1
|
Bayram C, Ozturk S, Karaosmanoglu B, Gultekinoglu M, Taskiran EZ, Ulubayram K, Majd H, Ahmed J, Edirisinghe M. Microfluidic Fabrication of Gelatin-Nano Hydroxyapatite Scaffolds for Enhanced Control of Pore Size Distribution and Osteogenic Differentiation of Dental Pulp Stem Cells. Macromol Biosci 2024; 24:e2400279. [PMID: 39388643 DOI: 10.1002/mabi.202400279] [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: 06/13/2024] [Revised: 09/13/2024] [Indexed: 10/12/2024]
Abstract
The combination of gelatin and hydroxyapatite (HA) has emerged as a promising strategy in dental tissue engineering due to its favorable biocompatibility, mechanical properties, and ability to support cellular activities essential for tissue regeneration, rendering them ideal components for hard tissue applications. Besides, precise control over interconnecting porosity is of paramount importance for tissue engineering materials. Conventional methods for creating porous scaffolds frequently encounter difficulties in regulating pore size distribution. This study demonstrates the fabrication of gelatin-nano HA scaffolds with uniform porosity using a T-type junction microfluidic device in a single-step process. Significant improvements in control over the pore size distribution are achieved by regulating the flow parameters, resulting in effective and time-efficient manufacturing comparable in quality to the innovative 3D bioprinting techniques. The overall porosity of the scaffolds exceeded 60%, with a remarkably narrow size distribution. The incorporation of nano-HAinto 3D porous gelatin scaffolds successfully induced osteogenic differentiation in stem cells at both the protein and gene levels, as evidenced by the significant increase in osteocalcin (OCN), an important marker of osteogenic differentiation. The OCN levels are 26 and 43 times higher for gelatin and gelatin-HA scaffolds, respectively, compared to the control group.
Collapse
Affiliation(s)
- Cem Bayram
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, 06800, Turkey
| | - Sukru Ozturk
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, 06100, Turkey
| | - Beren Karaosmanoglu
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Merve Gultekinoglu
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, 06800, Turkey
| | - Ekim Z Taskiran
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, 06100, Turkey
| | - Kezban Ulubayram
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara, 06100, Turkey
| | - Hamta Majd
- Department of Mechanical Engineering, University College London (UCL), London, WC1E7JE, UK
| | - Jubair Ahmed
- Department of Mechanical Engineering, University College London (UCL), London, WC1E7JE, UK
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London (UCL), London, WC1E7JE, UK
| |
Collapse
|
2
|
Wang J, Luo T, Chen J, Liu Z, Wang J, Zhang X, Li H, Ma Y, Zhang F, Ju H, Wang W, Wang Y, Zhu Q. Enhancement of Tumor Perfusion and Antiangiogenic Therapy in Murine Models of Clear Cell Renal Cell Carcinoma Using Ultrasound-Stimulated Microbubbles. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:680-689. [PMID: 38311538 DOI: 10.1016/j.ultrasmedbio.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/06/2024]
Abstract
OBJECTIVE To explore the effect of ultrasound-stimulated microbubble cavitation (USMC) on enhancing antiangiogenic therapy in clear cell renal cell carcinoma. MATERIALS AND METHODS We explored the effects of USMC with different mechanical indices (MIs) on tumor perfusion, 36 786-O tumor-bearing nude mice were randomly assigned into four groups: (i) control group, (ii) USMC0.25 group (MI = 0.25), (iii) USMC1.4 group (MI = 1.4) (iv) US1.4 group (MI = 1.4). Tumor perfusion was assessed by contrast-enhanced ultrasound (CEUS) before the USMC treatment and 30 min, 4h and 6h after the USMC treatment, respectively. Then we evaluated vascular normalization(VN) induced by low-MI (0.25) USMC treatment, 12 tumor-bearing nude mice were randomly divided into two groups: (i) control group (ii) USMC0.25 group. USMC treatment was performed, and tumor microvascular imaging and blood perfusion were analyzed by MicroFlow imaging (MFI) and CEUS 30 min after each treatment. In combination therapy, a total of 144 tumor-bearing nude mice were randomly assigned to six groups (n = 24): (i) control group, (ii) USMC1.4 group, (iii) USMC0.25 group, (iv) bevacizumab(BEV) group, (v) USMC1.4 +BEV group, (vi) USMC0.25 +BEV group. BEV was injected on the 6th, 10th, 14th, and 18th d after the tumors were inoculated, while USMC treatment was performed 24 h before and after every BEV administration. We examined the effects of the combination therapy through a series of experiments. RESULTS Tumor blood perfusion enhanced by USMC with low MI (0.25)could last for more than 6h, inducing tumor VN and promoting drug delivery. Compared with other groups, USMC0.25+BEV combination therapy had the strongest inhibition on tumor growth, led to the longest survival time of the mice. CONCLUSION The optimized USMC is a promising therapeutic approach that can be combined with antiangiogenic therapy to combat tumor progression.
Collapse
Affiliation(s)
- Juan Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tingting Luo
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jianghong Chen
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Juan Wang
- Department of Pathology,The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Hebei Province Key Laboratory of Environment and Human Health, Shijiiazhuang, Hebei, China
| | - Hui Li
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yulin Ma
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fan Zhang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongjuan Ju
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wengang Wang
- Department of Abdominal Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueheng Wang
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
3
|
Wu Y, Li J, Shu L, Tian Z, Wu S, Wu Z. Ultrasound combined with microbubble mediated immunotherapy for tumor microenvironment. Front Pharmacol 2024; 15:1304502. [PMID: 38487163 PMCID: PMC10937735 DOI: 10.3389/fphar.2024.1304502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/11/2024] [Indexed: 03/17/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in dynamically regulating the progress of cancer and influencing the therapeutic results. Targeting the tumor microenvironment is a promising cancer treatment method in recent years. The importance of tumor immune microenvironment regulation by ultrasound combined with microbubbles is now widely recognized. Ultrasound and microbubbles work together to induce antigen release of tumor cell through mechanical or thermal effects, promoting antigen presentation and T cells' recognition and killing of tumor cells, and improve tumor immunosuppression microenvironment, which will be a breakthrough in improving traditional treatment problems such as immune checkpoint blocking (ICB) and himeric antigen receptor (CAR)-T cell therapy. In order to improve the therapeutic effect and immune regulation of TME targeted tumor therapy, it is necessary to develop and optimize the application system of microbubble ultrasound for organs or diseases. Therefore, the combination of ultrasound and microbubbles in the field of TME will continue to focus on developing more effective strategies to regulate the immunosuppression mechanisms, so as to activate anti-tumor immunity and/or improve the efficacy of immune-targeted drugs, At present, the potential value of ultrasound combined with microbubbles in TME targeted therapy tumor microenvironment targeted therapy has great potential, which has been confirmed in the experimental research and application of breast cancer, colon cancer, pancreatic cancer and prostate cancer, which provides a new alternative idea for clinical tumor treatment. This article reviews the research progress of ultrasound combined with microbubbles in the treatment of tumors and their application in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Zuohui Wu
- Department of Ultrasound, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
4
|
Pellow C, Jafari Sojahrood A, Zhao X, Kolios MC, Exner AA, Goertz DE. Synchronous Intravital Imaging and Cavitation Monitoring of Antivascular Focused Ultrasound in Tumor Microvasculature Using Monodisperse Low Boiling Point Nanodroplets. ACS NANO 2024; 18:410-427. [PMID: 38147452 PMCID: PMC10786165 DOI: 10.1021/acsnano.3c07711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.
Collapse
Affiliation(s)
- Carly Pellow
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
| | - Amin Jafari Sojahrood
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Xiaoxiao Zhao
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| | - Michael C. Kolios
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Agata A. Exner
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - David E. Goertz
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| |
Collapse
|
5
|
Zhao P, Peng Y, Wang Y, Hu Y, Qin J, Li D, Yan K, Fan Z. Mechanistic study of ultrasound and microbubble enhanced cancer therapy in a 3D vascularized microfluidic cancer model. ULTRASONICS SONOCHEMISTRY 2023; 101:106709. [PMID: 38043461 PMCID: PMC10704430 DOI: 10.1016/j.ultsonch.2023.106709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Accumulating evidence has shown that ultrasound exposure combined with microbubbles can enhance cancer therapy. However, the underlying mechanisms at the tissue level have not been fully understood yet. The conventional cell culture in vitro lacks complex structure and interaction, while animal studies cannot provide micron-scale dynamic information. To bridge the gap, we designed and assembled a 3D vascularized microfluidic cancer model, particularly suitable for ultrasound and microbubble involved mechanistic studies. Using this model, we first studied SonoVue microbubble traveling dynamics in 3D tissue structure, then resolved SonoVue microbubble cavitation dynamics in tissue mimicking agarose gels at a frame rate of 0.675 M fps, and finally explored the impacts of ultrasound and microbubbles on cancer cell spheroids. Our results demonstrate that microbubble penetration in agarose gel was enhanced by increasing microbubble concentration, flow rate and decreasing viscosity of the gel, and little affected by mild acoustic radiation force. SonoVue microbubble exhibited larger expansion amplitudes in 2 %(w/v) agarose gels than in water, which can be explained theoretically by the relaxation of the cavitation medium. The immediate impacts of ultrasound and SonoVue microbubbles to cancer cell spheroids in the 3D tissue model included improved cancer cell spheroid penetration in micron-scale and sparse direct permanent cancer cell damage. Our study provides new insights of the mechanisms for ultrasound and microbubble enhanced cancer therapy at the tissue level.
Collapse
Affiliation(s)
- Pu Zhao
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Yingxiao Peng
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Yanjie Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yi Hu
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Jixing Qin
- State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing 100190, China
| | - Dachao Li
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Kun Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Ultrasound, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Zhenzhen Fan
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China; State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
6
|
Chen P, Zhang P, Shah NH, Cui Y, Wang Y. A Comprehensive Review of Inorganic Sonosensitizers for Sonodynamic Therapy. Int J Mol Sci 2023; 24:12001. [PMID: 37569377 PMCID: PMC10418994 DOI: 10.3390/ijms241512001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Sonodynamic therapy (SDT) is an emerging non-invasive cancer treatment method in the field of nanomedicine, which has the advantages of deep penetration, good therapeutic efficacy, and minimal damage to normal tissues. Sonosensitizers play a crucial role in the process of SDT, as their structure and properties directly determine the treatment outcome. Inorganic sonosensitizers, with their high stability and longer circulation time in the human body, have great potential in SDT. In this review, the possible mechanisms of SDT including the ultrasonic cavitation, reactive oxygen species generation, and activation of immunity are briefly discussed. Then, the latest research progress on inorganic sonosensitizers is systematically summarized. Subsequently, strategies for optimizing treatment efficacy are introduced, including combination therapy and image-guided therapy. The challenges and future prospects of sonodynamic therapy are discussed. It is hoped that this review will provide some guidance for the screening of inorganic sonosensitizers.
Collapse
Affiliation(s)
- Peng Chen
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Ping Zhang
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Navid Hussain Shah
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
| | - Yanyan Cui
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China; (P.C.); (P.Z.); (N.H.S.)
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| |
Collapse
|
7
|
Imran KM, Tintera B, Morrison HA, Tupik JD, Nagai-Singer MA, Ivester H, Council-Troche M, Edwards M, Coutermarsh-Ott S, Byron C, Clark-Deener S, Uh K, Lee K, Boulos P, Rowe C, Coviello C, Allen IC. Improved Therapeutic Delivery Targeting Clinically Relevant Orthotopic Human Pancreatic Tumors Engrafted in Immunocompromised Pigs Using Ultrasound-Induced Cavitation: A Pilot Study. Pharmaceutics 2023; 15:1585. [PMID: 37376034 PMCID: PMC10302458 DOI: 10.3390/pharmaceutics15061585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/03/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic tumors can be resistant to drug penetration due to high interstitial fluid pressure, dense stroma, and disarrayed vasculature. Ultrasound-induced cavitation is an emerging technology that may overcome many of these limitations. Low-intensity ultrasound, coupled with co-administered cavitation nuclei consisting of gas-stabilizing sub-micron scale SonoTran Particles, is effective at increasing therapeutic antibody delivery to xenograft flank tumors in mouse models. Here, we sought to evaluate the effectiveness of this approach in situ using a large animal model that mimics human pancreatic cancer patients. Immunocompromised pigs were surgically engrafted with human Panc-1 pancreatic ductal adenocarcinoma (PDAC) tumors in targeted regions of the pancreas. These tumors were found to recapitulate many features of human PDAC tumors. Animals were intravenously injected with the common cancer therapeutics Cetuximab, gemcitabine, and paclitaxel, followed by infusion with SonoTran Particles. Select tumors in each animal were targeted with focused ultrasound to induce cavitation. Cavitation increased the intra-tumor concentrations of Cetuximab, gemcitabine, and paclitaxel by 477%, 148%, and 193%, respectively, compared to tumors that were not targeted with ultrasound in the same animals. Together, these data show that ultrasound-mediated cavitation, when delivered in combination with gas-entrapping particles, improves therapeutic delivery in pancreatic tumors under clinically relevant conditions.
Collapse
Affiliation(s)
- Khan Mohammad Imran
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Benjamin Tintera
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Holly A. Morrison
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Juselyn D. Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Margaret A. Nagai-Singer
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Hannah Ivester
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
| | - McAlister Council-Troche
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Michael Edwards
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Christopher Byron
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Sherrie Clark-Deener
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| | - Kyungjun Uh
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
| | - Kiho Lee
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO 65211, USA
| | - Paul Boulos
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | - Cliff Rowe
- OxSonics Therapeutics, Oxford Science Park, Oxford OX4 4GA, UK
| | | | - Irving C. Allen
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA 24061, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
| |
Collapse
|
8
|
Ho YJ, Hsu HC, Wu BH, Lin YC, Liao LD, Yeh CK. Preventing ischemia-reperfusion injury by acousto-mechanical local oxygen delivery. J Control Release 2023; 356:481-492. [PMID: 36921723 DOI: 10.1016/j.jconrel.2023.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/28/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023]
Abstract
Ischemia-reperfusion (I/R) injury is a pathological process that causes vascular damage and dysfunction which increases recurrence and/or mortality in myocardial infarction, ischemic stroke, and organ transplantation. We hypothesized that ultrasound-stimulated oxygen-loaded microbubble (O2-MB) cavitation would enhance mechanical force on endothelium and simultaneously release oxygen locally at the targeted vessels. This cooperation between biomechanical and biochemical stimuli might modulate endothelial metabolism, providing a potential clinical approach to the prevention of I/R injury. Murine hindlimb and cardiac I/R models were used to demonstrate the feasibility of injury prevention by O2-MB cavitation. Increased mechanical force on endothelium induced eNOS-activated vasodilation and angiogenesis to prevent re-occlusion at the I/R vessels. Local oxygen therapy increased endothelial oxygenation that inhibited HIF-1α expression, increased ATP generation, and activated cyclin D1 for cell repair. Moreover, a decrease in interstitial H2O2 level reduced the expression of caspase3, NFκB, TNFα, and IL6, thus ameliorating inflammatory responses. O2-MB cavitation showed efficacy in maintaining cardiac function and preventing myocardial fibrosis after I/R. Finally, we present a potential pathway for the modulation of endothelial metabolism by O2-MB cavitation in relation to I/R injury, wound healing, and vascular bioeffects.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| | - Hui-Ching Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Bing-Huan Wu
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chun Lin
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
9
|
Li S, Zhang X. Three-dimensional acoustic radiation force of a eukaryotic cell arbitrarily positioned in a Gaussian beam. NANOTECHNOLOGY AND PRECISION ENGINEERING 2023. [DOI: 10.1063/10.0016831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Expressions are derived for calculating the three-dimensional acoustic radiation force (ARF) on a multilayer microsphere positioned arbitrarily in a Gaussian beam. A theoretical model of a three-layer microsphere with a cell membrane, cytoplasm, and nucleus is established to study how particle geometry and position affect the three-dimensional ARF, and its results agree well with finite-element numerical results. The microsphere can be moved relative to the beam axis by changing its structure and position in the beam, and the axial ARF increases with increasing outer-shell thickness and core size. This study offers a theoretical foundation for selecting suitable parameters for manipulating a three-layer microsphere in a Gaussian beam.
Collapse
Affiliation(s)
- Shuyuan Li
- Shaanxi Key Laboratory of Ultrasonics, School of Physics and Information Technology, Shaanxi Normal University, Xi’an 710119, China
| | - Xiaofeng Zhang
- Shaanxi Key Laboratory of Ultrasonics, School of Physics and Information Technology, Shaanxi Normal University, Xi’an 710119, China
| |
Collapse
|
10
|
Zhao X, Wright A, Goertz DE. An optical and acoustic investigation of microbubble cavitation in small channels under therapeutic ultrasound conditions. ULTRASONICS SONOCHEMISTRY 2023; 93:106291. [PMID: 36640460 PMCID: PMC9852793 DOI: 10.1016/j.ultsonch.2023.106291] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 06/04/2023]
Abstract
Therapeutic focused ultrasound in combination with encapsulated microbubbles is being widely investigated for its ability to elicit bioeffects in the microvasculature, such as transient permeabilization for drug delivery or at higher pressures to achieve 'antivascular' effects. While it is well established that the behaviors of microbubbles are altered when they are situated within sufficiently small vessels, there is a paucity of data examining how the bubble population dynamics and emissions change as a function of channel (vessel) diameter over a size range relevant to therapeutic ultrasound, particularly at pressures relevant to antivascular ultrasound. Here we use acoustic emissions detection and high-speed microscopy (10 kframes/s) to examine the behavior of a polydisperse clinically employed agent (Definity®) in wall-less channels as their diameters are scaled from 1200 to 15 µm. Pressures are varied from 0.1 to 3 MPa using either a 5 ms pulse or a sequence of 0.1 ms pulses spaced at 1 ms, both of which have been previously employed in an in vivo context. With increasing pressure, the 1200 µm channel - on the order of small arteries and veins - exhibited inertial cavitation, 1/2 subharmonics and 3/2 ultraharmonics, consistent with numerous previous reports. The 200 and 100 µm channels - in the size range of larger microvessels less affected by therapeutic focused ultrasound - exhibited a distinctly different behavior, having muted development of 1/2 subharmonics and 3/2 ultraharmonics and reduced persistence. These were associated with radiation forces displacing bubbles to the distal wall and inducing clusters that then rapidly dissipated along with emissions. As the diameter transitioned to 50 and then 15 µm - a size regime that is most relevant to therapeutic focused ultrasound - there was a higher threshold for the onset of inertial cavitation as well as subharmonics and ultraharmonics, which importantly had more complex orders that are not normally reported. Clusters also occurred in these channels (e.g. at 3 MPa, the mean lateral and axial sizes were 23 and 72 µm in the 15 µm channel; 50 and 90 µm in the 50 µm channel), however in this case they occupied the entire lumens and displaced the wall boundaries. Damage to the 15 µm channel was observed for both pulse types, but at a lower pressure for the long pulse. Experiments conducted with a 'nanobubble' (<0.45 µm) subpopulation of Definity followed broadly similar features to 'native' Definity, albeit at a higher pressure threshold for inertial cavitation. These results provide new insights into the behavior of microbubbles in small vessels at higher pressures and have implications for therapeutic focused ultrasound cavitation monitoring and control.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| | - Alex Wright
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada
| | - David E Goertz
- Department of Medical Biophysics, University of Toronto, M5G 1L7, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto M4N 3M5, Canada.
| |
Collapse
|
11
|
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
|
12
|
|
13
|
Li Y, Deng G, Hu X, Li C, Wang X, Zhu Q, Zheng K, Xiong W, Wu H. Recent advances in mesoporous silica nanoparticle-based targeted drug-delivery systems for cancer therapy. Nanomedicine (Lond) 2022; 17:1253-1279. [PMID: 36250937 DOI: 10.2217/nnm-2022-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Targeted drug-delivery systems are a growing research topic in tumor treatment. In recent years, mesoporous silica nanoparticles (MSNs) have been extensively studied and applied in noninvasive and biocompatible drug-delivery systems for tumor therapy due to their outstanding advantages, which include high surface area, large pore volume, tunable pore size, easy surface modification and stable framework. The advances in the application of MSNs for anticancer drug targeting are covered and highlighted in this review, and the challenges and prospects of MSN-based targeted drug-delivery systems are discussed. This review provides new insights for researchers interested in targeted drug-delivery systems against cancer.
Collapse
Affiliation(s)
- Ying Li
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Guoxing Deng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China.,School of Pharmacy, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xianlong Hu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Chenyang Li
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Xiaodong Wang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Qinchang Zhu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Wei Xiong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Haiqiang Wu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| |
Collapse
|
14
|
Breaking through the barrier: Modelling and exploiting the physical microenvironment to enhance drug transport and efficacy. Adv Drug Deliv Rev 2022; 184:114183. [PMID: 35278523 DOI: 10.1016/j.addr.2022.114183] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/03/2022] [Accepted: 03/06/2022] [Indexed: 02/08/2023]
Abstract
Pharmaceutical compounds are the main pillar in the treatment of various illnesses. To administer these drugs in the therapeutic setting, multiple routes of administration have been defined, including ingestion, inhalation, and injection. After administration, drugs need to find their way to the intended target for high effectiveness, and this penetration is greatly dependent on obstacles the drugs encounter along their path. Key hurdles include the physical barriers that are present within the body and knowledge of those is indispensable for progress in the development of drugs with increased therapeutic efficacy. In this review, we examine several important physical barriers, such as the blood-brain barrier, the gut-mucosal barrier, and the extracellular matrix barrier, and evaluate their influence on drug transport and efficacy. We explore various in vitro model systems that aid in understanding how parameters within the barrier model affect drug transfer and therapeutic effect. We conclude that physical barriers in the body restrict the quantity of drugs that can pass through, mainly as a consequence of the barrier architecture. In addition, the specific physical properties of the tissue can trigger intracellular changes, altering cell behavior in response to drugs. Though the barriers negatively influence drug distribution, physical stimulation of the surrounding environment may also be exploited as a mechanism to control drug release. This drug delivery approach is explored in this review as a potential alternative to the conventional ways of delivering therapeutics.
Collapse
|
15
|
Tang Y, Yu Z, Lu X, Fan Q, Huang W. Overcoming Vascular Barriers to Improve the Theranostic Outcomes of Nanomedicines. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103148. [PMID: 35246962 PMCID: PMC9069202 DOI: 10.1002/advs.202103148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/16/2022] [Indexed: 05/04/2023]
Abstract
Nanotheranostics aims to utilize nanomaterials to prevent, diagnose, and treat diseases to improve the quality of patients' lives. Blood vessels are responsible to deliver nutrients and oxygen to the whole body, eliminate waste, and provide access for patrolling immune cells for healthy tissues. Meanwhile, they can also nourish disease tissues, spread disease factors or cells into other healthy tissues, and deliver nanotheranostic agents to cover all the regions of a disease tissue. Thus, blood vessels are the first and the most important barrier for highly efficient nanotheranostics. Here, the structure and function of blood vessels are explored and how these characteristics affect nanotheranostics is discussed. Moreover, new mechanisms and related strategies about overcoming vascular obstacles for improved nanotheranostic outcomes are critically summarized, and their merits and demerits of each strategy are analyzed. Moreover, the present challenges to completely exhibit the potential of overcoming vascular barriers to improve the theranostic outcomes of nanomedicines in life science are also discussed. Finally, the future perspective is further discussed.
Collapse
Affiliation(s)
- Yufu Tang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
| | - Zhongzheng Yu
- School of Chemical and Biomedical EngineeringNanyang Technological UniversitySingapore637459Singapore
| | - Xiaomei Lu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
- Shaanxi Institute of Flexible Electronics (SIFE)Northwestern Polytechnical University (NPU)Xi'an710072China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM)Nanjing Tech University30 South Puzhu RoadNanjing211800P. R. China
- Key Laboratory for Organic Electronics and Information Displays and Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
- Shaanxi Institute of Flexible Electronics (SIFE)Northwestern Polytechnical University (NPU)Xi'an710072China
| |
Collapse
|
16
|
Overcoming Hypoxia-Induced Drug Resistance via Promotion of Drug Uptake and Reoxygenation by Acousto–Mechanical Oxygen Delivery. Pharmaceutics 2022; 14:pharmaceutics14050902. [PMID: 35631488 PMCID: PMC9144555 DOI: 10.3390/pharmaceutics14050902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
Hypoxia-induced drug resistance (HDR) is a critical issue in cancer therapy. The presence of hypoxic tumor cells impedes drug uptake and reduces the cytotoxicity of chemotherapeutic drugs, leading to HDR and increasing the probability of tumor recurrence and metastasis. Microbubbles, which are used as an ultrasound contrast agent and drug/gas carrier, can locally deliver drugs/gas and produce an acousto–mechanical effect to enhance cell permeability under ultrasound sonication. The present study applied oxygen-loaded microbubbles (OMBs) to evaluate the mechanisms of overcoming HDR via promotion of drug uptake and reoxygenation. A hypoxic mouse prostate tumor cell model was established by hypoxic incubation for 4 h. After OMB treatment, the permeability of HDR cells was enhanced by 23 ± 5% and doxorubicin uptake was increased by 11 ± 7%. The 61 ± 14% reoxygenation of HDR cells increased the cytotoxicity of doxorubicin from 18 ± 4% to 58 ± 6%. In combination treatment with OMB and doxorubicin, the relative contributions of uptake promotion and reoxygenation towards overcoming HDR were 11 ± 7% and 28 ± 10%, respectively. Our study demonstrated that reoxygenation of hypoxic conditions is a critical mechanism in the inhibition of HDR and enhancing the outcome of OMB treatment.
Collapse
|
17
|
Keller SB, Wang YN, Totten S, Yeung RS, Averkiou MA. Safety of Image-Guided Treatment of the Liver with Ultrasound and Microbubbles in an in Vivo Porcine Model. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3211-3220. [PMID: 34362584 DOI: 10.1016/j.ultrasmedbio.2021.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/15/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Ultrasound and microbubbles are useful for both diagnostic imaging and targeted drug delivery, making them ideal conduits for theranostic interventions. Recent reports have indicated the preclinical success of microbubble cavitation for enhancement of chemotherapy in abdominal tumors; however, there have been limited studies and variable efficacy in clinical implementation of this technique. This is likely because in contrast to the high pressures and long cycle lengths seen in successful preclinical work, current clinical implementation of microbubble cavitation for drug delivery generally involves low acoustic pressures and short cycle lengths to fit within clinical guidelines. To translate the preclinical parameter space to clinical adoption, a relevant safety study in a healthy large animal is required. Therefore, the purpose of this work was to evaluate the safety of ultrasound cavitation treatment (USCTx) in a healthy porcine model using a modified Philips EPIQ with S5-1 as the focused source. We performed USCTx on eight healthy pigs and monitored health over the course of 1 wk. We then performed an acute study of USCTx to evaluate immediate tissue damage. Contrast-enhanced ultrasound exams were performed before and after each treatment to investigate perfusion changes within the treated areas, and blood and urine were evaluated for liver damage biomarkers. We illustrate, through quantitative analysis of contrast-enhanced ultrasound data, blood and urine analyses and histology, that this technique and the parameter space considered are safe within the time frame evaluated. With its safety confirmed using a clinical-grade ultrasound scanner and contrast agent, USCTx could be easily translated into clinical trials for improvement of chemotherapy delivery. This represents the first safety study assessing the bio-effects of microbubble cavitation from relevant ultrasound parameters in a large animal model.
Collapse
Affiliation(s)
- Sara B Keller
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Yak-Nam Wang
- Applied Physics Laboratory, University of Washington, Seattle, Washington, USA
| | - Stephanie Totten
- Applied Physics Laboratory, University of Washington, Seattle, Washington, USA
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
18
|
Keller SB, Averkiou MA. The Role of Ultrasound in Modulating Interstitial Fluid Pressure in Solid Tumors for Improved Drug Delivery. Bioconjug Chem 2021; 33:1049-1056. [PMID: 34514776 DOI: 10.1021/acs.bioconjchem.1c00422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The unique microenvironment of solid tumors, including desmoplasia within the extracellular matrix, enhanced vascular permeability, and poor lymphatic drainage, leads to an elevated interstitial fluid pressure which is a major barrier to drug delivery. Reducing tumor interstitial fluid pressure is one proposed method of increasing drug delivery to the tumor. The goal of this topical review is to describe recent work using focused ultrasound with or without microbubbles to modulate tumor interstitial fluid pressure, through either thermal or mechanical effects on the extracellular matrix and the vasculature. Furthermore, we provide a review on techniques in which ultrasound imaging may be used to diagnose elevated interstitial fluid pressure within solid tumors. Ultrasound-based techniques show high promise in diagnosing and treating elevated interstitial pressure to enhance drug delivery.
Collapse
Affiliation(s)
- Sara B Keller
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Michalakis A Averkiou
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
19
|
Ho YJ, Huang CC, Fan CH, Liu HL, Yeh CK. Ultrasonic technologies in imaging and drug delivery. Cell Mol Life Sci 2021; 78:6119-6141. [PMID: 34297166 PMCID: PMC11072106 DOI: 10.1007/s00018-021-03904-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Ultrasonic technologies show great promise for diagnostic imaging and drug delivery in theranostic applications. The development of functional and molecular ultrasound imaging is based on the technical breakthrough of high frame-rate ultrasound. The evolution of shear wave elastography, high-frequency ultrasound imaging, ultrasound contrast imaging, and super-resolution blood flow imaging are described in this review. Recently, the therapeutic potential of the interaction of ultrasound with microbubble cavitation or droplet vaporization has become recognized. Microbubbles and phase-change droplets not only provide effective contrast media, but also show great therapeutic potential. Interaction with ultrasound induces unique and distinguishable biophysical features in microbubbles and droplets that promote drug loading and delivery. In particular, this approach demonstrates potential for central nervous system applications. Here, we systemically review the technological developments of theranostic ultrasound including novel ultrasound imaging techniques, the synergetic use of ultrasound with microbubbles and droplets, and microbubble/droplet drug-loading strategies for anticancer applications and disease modulation. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theranostic tool.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
20
|
Krafft MP, Riess JG. Therapeutic oxygen delivery by perfluorocarbon-based colloids. Adv Colloid Interface Sci 2021; 294:102407. [PMID: 34120037 DOI: 10.1016/j.cis.2021.102407] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
After the protocol-related indecisive clinical trial of Oxygent, a perfluorooctylbromide/phospholipid nanoemulsion, in cardiac surgery, that often unduly assigned the observed untoward effects to the product, the development of perfluorocarbon (PFC)-based O2 nanoemulsions ("blood substitutes") has come to a low. Yet, significant further demonstrations of PFC O2-delivery efficacy have continuously been reported, such as relief of hypoxia after myocardial infarction or stroke; protection of vital organs during surgery; potentiation of O2-dependent cancer therapies, including radio-, photodynamic-, chemo- and immunotherapies; regeneration of damaged nerve, bone or cartilage; preservation of organ grafts destined for transplantation; and control of gas supply in tissue engineering and biotechnological productions. PFC colloids capable of augmenting O2 delivery include primarily injectable PFC nanoemulsions, microbubbles and phase-shift nanoemulsions. Careful selection of PFC and other colloid components is critical. The basics of O2 delivery by PFC nanoemulsions will be briefly reminded. Improved knowledge of O2 delivery mechanisms has been acquired. Advanced, size-adjustable O2-delivering nanoemulsions have been designed that have extended room-temperature shelf-stability. Alternate O2 delivery options are being investigated that rely on injectable PFC-stabilized microbubbles or phase-shift PFC nanoemulsions. The latter combine prolonged circulation in the vasculature, capacity for penetrating tumor tissues, and acute responsiveness to ultrasound and other external stimuli. Progress in microbubble and phase-shift emulsion engineering, control of phase-shift activation (vaporization), understanding and control of bubble/ultrasound/tissue interactions is discussed. Control of the phase-shift event and of microbubble size require utmost attention. Further PFC-based colloidal systems, including polymeric micelles, PFC-loaded organic or inorganic nanoparticles and scaffolds, have been devised that also carry substantial amounts of O2. Local, on-demand O2 delivery can be triggered by external stimuli, including focused ultrasound irradiation or tumor microenvironment. PFC colloid functionalization and targeting can help adjust their properties for specific indications, augment their efficacy, improve safety profiles, and expand the range of their indications. Many new medical and biotechnological applications involving fluorinated colloids are being assessed, including in the clinic. Further uses of PFC-based colloidal nanotherapeutics will be briefly mentioned that concern contrast diagnostic imaging, including molecular imaging and immune cell tracking; controlled delivery of therapeutic energy, as for noninvasive surgical ablation and sonothrombolysis; and delivery of drugs and genes, including across the blood-brain barrier. Even when the fluorinated colloids investigated are designed for other purposes than O2 supply, they will inevitably also carry and deliver a certain amount of O2, and may thus be considered for O2 delivery or co-delivery applications. Conversely, O2-carrying PFC nanoemulsions possess by nature a unique aptitude for 19F MR imaging, and hence, cell tracking, while PFC-stabilized microbubbles are ideal resonators for ultrasound contrast imaging and can undergo precise manipulation and on-demand destruction by ultrasound waves, thereby opening multiple theranostic opportunities.
Collapse
Affiliation(s)
- Marie Pierre Krafft
- University of Strasbourg, Institut Charles Sadron (CNRS), 23 rue du Loess, 67034 Strasbourg, France.
| | - Jean G Riess
- Harangoutte Institute, 68160 Ste Croix-aux-Mines, France
| |
Collapse
|
21
|
Ho YJ, Chang HC, Lin CW, Fan CH, Lin YC, Wei KC, Yeh CK. Oscillatory behavior of microbubbles impacts efficacy of cellular drug delivery. J Control Release 2021; 333:316-327. [PMID: 33811982 DOI: 10.1016/j.jconrel.2021.03.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 01/16/2023]
Abstract
Drug-loaded microbubbles have been proven to be an effective strategy for non-invasive and local drug delivery when combined with ultrasound excitation for targeted drug release. Inertial cavitation is speculated to be a major mechanism for releasing drugs from drug-loaded microbubbles, but it results in lethal cellular pore damage that greatly limits its application. Thus, we investigated the cellular vesicle attachment and uptake to evaluate the efficiency of drug delivery by modulating the behaviors of targeted microbubble oscillation. The efficiency of vesicle attachment on the targeted cell membrane was 36.5 ± 15.9% and 3.8 ± 2.3% under stable and inertial cavitation, respectively. Further, stable cavitation enhanced cell permeability (26.8 ± 3.2%), maintained cell viability (90.8 ± 2.1%), and showed 7.9 ± 1.9-fold enhancement of in vivo vesicle release on tumor vessels. Therefore, our results reveal the ability to improve drug delivery via stable cavitation induced by targeted microbubbles. We propose that this strategy might be suitable for tissue repair or neuromodulation.
Collapse
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ho-Chun Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Wei Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan; Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan.
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, New Taipei City, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
22
|
Keller SB, Suo D, Wang YN, Kenerson H, Yeung RS, Averkiou MA. Image-Guided Treatment of Primary Liver Cancer in Mice Leads to Vascular Disruption and Increased Drug Penetration. Front Pharmacol 2020; 11:584344. [PMID: 33101038 PMCID: PMC7554611 DOI: 10.3389/fphar.2020.584344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022] Open
Abstract
Despite advances in interventional procedures and chemotherapeutic drug development, hepatocellular carcinoma (HCC) is still the fourth leading cause of cancer-related deaths worldwide with a <30% 5-year survival rate. This poor prognosis can be attributed to the fact that HCC most commonly occurs in patients with pre-existing liver conditions, rendering many treatment options too aggressive. Patient survival rates could be improved by a more targeted approach. Ultrasound-induced cavitation can provide a means for overcoming traditional barriers defining drug uptake. The goal of this work was to evaluate preclinical efficacy of image-guided, cavitation-enabled drug delivery with a clinical ultrasound scanner. To this end, ultrasound conditions (unique from those used in imaging) were designed and implemented on a Philips EPIQ and S5-1 phased array probe to produced focused ultrasound for cavitation treatment. Sonovue® microbubbles which are clinically approved as an ultrasound contrast agent were used for both imaging and cavitation treatment. A genetically engineered mouse model was bred and used as a physiologically relevant preclinical analog to human HCC. It was observed that image-guided and targeted microbubble cavitation resulted in selective disruption of the tumor blood flow and enhanced doxorubicin uptake and penetration. Histology results indicate that no gross morphological damage occurred as a result of this process. The combination of these effects may be exploited to treat HCC and other challenging malignancies and could be implemented with currently available ultrasound scanners and reagents.
Collapse
Affiliation(s)
- Sara B Keller
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Dingjie Suo
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Yak-Nam Wang
- Applied Physics Laboratory, University of Washington, Seattle, WA, United States
| | - Heidi Kenerson
- Department of Surgery, University of Washington, Seattle, WA, United States
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, WA, United States
| | | |
Collapse
|
23
|
Esmaeili J, Rezaei FS, Beram FM, Barati A. Integration of microbubbles with biomaterials in tissue engineering for pharmaceutical purposes. Heliyon 2020; 6:e04189. [PMID: 32577567 PMCID: PMC7303999 DOI: 10.1016/j.heliyon.2020.e04189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/24/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
Tissue engineering with the aid of biomaterials is a novel and promising knowledge aiming at improving human life expectancy. Besides, microbubbles are increasingly employed in biomedical applications due to their capability as a reservoir of therapeutic agents and oxygen molecules. In the present study, Microbubbles as the backbone of the research are produced as one of the potent devices in tissue engineering approaches, including drug delivery, wound healing, 3D printing, and scaffolding. It was shown that microbubbles are capable of promoting oxygen penetration and boosting the wound healing process by supplying adequate oxygen. Microbubbles also demonstrated their strength and potency in advancing drug delivery systems by reinforcing mass transfer phenomena. Furthermore, microbubbles developed the mechanical and biological characteristics of engineered scaffolds by manipulating the pores. Increasing cell survival, the biological activity of cells, angiogenesis, cell migration, and also nutrient diffusion into the inner layers of the scaffold were other achievements by microbubbles. In conclusion, the interest of biomedical communities in simultaneous usage of microbubbles and biomaterials under tissue engineering approaches experiences remarkable growth in Pharmaceutical studies.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
- Histogenotech Co., R&D Department, Tehran, Iran
| | - Farnoush Sadat Rezaei
- Department of Chemical Engineering, Faculty of Engineering, Amir Kabir University, Tehran, Iran
| | | | - Abolfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
| |
Collapse
|
24
|
Kooiman K, Roovers S, Langeveld SAG, Kleven RT, Dewitte H, O'Reilly MA, Escoffre JM, Bouakaz A, Verweij MD, Hynynen K, Lentacker I, Stride E, Holland CK. Ultrasound-Responsive Cavitation Nuclei for Therapy and Drug Delivery. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1296-1325. [PMID: 32165014 PMCID: PMC7189181 DOI: 10.1016/j.ultrasmedbio.2020.01.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 05/03/2023]
Abstract
Therapeutic ultrasound strategies that harness the mechanical activity of cavitation nuclei for beneficial tissue bio-effects are actively under development. The mechanical oscillations of circulating microbubbles, the most widely investigated cavitation nuclei, which may also encapsulate or shield a therapeutic agent in the bloodstream, trigger and promote localized uptake. Oscillating microbubbles can create stresses either on nearby tissue or in surrounding fluid to enhance drug penetration and efficacy in the brain, spinal cord, vasculature, immune system, biofilm or tumors. This review summarizes recent investigations that have elucidated interactions of ultrasound and cavitation nuclei with cells, the treatment of tumors, immunotherapy, the blood-brain and blood-spinal cord barriers, sonothrombolysis, cardiovascular drug delivery and sonobactericide. In particular, an overview of salient ultrasound features, drug delivery vehicles, therapeutic transport routes and pre-clinical and clinical studies is provided. Successful implementation of ultrasound and cavitation nuclei-mediated drug delivery has the potential to change the way drugs are administered systemically, resulting in more effective therapeutics and less-invasive treatments.
Collapse
Affiliation(s)
- Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Silke Roovers
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Simone A G Langeveld
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert T Kleven
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Medical School of the Vrije Universiteit Brussel, Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Martin D Verweij
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Christy K Holland
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA; Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
25
|
Ho YJ, Hsu HC, Kang ST, Fan CH, Chang CW, Yeh CK. Ultrasonic Transdermal Delivery System with Acid–Base Neutralization-Generated CO2 Microbubble Cavitation. ACS APPLIED BIO MATERIALS 2020; 3:1968-1975. [DOI: 10.1021/acsabm.9b01126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Yi-Ju Ho
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hui-Ching Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Shih-Tsung Kang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
- Trust Bio-sonics, Zhubei City 30261, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
26
|
Zhang L, Zhang S, Chen H, Liang Y, Zhao B, Luo W, Xiao Q, Li J, Zhu J, Peng C, Zhang Y, Hong Z, Wang Y, Li Y. An acoustic/thermo-responsive hybrid system for advanced doxorubicin delivery in tumor treatment. Biomater Sci 2020; 8:2202-2211. [PMID: 32100739 DOI: 10.1039/c9bm01794a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The efficiency of drug delivery and bioavailability to tumor cells are crucial for effective cancer chemotherapy. Herein, a doxorubicin (DOX) encapsulated lysolipid-based thermosensitive liposome decorated with cRGD peptide (RTSL) is conjugated on the surface of an IR780-loaded microbubble (IMB) to synthesize RTSL-IMBs. Sequentially taking advantage of acoustic-assisted early extravasation and thermo-triggered interstitium ultrafast drug release, RTSL-IMBs combine with ultrasound (US) and laser irradiation can advance drug delivery and bioavailability. In vitro experiments demonstrate that RTSL-IMBs associated with a two-step protocol (subsequently US irradiation for 1 min and laser irradiation for 5 min) can dramatically enhance the cellular uptake and bioavailability of DOX. In vivo fluorescence imaging studies reveal that the combination of RTSL-IMBs and US shows a 2.8-fold intratumoral drug accumulation increase at 0.5 h post-injection, while it will take 48 h to reach the same level of intratumoral drug accumulation for the RTSL-IMB group alone. Interestingly, the following localized application of a laser can further increase drug accumulation and slow tumor clearance. Histological analysis demonstrates that the combinational RTSL-IMBs, US and laser significantly improve the drug penetration distance and delivery efficiency in the tumor core. In this study, the acoustic/thermo-responsive hybrid system shows potential for advancing DOX chemotherapy in breast cancer cell MCF-7 xenograft nude mice.
Collapse
Affiliation(s)
- Li Zhang
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Superhydrophobic drug-loaded mesoporous silica nanoparticles capped with β-cyclodextrin for ultrasound image-guided combined antivascular and chemo-sonodynamic therapy. Biomaterials 2020; 232:119723. [DOI: 10.1016/j.biomaterials.2019.119723] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/15/2019] [Accepted: 12/21/2019] [Indexed: 01/13/2023]
|
28
|
Chen Y, Liang Y, Jiang P, Li F, Yu B, Yan F. Lipid/PLGA Hybrid Microbubbles as a Versatile Platform for Noninvasive Image-Guided Targeted Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2019; 11:41842-41852. [PMID: 31633326 DOI: 10.1021/acsami.9b10188] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Microbubbles (MBs) have recently emerged as promising theranostic carriers for ultrasound contrast imaging and drug delivery. However, conventional lipid-based MBs have a poor drug encapsulation efficiency, and polymer-based MBs show a weak capability in contrast imaging and ultrasound-triggered drug release. Here, we developed a novel type of multiporous lipid/PLGA hybrid MBs (lipid/PLGA MBs) that solved the dilemma of MBs as imaging agents and drug carriers. The lipid/PLGA MBs were designed through regulating the elasticity of the bubble shells using lipids to incorporate into the PLGA shells and ammonium bicarbonate as a gas-generating agent. The softened shells and the porous bubble structure make them be able to generate stronger harmonic signals and be more vulnerable to ultrasound irradiation, leading to their excellent performance in ultrasound contrast imaging and ultrasound-triggered MB destruction in vitro and in vivo. By using doxorubicin (Dox) as a model drug, the Dox-loaded lipid/PLGA MBs (Dox-lipid/PLGA MBs) were prepared and achieved a high drug encapsulation efficiency. The real-time tracking of drug delivery and on-command controlled drug release by ultrasound were successfully realized in the tumor-bearing mice. A significantly enhanced tumor growth inhibition effect could be observed when using Dox-lipid/PLGA MBs combined with ultrasound irradiation, compared with free Dox and Dox-lipid/PLGA MBs without ultrasound. Our study provides an innovative multifunctional platform of MBs for ultrasound contrast imaging and drug delivery applications.
Collapse
Affiliation(s)
- Yan Chen
- Ultrasound Medical Center , Zhujiang Hospital of Southern Medical University , Guangzhou 510282 , China
| | - Yangbiao Liang
- Ultrasound Medical Center , Zhujiang Hospital of Southern Medical University , Guangzhou 510282 , China
| | - Peng Jiang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences , Shenzhen 518055 , China
| | - Bo Yu
- Ultrasound Medical Center , Zhujiang Hospital of Southern Medical University , Guangzhou 510282 , China
| | - Fei Yan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology , Chinese Academy of Sciences , Shenzhen 518055 , China
| |
Collapse
|
29
|
Ho YJ, Chu SW, Liao EC, Fan CH, Chan HL, Wei KC, Yeh CK. Normalization of Tumor Vasculature by Oxygen Microbubbles with Ultrasound. Am J Cancer Res 2019; 9:7370-7383. [PMID: 31695774 PMCID: PMC6831304 DOI: 10.7150/thno.37750] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/30/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor microenvironment influences the efficacy of anti-cancer therapies. The dysfunctional tumor vasculature limits the efficiency of oxygenation and drug delivery to reduce treatment outcome. A concept of tumor vascular normalization (VN), which inhibits angiogenesis to improve vessel maturity, blood perfusion, and oxygenation, has been demonstrated under the anti-angiogenic therapy. The efficiency of drug delivery and penetration is increased by enhancing perfusion and reducing interstitial fluid pressure during the time window of VN. However, anti-angiogenic agents only induce transient VN and then prune vessels to aggravate tumor hypoxia. To repair tumor vessels without altering vessel density, we proposed to induce tumor VN by local oxygen release via oxygen microbubbles with ultrasound. With tumor perfusion enhancement under ultrasound contrast imaging tracing, the time window of VN was defined as 2-8 days after a single oxygen microbubble treatment. The enhanced tumor oxygenation after oxygen microbubble treatment inhibited hypoxia inducible factor-1 alpha (HIF-1α)/vascular endothelial growth factor (VEGF) pathway to improve the morphology and function of tumor vasculature. The pericyte coverage and Hoechst penetration of tumor vessels increased without any changes to the vessel density. Finally, the intratumoral accumulation of anti-cancer drug doxorubicin could be increased 3-4 folds during tumor VN. These findings demonstrate that regulating tumor oxygenation by oxygen microbubbles could normalize dysfunctional vessels to enhance vascular maturity, blood perfusion, and drug penetration. Furthermore, ultrasound perfusion imaging provides a simple and non-invasive way to detect the VN time window, which increases the feasibility of VN in clinical cancer applications.
Collapse
|
30
|
Simpson JD, Smith SA, Thurecht KJ, Such G. Engineered Polymeric Materials for Biological Applications: Overcoming Challenges of the Bio-Nano Interface. Polymers (Basel) 2019; 11:E1441. [PMID: 31480780 PMCID: PMC6780590 DOI: 10.3390/polym11091441] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/11/2022] Open
Abstract
Nanomedicine has generated significant interest as an alternative to conventional cancertherapy due to the ability for nanoparticles to tune cargo release. However, while nanoparticletechnology has promised significant benefit, there are still limited examples of nanoparticles inclinical practice. The low translational success of nanoparticle research is due to the series ofbiological roadblocks that nanoparticles must migrate to be effective, including blood and plasmainteractions, clearance, extravasation, and tumor penetration, through to cellular targeting,internalization, and endosomal escape. It is important to consider these roadblocks holistically inorder to design more effective delivery systems. This perspective will discuss how nanoparticlescan be designed to migrate each of these biological challenges and thus improve nanoparticledelivery systems in the future. In this review, we have limited the literature discussed to studiesinvestigating the impact of polymer nanoparticle structure or composition on therapeutic deliveryand associated advancements. The focus of this review is to highlight the impact of nanoparticlecharacteristics on the interaction with different biological barriers. More specific studies/reviewshave been referenced where possible.
Collapse
Affiliation(s)
- Joshua D Simpson
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, the University of Queensland, St Lucia QLD 4072, Australia;
| | - Samuel A Smith
- School of Chemistry, University of Melbourne, Parkville VIC 3010, Australia;
| | - Kristofer J. Thurecht
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, the University of Queensland, St Lucia QLD 4072, Australia;
| | - Georgina Such
- School of Chemistry, University of Melbourne, Parkville VIC 3010, Australia;
| |
Collapse
|
31
|
Yildirim A, Blum NT, Goodwin AP. Colloids, nanoparticles, and materials for imaging, delivery, ablation, and theranostics by focused ultrasound (FUS). Theranostics 2019; 9:2572-2594. [PMID: 31131054 PMCID: PMC6525987 DOI: 10.7150/thno.32424] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 12/15/2022] Open
Abstract
This review focuses on different materials and contrast agents that sensitize imaging and therapy with Focused Ultrasound (FUS). At high intensities, FUS is capable of selectively ablating tissue with focus on the millimeter scale, presenting an alternative to surgical intervention or management of malignant growth. At low intensities, FUS can be also used for other medical applications such as local delivery of drugs and blood brain barrier opening (BBBO). Contrast agents offer an opportunity to increase selective acoustic absorption or facilitate destructive cavitation processes by converting incident acoustic energy into thermal and mechanical energy. First, we review the history of FUS and its effects on living tissue. Next, we present different colloidal or nanoparticulate approaches to sensitizing FUS, for example using microbubbles, phase-shift emulsions, hollow-shelled nanoparticles, or hydrophobic silica surfaces. Exploring the science behind these interactions, we also discuss ways to make stimulus-responsive, or "turn-on" contrast agents for improved selectivity. Finally, we discuss acoustically-active hydrogels and membranes. This review will be of interest to those working in materials who wish to explore new applications in acoustics and those in acoustics who are seeking new agents to improve the efficacy of their approaches.
Collapse
Affiliation(s)
- Adem Yildirim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
- Present address: CEDAR, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97239 USA
| | - Nicholas T. Blum
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303 USA
| |
Collapse
|
32
|
Feng Y, He Z, Mao C, Shui X, Cai L. Therapeutic Effects of Resveratrol Liposome on Muscle Injury in Rats. Med Sci Monit 2019; 25:2377-2385. [PMID: 30936416 PMCID: PMC6457134 DOI: 10.12659/msm.913409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In this study we prepared liposome microbubbles loading resveratrol (LMLR) and evaluated its therapeutic effect on injury of gastrocnemius muscle in rats. MATERIAL AND METHODS LMLR was prepared and characterized by particle size, potential, and microscopy, and a rat model of acute blunt injury of gastrocnemius muscle was established. After treatments with resveratrol or LMLR, the therapeutic effects were evaluated by hematoxylin-eosin (HE) staining. The expression of MHCIIB and vimentin in mRNA level was measured by real-time PCR. The expression of desmin and collagen I protein was assessed by immunohistochemistry. RESULTS LMLR showed regular cycle shape in a size of ~1000 nm. LMLR was negatively charged (-30 mV). The in vitro release of LMLR was close to 80% at 10 h and 90% at 48 h. Acute gastrocnemius muscle injury was established in rats and tissue recovery was observed after LMLR treatment as evidenced by HE staining, decreased expression of MHCIIB, and increased expression of vimentin. Moreover, LMLR treatment obviously facilitated desmin expression and reduced collagen I expression. CONCLUSIONS LMLR is effective in treating acute blunt injury of gastrocnemius muscle in rats.
Collapse
Affiliation(s)
- Yongzeng Feng
- Department of Orthopedic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang, P.R. China
| | - Zili He
- Department of Orthopedic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang, P.R. China
| | - Cong Mao
- Department of Orthopedic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang, P.R. China
| | - Xiaolong Shui
- Department of Orthopedic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang, P.R. China
| | - Leyi Cai
- Department of Orthopedic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou, Zhejiang, P.R. China
| |
Collapse
|
33
|
Jiang ZQ, Wang YY, Yao J, Wu DJ, Liu XJ. Acoustic radiation forces on three-layered drug particles in focused Gaussian beams. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 145:1331. [PMID: 31067931 DOI: 10.1121/1.5093544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/19/2019] [Indexed: 05/19/2023]
Abstract
Drug delivery by acoustic waves is a crucial technology for targeted therapy. Recently, a three-layered drug micro-particle was proposed and fabricated, the second shell of which greatly improves both the encapsulation of the drug and the flexibility in its release rate. In this work, the acoustic radiation force (ARF) of an acoustic focused Gaussian beam on a three-layered particle comprising an inner drug core (D), a middle layer of poly(lactide-co-glycolide) (PLGA), and an outer chitosan shell (CS) is investigated. A three-layered elastic shell (TES) mimics the D-PLGA-CS structure, and the acoustic scattering from and ARF of the D-PLGA-CS are studied using Mie theory. This paper focuses on how the geometry and acoustic parameters of the outer shell influence the ARF, finding that the Poisson's ratio of the outer shell affects the ARF more than does the density or Young's modulus. In addition, this paper finds that the choice of the inner drug has little effect on the ARF acting on the D-PLGA-CS particle. The present work may benefit the acoustic manipulation of both TESs and three-layered drugs.
Collapse
Affiliation(s)
- Zhong-Qiu Jiang
- Jiangsu Key Laboratory on Opto-Electronic Technology, School of Physics and Technology, Nanjing Normal University, Nanjing 210023, China
| | - Yuan-Yuan Wang
- Jiangsu Key Laboratory on Opto-Electronic Technology, School of Physics and Technology, Nanjing Normal University, Nanjing 210023, China
| | - Jie Yao
- Jiangsu Key Laboratory on Opto-Electronic Technology, School of Physics and Technology, Nanjing Normal University, Nanjing 210023, China
| | - Da-Jian Wu
- Jiangsu Key Laboratory on Opto-Electronic Technology, School of Physics and Technology, Nanjing Normal University, Nanjing 210023, China
| | - Xiao-Jun Liu
- Key Laboratory of Modern Acoustics, Department of Physics and Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| |
Collapse
|
34
|
Najafi M, Goradel NH, Farhood B, Salehi E, Solhjoo S, Toolee H, Kharazinejad E, Mortezaee K. Tumor microenvironment: Interactions and therapy. J Cell Physiol 2018; 234:5700-5721. [PMID: 30378106 DOI: 10.1002/jcp.27425] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
Tumor microenvironment (TME) is a host for a complex network of heterogeneous stromal cells with overlapping or opposing functions depending on the dominant signals within this milieu. Reciprocal paracrine interactions between cancer cells with cells within the tumor stroma often reshape the TME in favor of the promotion of tumor. These complex interactions require more sophisticated approaches for cancer therapy, and, therefore, advancing knowledge about dominant drivers of cancer within the TME is critical for designing therapeutic schemes. This review will provide knowledge about TME architecture, multiple signaling, and cross communications between cells within this milieu, and its targeting for immunotherapy of cancer.
Collapse
Affiliation(s)
- Masoud Najafi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Department of Radiology and Medical Physics, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Eniseh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somaye Solhjoo
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Heidar Toolee
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|