonance imaging. In this regard, contrast-enhanced ultrasonography provides highly precise functional information on macrovascular and microvascular perfusion, while brain elastography offers information on brain stiffness that may be associated with relevant physiological factors of diagnostic, therapeutic, and/or prognostic utility. This review details the technical background, current understanding and utility, and future directions of these two emerging advanced ultrasound techniques for neonatal brain applications.
Collapse
Affiliation(s)
- Misun Hwang
- Department of Radiology, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zeng Zhang
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Joseph Katz
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Colbey Freeman
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Todd Kilbaugh
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
88
|
Hebert JF, Burfeind KG, Malinoski D, Hutchens MP. Molecular Mechanisms of Rhabdomyolysis-Induced Kidney Injury: From Bench to Bedside. Kidney Int Rep 2022; 8:17-29. [PMID: 36644345 PMCID: PMC9831947 DOI: 10.1016/j.ekir.2022.09.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 01/18/2023] Open
Abstract
Rhabdomyolysis-induced acute kidney injury (RIAKI) occurs following damage to the muscular sarcolemma sheath, resulting in the leakage of myoglobin and other metabolites that cause kidney damage. Currently, the sole recommended clinical treatment for RIAKI is aggressive fluid resuscitation, but other potential therapies, including pretreatments for those at risk for developing RIAKI, are under investigation. This review outlines the mechanisms and clinical significance of RIAKI, investigational treatments and their specific targets, and the status of ongoing research trials.
Collapse
Affiliation(s)
- Jessica F. Hebert
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA,Correspondence: Jessica F. Hebert, Oregon Health and Science University, Department of Anesthesiology and Perioperative Medicine, Portland, Oregon, USA.
| | - Kevin G. Burfeind
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Darren Malinoski
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA,Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| | - Michael P. Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA,Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| |
Collapse
|
89
|
Wang YC, Tian JY, Han YY, Liu YF, Chen SY, Guo FJ. Evaluation of the potential of ultrasound-mediated drug delivery for the treatment of ovarian cancer through preclinical studies. Front Oncol 2022; 12:978603. [PMID: 36132133 PMCID: PMC9483181 DOI: 10.3389/fonc.2022.978603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian cancer (OC) has the greatest mortality rate among gynecological cancers, with a five-year survival rate of <50%. Contemporary adjuvant chemotherapy mostly fails in the case of OCs that are refractory, metastatic, recurrent, and drug-resistant. Emerging ultrasound (US)-mediated technologies show remarkable promise in overcoming these challenges. Absorption of US waves by the tissue results in the generation of heat due to its thermal effect causing increased diffusion of drugs from the carriers and triggering sonoporation by increasing the permeability of the cancer cells. Certain frequencies of US waves could also produce a cavitation effect on drug-filled microbubbles (MBs, phospholipid bilayers) thereby generating shear force and acoustic streaming that could assist drug release from the MBs, and promote the permeability of the cell membrane. A new class of nanoparticles that carry therapeutic agents and are guided by US contrast agents for precision delivery to the site of the ovarian tumor has been developed. Phase-shifting of nanoparticles by US sonication has also been engineered to enhance the drug delivery to the ovarian tumor site. These technologies have been used for targeting the ovarian cancer stem cells and protein moieties that are particularly elevated in OCs including luteinizing hormone-releasing hormone, folic acid receptor, and vascular endothelial growth factor. When compared to healthy ovarian tissue, the homeostatic parameters at the tissue microenvironment including pH, oxygen levels, and glucose metabolism differ significantly in ovarian tumors. US-based technologies have been developed to take advantage of these tumor-specific alterations for precision drug delivery. Preclinical efficacy of US-based targeting of currently used clinical chemotherapies presented in this review has the potential for rapid human translation, especially for formulations that use all substances that are deemed to be generally safe by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Yi-Chao Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Jing-Yan Tian
- Department of Urology, The Second Division of the First Hospital of Jilin University, Changchun, China
| | - Ying-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yun-Fei Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Si-Yao Chen
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Feng-Jun Guo
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Feng-Jun Guo,
| |
Collapse
|
90
|
Plazonic F, LuTheryn G, Hind C, Clifford M, Gray M, Stride E, Glynne-Jones P, Hill M, Sutton JM, Carugo D. Bactericidal Effect of Ultrasound-Responsive Microbubbles and Sub-inhibitory Gentamicin against Pseudomonas aeruginosa Biofilms on Substrates With Differing Acoustic Impedance. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1888-1898. [PMID: 35798625 DOI: 10.1016/j.ultrasmedbio.2022.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
The aim of this research was to explore the interaction between ultrasound-activated microbubbles (MBs) and Pseudomonas aeruginosa biofilms, specifically the effects of MB concentration, ultrasound exposure and substrate properties on bactericidal efficacy. Biofilms were grown using a Centre for Disease Control (CDC) bioreactor on polypropylene or stainless-steel coupons as acoustic analogues for soft and hard tissue, respectively. Biofilms were treated with different concentrations of phospholipid-shelled MBs (107-108 MB/mL), a sub-inhibitory concentration of gentamicin (4 µg/mL) and 1-MHz ultrasound with a continuous or pulsed (100-kHz pulse repetition frequency, 25% duty cycle, 0.5-MPa peak-to-peak pressure) wave. The effect of repeated ultrasound exposure with intervals of either 15- or 60-min was also investigated. With polypropylene coupons, the greatest bactericidal effect was achieved with 2 × 5 min of pulsed ultrasound separated by 60 min and a microbubble concentration of 5 × 107 MBs/mL. A 0.76 log (83%) additional reduction in the number of bacteria was achieved compared with the use of an antibiotic alone. With stainless-steel coupons, a 67% (0.46 log) reduction was obtained under the same exposure conditions, possibly due to enhancement of a standing wave field which inhibited MB penetration in the biofilm. These findings demonstrate the importance of treatment parameter selection in antimicrobial applications of MBs and ultrasound in different tissue environments.
Collapse
Affiliation(s)
- Filip Plazonic
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Gareth LuTheryn
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK; Department of Pharmaceutics, School of Pharmacy, University College London, London, UK; National Biofilms Innovation Centre, University of Southampton, Southampton, UK
| | - Charlotte Hind
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Melanie Clifford
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Michael Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Peter Glynne-Jones
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Martyn Hill
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - J Mark Sutton
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK; Institute of Pharmaceutical Science, King's College London, London, UK
| | - Dario Carugo
- Department of Pharmaceutics, School of Pharmacy, University College London, London, UK.
| |
Collapse
|
91
|
Khan AH, Jiang X, Kaushik A, Nair HS, Edirisinghe M, Mercado-Shekhar KP, Shekhar H, Dalvi SV. Combining Ultrasound and Capillary-Embedded T-Junction Microfluidic Devices to Scale Up the Production of Narrow-Sized Microbubbles through Acoustic Fragmentation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:10288-10304. [PMID: 35943351 DOI: 10.1021/acs.langmuir.2c01676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microbubbles are tiny gas-filled bubbles that have a variety of applications in ultrasound imaging and therapeutic drug delivery. Microbubbles can be synthesized using a number of techniques including sonication, amalgamation, and saline shaking. These approaches can produce highly concentrated microbubble suspensions but offer minimal control over the size and polydispersity of the microbubbles. One of the simplest and effective methods for producing monodisperse microbubbles is capillary-embedded T-junction microfluidic devices, which offer great control over the microbubble size. However, lower production rates (∼200 bubbles/s) and large microbubble sizes (∼300 μm) limit the applicability of such devices for biomedical applications. To overcome the limitations of these technologies, we demonstrate in this work an alternative approach to combine a capillary-embedded T-junction device with ultrasound to enhance the generation of narrow-sized microbubbles in aqueous suspensions. Two T-junction microfluidic devices were connected in parallel and combined with an ultrasonic horn to produce lipid-coated SF6 core microbubbles in the size range of 1-8 μm. The rate of microbubble production was found to increase from 180 microbubbles/s in the absence of ultrasound to (6.5 ± 1.2) × 106 bubble/s in the presence of ultrasound (100% ultrasound amplitude). When stored in a closed environment, the microbubbles were observed to be stable for up to 30 days, with the concentration of the microbubble suspension decreasing from ∼2.81 × 109/mL to ∼2.3 × 106/mL and the size changing from 1.73 ± 0.2 to 1.45 ± 0.3 μm at the end of 30 days. The acoustic response of these microbubbles was examined using broadband attenuation spectroscopy, and flow phantom imaging was performed to determine the ability of these microbubble suspensions to enhance the contrast relative to the surrounding tissue. Overall, this approach of coupling ultrasound with microfluidic parallelization enabled the continuous production of stable microbubbles at high production rates and low polydispersity using simple T-junction devices.
Collapse
Affiliation(s)
- Aaqib H Khan
- Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Xinyue Jiang
- Department of Mechanical Engineering, University College London (UCL), London WC1E 7JE, U.K
| | - Anuj Kaushik
- Electrical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Hari S Nair
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London (UCL), London WC1E 7JE, U.K
| | - Karla P Mercado-Shekhar
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Himanshu Shekhar
- Electrical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Sameer V Dalvi
- Chemical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
92
|
Barmin RA, Dasgupta A, Bastard C, De Laporte L, Rütten S, Weiler M, Kiessling F, Lammers T, Pallares RM. Engineering the Acoustic Response and Drug Loading Capacity of PBCA-Based Polymeric Microbubbles with Surfactants. Mol Pharm 2022; 19:3256-3266. [PMID: 35905480 DOI: 10.1021/acs.molpharmaceut.2c00416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gas-filled microbubbles (MB) are routinely used in the clinic as ultrasound contrast agents. MB are also increasingly explored as drug delivery vehicles based on their ultrasound stimuli-responsiveness and well-established shell functionalization routes. Broadening the range of MB properties can enhance their performance in both imaging and drug delivery applications. This can be promoted by systematically varying the reagents used in the synthesis of MB, which in the case of polymeric MB include surfactants. We therefore set out to study the effect of key surfactant characteristics, such as the chemical structure, molecular weight, and hydrophilic-lipophilic balance on the formation of poly(butyl cyanoacrylate) (PBCA) MB, as well as on their properties, including shell thickness, drug loading capacity, ultrasound contrast, and acoustic stability. Two different surfactant families (i.e., Triton X and Tween) were employed, which show opposite molecular weight vs hydrophilic-lipophilic balance trends. For both surfactant types, we found that the shell thickness of PBCA MB increased with higher-molecular-weight surfactants and that the resulting MB with thicker shells showed higher drug loading capacities and acoustic stability. Furthermore, the higher proportion of smaller polymer chains of the Triton X-based MB (as compared to those of the Tween-based ones) resulted in lower polymer entanglement, improving drug loading capacity and ultrasound contrast response. These findings open up new avenues to fine-tune the shell properties of polymer-based MB for enhanced ultrasound imaging and drug delivery applications.
Collapse
Affiliation(s)
- Roman A Barmin
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Céline Bastard
- DWI - Leibniz Institute for Interactive Materials, 52074 Aachen, Germany.,Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany.,Institute of Applied Medical Engineering, Department of Advanced Materials for Biomedicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Laura De Laporte
- DWI - Leibniz Institute for Interactive Materials, 52074 Aachen, Germany.,Institute for Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany.,Institute of Applied Medical Engineering, Department of Advanced Materials for Biomedicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Stephan Rütten
- Electron Microscope Facility, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Marek Weiler
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
93
|
Caraway CA, Gaitsch H, Wicks EE, Kalluri A, Kunadi N, Tyler BM. Polymeric Nanoparticles in Brain Cancer Therapy: A Review of Current Approaches. Polymers (Basel) 2022; 14:2963. [PMID: 35890738 PMCID: PMC9322801 DOI: 10.3390/polym14142963] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
Translation of novel therapies for brain cancer into clinical practice is of the utmost importance as primary brain tumors are responsible for more than 200,000 deaths worldwide each year. While many research efforts have been aimed at improving survival rates over the years, prognosis for patients with glioblastoma and other primary brain tumors remains poor. Safely delivering chemotherapeutic drugs and other anti-cancer compounds across the blood-brain barrier and directly to tumor cells is perhaps the greatest challenge in treating brain cancer. Polymeric nanoparticles (NPs) are powerful, highly tunable carrier systems that may be able to overcome those obstacles. Several studies have shown appropriately-constructed polymeric NPs cross the blood-brain barrier, increase drug bioavailability, reduce systemic toxicity, and selectively target central nervous system cancer cells. While no studies relating to their use in treating brain cancer are in clinical trials, there is mounting preclinical evidence that polymeric NPs could be beneficial for brain tumor therapy. This review includes a variety of polymeric NPs and how their associated composition, surface modifications, and method of delivery impact their capacity to improve brain tumor therapy.
Collapse
Affiliation(s)
- Chad A. Caraway
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Hallie Gaitsch
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- NIH-Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Elizabeth E. Wicks
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- University of Mississippi School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Anita Kalluri
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Navya Kunadi
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Betty M. Tyler
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| |
Collapse
|
94
|
Estifeeva TM, Barmin RA, Rudakovskaya PG, Nechaeva AM, Luss AL, Mezhuev YO, Chernyshev VS, Krivoborodov EG, Klimenko OA, Sindeeva OA, Demina PA, Petrov KS, Chuprov-Netochin RN, Fedotkina EP, Korotchenko OE, Sencha EA, Sencha AN, Shtilman MI, Gorin DA. Hybrid (Bovine Serum Albumin)/Poly( N-vinyl-2-pyrrolidone- co-acrylic acid)-Shelled Microbubbles as Advanced Ultrasound Contrast Agents. ACS APPLIED BIO MATERIALS 2022; 5:3338-3348. [PMID: 35791763 DOI: 10.1021/acsabm.2c00331] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Microbubbles are routinely used ultrasound contrast agents in the clinic. While a soft protein shell is commercially preferable for imaging purposes, a rigid polymer shell demonstrates prolonged agent stability. Hence, combining polymers and proteins in one shell composition can advance microbubble properties. We formulated the hybrid "protein-copolymer" microbubble shell with a complex of bovine serum albumin and an amphiphilic copolymer of N-vinyl-2-pyrrolidone and acrylic acid. The resulting microbubbles demonstrated advanced physicochemical and acoustic properties, preserving in vitro biocompatibility. Adjusting the mass ratio between protein and copolymer allowed fine tuning of the microbubble properties of concentration (by two orders, up to 1010 MBs/mL), mean size (from 0.8 to 5 μm), and shell thickness (from 28 to 50 nm). In addition, the minimum air-liquid surface tension for the "protein-copolymer" solution enabled the highest bubble concentration. At the same time, a higher copolymer amount in the bubble shell increased the bubble size and tuned duration and intensity of the contrast during an ultrasound procedure. Demonstrated results exemplify the potential of the hybrid "protein-polymer" microbubble shell, allowing tailoring of microbubble properties for image-guided applications, combining advances of each material involved in the formulation.
Collapse
Affiliation(s)
- Tatyana M Estifeeva
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Roman A Barmin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Nobel str. 3, 121205 Moscow, Russia
| | - Polina G Rudakovskaya
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Nobel str. 3, 121205 Moscow, Russia
| | - Anna M Nechaeva
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Anna L Luss
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Yaroslav O Mezhuev
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Vasiliy S Chernyshev
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Nobel str. 3, 121205 Moscow, Russia
| | - Efrem G Krivoborodov
- Institute of Chemistry and Sustainable Development, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Oleg A Klimenko
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Nobel str. 3, 121205 Moscow, Russia.,P.N. Lebedev Physical Institute of the Russian Academy of Sciences, Leninskiy Prospekt 53, 119991 Moscow, Russia
| | - Olga A Sindeeva
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Nobelya Str. 3, 121205 Moscow, Russia
| | - Polina A Demina
- Federal Scientific Research Centre ″Crystallography and Photonics″ of the Russian Academy of Sciences, Leninskiy avenue 59, 119333 Moscow, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997 Moscow, Russia
| | - Kirill S Petrov
- Hadassah Medical Moscow, Bolshoy Boulevard 46, 121205 Moscow, Russia
| | - Roman N Chuprov-Netochin
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky Lane 9, 141700 Dolgoprudny, Moscow Region, Russia
| | - Elena P Fedotkina
- Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, Akademika Oparina str. 4, 117198 Moscow, Russia
| | - Olga E Korotchenko
- Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, Akademika Oparina str. 4, 117198 Moscow, Russia
| | - Ekaterina A Sencha
- Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, Akademika Oparina str. 4, 117198 Moscow, Russia
| | - Alexander N Sencha
- Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, Akademika Oparina str. 4, 117198 Moscow, Russia
| | - Mikhail I Shtilman
- Department of Biomaterials, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Nobel str. 3, 121205 Moscow, Russia
| |
Collapse
|
95
|
Faiz K, Lam FC, Chen J, Kasper EM, Salehi F. The Emerging Applications of Nanotechnology in Neuroimaging: A Comprehensive Review. Front Bioeng Biotechnol 2022; 10:855195. [PMID: 35875504 PMCID: PMC9297121 DOI: 10.3389/fbioe.2022.855195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/06/2022] [Indexed: 11/19/2022] Open
Abstract
Neuroimaging modalities such as computer tomography and magnetic resonance imaging have greatly improved in their ability to achieve higher spatial resolution of neurovascular and soft tissue neuroanatomy, allowing for increased accuracy in the diagnosis of neurological conditions. However, the use of conventional contrast agents that have short tissue retention time and associated renal toxicities, or expensive radioisotope tracers that are not widely available, continue to limit the sensitivity of these imaging modalities. Nanoparticles can potentially address these shortcomings by enhancing tissue retention and improving signal intensity in the brain and neural axis. In this review, we discuss the use of different types of nanotechnology to improve the detection, diagnosis, and treatment of a wide range of neurological diseases.
Collapse
Affiliation(s)
- Khunza Faiz
- Department of Radiology, McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
| | - Fred C. Lam
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, United States
- Division of Neurosurgery, Saint Elizabeth Medical Center, Brighton, MA, United States
- *Correspondence: Fred C. Lam, ; Ekkehard M. Kasper, ; Fateme Salehi,
| | - Jay Chen
- Department of Radiology, McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
| | - Ekkehard M. Kasper
- Division of Neurosurgery, Saint Elizabeth Medical Center, Brighton, MA, United States
- *Correspondence: Fred C. Lam, ; Ekkehard M. Kasper, ; Fateme Salehi,
| | - Fateme Salehi
- Department of Radiology, McMaster University Faculty of Health Sciences, Hamilton, ON, Canada
- *Correspondence: Fred C. Lam, ; Ekkehard M. Kasper, ; Fateme Salehi,
| |
Collapse
|
96
|
Sharma D, Czarnota GJ. Involvement of Ceramide Signalling in Radiation-Induced Tumour Vascular Effects and Vascular-Targeted Therapy. Int J Mol Sci 2022; 23:ijms23126671. [PMID: 35743121 PMCID: PMC9223569 DOI: 10.3390/ijms23126671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Sphingolipids are well-recognized critical components in several biological processes. Ceramides constitute a class of sphingolipid metabolites that are involved in important signal transduction pathways that play key roles in determining the fate of cells to survive or die. Ceramide accumulated in cells causes apoptosis; however, ceramide metabolized to sphingosine promotes cell survival and angiogenesis. Studies suggest that vascular-targeted therapies increase endothelial cell ceramide resulting in apoptosis that leads to tumour cure. Specifically, ultrasound-stimulated microbubbles (USMB) used as vascular disrupting agents can perturb endothelial cells, eliciting acid sphingomyelinase (ASMase) activation accompanied by ceramide release. This phenomenon results in endothelial cell death and vascular collapse and is synergistic with other antitumour treatments such as radiation. In contrast, blocking the generation of ceramide using multiple approaches, including the conversion of ceramide to sphingosine-1-phosphate (S1P), abrogates this process. The ceramide-based cell survival "rheostat" between these opposing signalling metabolites is essential in the mechanotransductive vascular targeting following USMB treatment. In this review, we aim to summarize the past and latest findings on ceramide-based vascular-targeted strategies, including novel mechanotransductive methodologies.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
- Correspondence: ; Tel.: +1-416-480-6100 (ext. 89533)
| | - Gregory J. Czarnota
- Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
97
|
Zhu Y, Peng N, Wang J, Jin Z, Zhu L, Wang Y, Chen S, Hu Y, Zhang T, Song Q, Xie F, Yan L, Li Y, Xiao J, Li X, Jiang B, Peng J, Wang Y, Luo Y. Peripheral nerve defects repaired with autogenous vein grafts filled with platelet-rich plasma and active nerve microtissues and evaluated by novel multimodal ultrasound techniques. Biomater Res 2022; 26:24. [PMID: 35690849 PMCID: PMC9188244 DOI: 10.1186/s40824-022-00264-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Developing biocompatible nerve conduits that accelerate peripheral nerve regeneration, lengthening and functional recovery remains a challenge. The combined application of nerve microtissues and platelet-rich plasma (PRP) provides abundant Schwann cells (SCs) and various natural growth factors and can compensate for the deficiency of SCs in the nerve bridge, as well as the limitations of applying a single type of growth factor. Multimodal ultrasound evaluation can provide additional information on the stiffness and microvascular flow perfusion of the tissue. This study was designed to investigate the effectiveness of a novel tissue-engineered nerve graft composed of an autogenous vein, nerve microtissues and PRP in reconstructing a 12-mm tibial nerve defect and to explore the value of multimodal ultrasound techniques in evaluating the prognosis of nerve repair. METHODS In vitro, nerve microtissue activity was first investigated, and the effects on SC proliferation, migration, factor secretion, and axonal regeneration of dorsal root ganglia (DRG) were evaluated by coculture with nerve microtissues and PRP. In vivo, seventy-five rabbits were equally and randomly divided into Hollow, PRP, Micro-T (Microtissues), Micro-T + PRP and Autograft groups. By analysing the neurological function, electrophysiological recovery, and the comparative results of multimodal ultrasound and histological evaluation, we investigated the effect of these new nerve grafts in repairing tibial nerve defects. RESULTS Our results showed that the combined application of nerve microtissues and PRP could significantly promote the proliferation, secretion and migration of SCs and the regeneration of axons in the early stage. The Micro-T + PRP group and Autograft groups exhibited the best nerve repair 12 weeks postoperatively. In addition, the changes in target tissue stiffness and microvascular perfusion on multimodal ultrasound (shear wave elastography; contrast-enhanced ultrasonography; Angio PlaneWave UltrasenSitive, AngioPLUS) were significantly correlated with the histological results, such as collagen area percentage and VEGF expression, respectively. CONCLUSION Our novel tissue-engineered nerve graft shows excellent efficacy in repairing 12-mm defects of the tibial nerve in rabbits. Moreover, multimodal ultrasound may provide a clinical reference for prognosis by quantitatively evaluating the stiffness and microvescular flow of nerve grafts and targeted muscles.
Collapse
Affiliation(s)
- Yaqiong Zhu
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China.,Beijing Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing, China.,Key Lab of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, China.,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China
| | - Nan Peng
- Department of Geriatric Rehabilitation, The Second Center of Chinese PLA General Hospital, Beijing, China
| | - Jing Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui Province, China
| | - Zhuang Jin
- General hospital of Northern Theater Command, Liaoning, China
| | - Lianhua Zhu
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing, China.,Key Lab of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, China
| | - Siming Chen
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Yongqiang Hu
- Department of Anesthesiology, JiangXi PingXiang People's Hospital, Jiangxi, China
| | - Tieyuan Zhang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing, China.,Key Lab of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, China
| | - Qing Song
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Fang Xie
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Lin Yan
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Yingying Li
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Jing Xiao
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Xinyang Li
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Bo Jiang
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Chinese PLA General Hospital, Beijing, China. .,Key Lab of Musculoskeletal Trauma & War Injuries, Chinese PLA General Hospital, Beijing, China.
| | - Yuexiang Wang
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China.
| | - Yukun Luo
- Departments of Ultrasound, The First Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
98
|
Jung O, Thomas A, Burks SR, Dustin ML, Frank JA, Ferrer M, Stride E. Neuroinflammation associated with ultrasound-mediated permeabilization of the blood-brain barrier. Trends Neurosci 2022; 45:459-470. [PMID: 35461727 PMCID: PMC9117477 DOI: 10.1016/j.tins.2022.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/17/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
The blood-brain barrier (BBB) continues to represent one of the most significant challenges for successful drug-based treatments of neurological disease. Mechanical modulation of the BBB using focused ultrasound (FUS) and microbubbles (MBs) has shown considerable promise in enhancing the delivery of therapeutics to the brain, but questions remain regarding possible long-term effects of such forced disruption. This review examines the evidence for inflammation associated with ultrasound-induced BBB disruption and potential strategies for managing such inflammatory effects to improve both the efficacy and safety of therapeutic ultrasound in neurological applications.
Collapse
Affiliation(s)
- Olive Jung
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK; 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Alec Thomas
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Scott R Burks
- The Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Michael L Dustin
- Nuffield Department of Orthopedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Joseph A Frank
- The Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Eleanor Stride
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
| |
Collapse
|
99
|
Spiekhout S, Voorneveld J, van Elburg B, Renaud G, Segers T, Lajoinie GPR, Versluis M, Verweij MD, de Jong N, Bosch JG. Time-resolved absolute radius estimation of vibrating contrast microbubbles using an acoustical camera. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2022; 151:3993. [PMID: 35778226 DOI: 10.1121/10.0011619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/20/2022] [Indexed: 06/15/2023]
Abstract
Ultrasound (US) contrast agents consist of microbubbles ranging from 1 to 10 μm in size. The acoustical response of individual microbubbles can be studied with high-frame-rate optics or an "acoustical camera" (AC). The AC measures the relative microbubble oscillation while the optical camera measures the absolute oscillation. In this article, the capabilities of the AC are extended to measure the absolute oscillations. In the AC setup, microbubbles are insonified with a high- (25 MHz) and low-frequency US wave (1-2.5 MHz). Other than the amplitude modulation (AM) from the relative size change of the microbubble (employed in Renaud, Bosch, van der Steen, and de Jong (2012a). "An 'acoustical camera' for in vitro characterization of contrast agent microbubble vibrations," Appl. Phys. Lett. 100(10), 101911, the high-frequency response from individual vibrating microbubbles contains a phase modulation (PM) from the microbubble wall displacement, which is the extension described here. The ratio of PM and AM is used to determine the absolute radius, R0. To test this sizing, the size distributions of two monodisperse microbubble populations ( R = 2.1 and 3.5 μm) acquired with the AC were matched to the distribution acquired with a Coulter counter. As a result of measuring the absolute size of the microbubbles, this "extended AC" can capture the full radial dynamics of single freely floating microbubbles with a throughput of hundreds of microbubbles per hour.
Collapse
Affiliation(s)
- Sander Spiekhout
- Biomedical Engineering, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jason Voorneveld
- Biomedical Engineering, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Benjamin van Elburg
- Physics of Fluids Group, Department of Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, The Netherlands
| | - Guillaume Renaud
- Laboratory of Medical Imaging, Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| | - Tim Segers
- Biomedical and Environmental Sensor Systems (BIOS) Lab-on-a-Chip Group, Max Planck Center for Complex Fluid Dynamics, University of Twente, Enschede, The Netherlands
| | - Guillaume P R Lajoinie
- Physics of Fluids Group, Department of Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, The Netherlands
| | - Michel Versluis
- Physics of Fluids Group, Department of Science and Technology, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, Enschede, The Netherlands
| | - Martin D Verweij
- Laboratory of Medical Imaging, Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| | - Nico de Jong
- Laboratory of Medical Imaging, Department of Imaging Physics, Delft University of Technology, Delft, The Netherlands
| | - Johannes G Bosch
- Biomedical Engineering, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
100
|
Lv W, Liu Y, Li S, Lv L, Lu H, Xin H. Advances of nano drug delivery system for the theranostics of ischemic stroke. J Nanobiotechnology 2022; 20:248. [PMID: 35641956 PMCID: PMC9153106 DOI: 10.1186/s12951-022-01450-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/05/2022] [Indexed: 02/07/2023] Open
Abstract
From the global perspective, stroke refers to a highly common cause of disability and death. Ischemic stroke (IS), attributed to blood vessel blockage, preventing the flow of blood to brain, acts as the most common form of stroke. Thus far, thrombolytic therapy is the only clinical treatment for IS with the approval from the FDA. Moreover, the physiology barrier complicates therapeutically and diagnostically related intervention development of IS. Accordingly, developing efficient and powerful curative approaches for IS diagnosis and treatment is urgently required. The advent of nanotechnology has brought dawn and hope to better curative and imaging forms for the management of IS. This work reviews the recent advances and challenges correlated with the nano drug delivery system for IS therapy and diagnosis. The overview of the current knowledge of the important molecular pathological mechanisms in cerebral ischemia and how the drugs cross the blood brain barrier will also be briefly summarized.
Collapse
Affiliation(s)
- Wei Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Yijiao Liu
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Shengnan Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Lingyan Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, 214400, Jiangyin, China
| | - Hongdan Lu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China.
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|