1
|
Mohammadi R, Ghani S, Arezumand R, Farhadi S, Khazaee-Poul Y, Kazemi B, Yarian F, Noruzi S, Alibakhshi A, Jalili M, Aghamiri S. Physicochemical Stimulus-Responsive Systems Targeted with Antibody Derivatives. Curr Mol Med 2024; 24:1250-1268. [PMID: 37594115 DOI: 10.2174/1566524023666230818093016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 08/19/2023]
Abstract
The application of monoclonal antibodies and antibody fragments with the advent of recombinant antibody technology has made notable progress in clinical trials to provide a regulated drug release and extra targeting to the special conditions in the function site. Modification of antibodies has facilitated using mAbs and antibody fragments in numerous models of therapeutic and detection utilizations, such as stimuliresponsive systems. Antibodies and antibody derivatives conjugated with diverse stimuliresponsive materials have been constructed for drug delivery in response to a wide range of endogenous (electric, magnetic, light, radiation, ultrasound) and exogenous (temperature, pH, redox potential, enzymes) stimuli. In this report, we highlighted the recent progress on antibody-conjugated stimuli-responsive and dual/multi-responsive systems that affect modern medicine by improving a multitude of diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Rezvan Mohammadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Ghani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghaye Arezumand
- Department of Advanced Technology, School of Medicine, North Khorasan University of Medical Sciences, North Khorasan, Iran
| | - Shohreh Farhadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yalda Khazaee-Poul
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Yarian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Somaye Noruzi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Jalili
- Preventive and Clinical Nutrition Group, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Shahin Aghamiri
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Liu H, Zhang J, Luo J, Wen D. Impact of nanodroplets on cone-textured surfaces. Phys Rev E 2023; 107:065101. [PMID: 37464703 DOI: 10.1103/physreve.107.065101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/10/2023] [Indexed: 07/20/2023]
Abstract
Molecular dynamics simulations have been performed to study the dynamics of nanodroplets impacting on a flat superhydrophobic surface and surfaces covered with nanocone structures. We present a panorama of nanodroplet behaviors for a wide range of impact velocities and different cone geometrics, and develop a model to predict whether a nanodroplet impacting onto cone-textured surfaces will touch the underlying substrate during impact. The advantages and disadvantages of applying nanocone structures to the solid surface are revealed by the investigations into restitution coefficient and contact time. The effects of nanocone structures on droplet bouncing dynamics are probed using momentum analysis rather than conventional energy analysis. We further demonstrate that a single Weber number is inadequate for unifying the dynamics of macroscale and nanoscale droplets on cone-textured surfaces, and propose a combined dimensionless number to address it. The extensive findings of this study carry noteworthy implications for engineering applications, such as nanoprinting and nanomedicine on functional patterned surfaces, providing fundamental support for these technologies.
Collapse
Affiliation(s)
- Hanyi Liu
- School of Aeronautic Science and Engineering, Beihang University, Beijing 100191, China
| | - Jun Zhang
- School of Aeronautic Science and Engineering, Beihang University, Beijing 100191, China
| | - Jia Luo
- School of Aeronautic Science and Engineering, Beihang University, Beijing 100191, China
| | - Dongsheng Wen
- School of Aeronautic Science and Engineering, Beihang University, Beijing 100191, China
- TUM School of Engineering and Design, Technical University of Munich, 80333 Munich, Germany
| |
Collapse
|
3
|
Kaykanat SI, Uguz AK. The role of acoustofluidics and microbubble dynamics for therapeutic applications and drug delivery. BIOMICROFLUIDICS 2023; 17:021502. [PMID: 37153864 PMCID: PMC10162024 DOI: 10.1063/5.0130769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/18/2023] [Indexed: 05/10/2023]
Abstract
Targeted drug delivery is proposed to reduce the toxic effects of conventional therapeutic methods. For that purpose, nanoparticles are loaded with drugs called nanocarriers and directed toward a specific site. However, biological barriers challenge the nanocarriers to convey the drug to the target site effectively. Different targeting strategies and nanoparticle designs are used to overcome these barriers. Ultrasound is a new, safe, and non-invasive drug targeting method, especially when combined with microbubbles. Microbubbles oscillate under the effect of the ultrasound, which increases the permeability of endothelium, hence, the drug uptake to the target site. Consequently, this new technique reduces the dose of the drug and avoids its side effects. This review aims to describe the biological barriers and the targeting types with the critical features of acoustically driven microbubbles focusing on biomedical applications. The theoretical part covers the historical developments in microbubble models for different conditions: microbubbles in an incompressible and compressible medium and bubbles encapsulated by a shell. The current state and the possible future directions are discussed.
Collapse
Affiliation(s)
- S. I. Kaykanat
- Department of Chemical Engineering, Boğaziçi University, 34342 Bebek, Istanbul, Türkiye
| | | |
Collapse
|
4
|
Liu Y, Yang S, Zhou Q, Zhou J, Li J, Ma Y, Hu B, Liu C, Zhao Y. Nanobubble-based anti-hepatocellular carcinoma therapy combining immune check inhibitors and sonodynamic therapy. NANOSCALE ADVANCES 2022; 4:4847-4862. [PMID: 36545394 PMCID: PMC9642800 DOI: 10.1039/d2na00322h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/27/2022] [Indexed: 05/16/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide, posing a global threat to human healthcare, and current approved treatment strategies do not produce satisfactory outcomes. Here, nanobubbles (NBs) were prepared that carried Immune Check Inhibitors (ICIs), PD-L1 antibody (PD-L1 Ab) and sonodynamic agent CHLORIN E6 (Ce6); the anti-cancer properties of these NBs were analyzed from the point of view of immune and sonodynamic therapies. The PD-L1 Ab/Ce6-NBs could inhibit tumor growth through regulating reactive oxygen species (ROS) production, apoptosis, and most importantly, the function of associated immunocytes, including natural killer cells and lymphocytes. The tumor tissues highly expressed markers of immunogenic tumor cell death (ICD) in which the expression of calreticulin (CRT) and ICD-related immune cytokines (CD80, CD86, INF-γ, and IL-2) were increased in PD-L1 Ab/Ce6-NB treated mice. PD-L1 Ab/Ce6-NBs also promoted murine spleen lymphocyte proliferation and cytotoxic activity, as well as CD8+ T cell infiltration in the tumor tissues, and downregulation of the PD-L1 protein and mRNA expression. Furthermore, Bax expression was increased and Bcl-2 was inhibited at the mRNA and protein levels in a murine subcutaneous transplanted tumor model. These results indicate that PD-L1 Ab/Ce6-NBs can induce ROS-dependent ICD to further boost anti-cancer immune responses under the action of targeting the PD-1/PD-L1 immune check point in the tumor microenvironment as a promising therapeutic agent for HCC.
Collapse
Affiliation(s)
- Yun Liu
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University Yichang China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University Yichang China
- Echo Laboratory, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University Wuhan China
| | - ShiQi Yang
- Medical College, China Three Gorges University No.8 Daxue Road, Xiling District Yichang China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University Yichang China
| | - Qing Zhou
- Echo Laboratory, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University Wuhan China
| | - Jun Zhou
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University Yichang China
| | - Jinlin Li
- Department of Ultrasound Imaging, Affiliated Renhe Hospital of China Three Gorges University Yichang China
| | - Yao Ma
- Department of Ultrasound Imaging, The First College of Clinical Medical Science, China Three Gorges University Yichang China
| | - Bing Hu
- Department of Ultrasound Imaging, Affiliated Renhe Hospital of China Three Gorges University Yichang China
| | - Chaoqi Liu
- Medical College, China Three Gorges University No.8 Daxue Road, Xiling District Yichang China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University Yichang China
| | - Yun Zhao
- Medical College, China Three Gorges University No.8 Daxue Road, Xiling District Yichang China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University Yichang China
| |
Collapse
|
5
|
Vidallon MLP, Teo BM, Bishop AI, Tabor RF. Next-Generation Colloidal Materials for Ultrasound Imaging Applications. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1373-1396. [PMID: 35641393 DOI: 10.1016/j.ultrasmedbio.2022.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 06/15/2023]
Abstract
Ultrasound has important applications, predominantly in the field of diagnostic imaging. Presently, colloidal systems such as microbubbles, phase-change emulsion droplets and particle systems with acoustic properties and multiresponsiveness are being developed to address typical issues faced when using commercial ultrasound contrast agents, and to extend the utility of such systems to targeted drug delivery and multimodal imaging. Current technologies and increasing research data on the chemistry, physics and materials science of new colloidal systems are also leading to the development of more complex, novel and application-specific colloidal assemblies with ultrasound contrast enhancement and other properties, which could be beneficial for multiple biomedical applications, especially imaging-guided treatments. In this article, we review recent developments in new colloids with applications that use ultrasound contrast enhancement. This work also highlights the emergence of colloidal materials fabricated from or modified with biologically derived and bio-inspired materials, particularly in the form of biopolymers and biomembranes. Challenges, limitations, potential developments and future directions of these next-generation colloidal systems are also presented and discussed.
Collapse
Affiliation(s)
| | - Boon Mian Teo
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| | - Alexis I Bishop
- School of Physics and Astronomy, Monash University, Clayton, Victoria, Australia
| | - Rico F Tabor
- School of Chemistry, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
6
|
Alphandéry E. Ultrasound and nanomaterial: an efficient pair to fight cancer. J Nanobiotechnology 2022; 20:139. [PMID: 35300712 PMCID: PMC8930287 DOI: 10.1186/s12951-022-01243-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/02/2022] [Indexed: 01/12/2023] Open
Abstract
Ultrasounds are often used in cancer treatment protocols, e.g. to collect tumor tissues in the right location using ultrasound-guided biopsy, to image the region of the tumor using more affordable and easier to use apparatus than MRI and CT, or to ablate tumor tissues using HIFU. The efficacy of these methods can be further improved by combining them with various nano-systems, thus enabling: (i) a better resolution of ultrasound imaging, allowing for example the visualization of angiogenic blood vessels, (ii) the specific tumor targeting of anti-tumor chemotherapeutic drugs or gases attached to or encapsulated in nano-systems and released in a controlled manner in the tumor under ultrasound application, (iii) tumor treatment at tumor site using more moderate heating temperatures than with HIFU. Furthermore, some nano-systems display adjustable sizes, i.e. nanobubbles can grow into micro-bubbles. Such dual size is advantageous since it enables gathering within the same unit the targeting properties of nano bubbles via EPR effect and the enhanced ultrasound contrasting properties of micro bubbles. Interestingly, the way in which nano-systems act against a tumor could in principle also be adjusted by accurately selecting the nano-system among a large choice and by tuning the values of the ultrasound parameters, which can lead, due to their mechanical nature, to specific effects such as cavitation that are usually not observed with purely electromagnetic waves and can potentially help destroying the tumor. This review highlights the clinical potential of these combined treatments that can improve the benefit/risk ratio of current cancer treatments.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS, 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de. Cosmochimie, IMPMC, 75005, Paris, France. .,Nanobacterie SARL, 36 boulevard Flandrin, 75116, Paris, France. .,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
7
|
Epigenetic Priming with Decitabine Augments the Therapeutic Effect of Cisplatin on Triple-Negative Breast Cancer Cells through Induction of Proapoptotic Factor NOXA. Cancers (Basel) 2022; 14:cancers14010248. [PMID: 35008411 PMCID: PMC8749981 DOI: 10.3390/cancers14010248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 12/10/2022] Open
Abstract
Epigenetic alterations caused by aberrant DNA methylation have a crucial role in cancer development, and the DNA-demethylating agent decitabine, is used to treat hematopoietic malignancy. Triple-negative breast cancers (TNBCs) have shown sensitivity to decitabine; however, the underlying mechanism of its anticancer effect and its effectiveness in treating TNBCs are not fully understood. We analyzed the effects of decitabine on nine TNBC cell lines and examined genes associated with its cytotoxic effects. According to the effect of decitabine, we classified the cell lines into cell death (D)-type, growth inhibition (G)-type, and resistant (R)-type. In D-type cells, decitabine induced the expression of apoptotic regulators and, among them, NOXA was functionally involved in decitabine-induced apoptosis. In G-type cells, induction of the cyclin-dependent kinase inhibitor, p21, and cell cycle arrest were observed. Furthermore, decitabine enhanced the cytotoxic effect of cisplatin mediated by NOXA in D-type and G-type cells. In contrast, the sensitivity to cisplatin was high in R-type cells, and no enhancing effect by decitabine was observed. These results indicate that decitabine enhances the proapoptotic effect of cisplatin on TNBC cell lines that are less sensitive to cisplatin, indicating the potential for combination therapy in TNBC.
Collapse
|
8
|
Song HW, Lee HS, Kim SJ, Kim HY, Choi YH, Kang B, Kim CS, Park JO, Choi E. Sonazoid-Conjugated Natural Killer Cells for Tumor Therapy and Real-Time Visualization by Ultrasound Imaging. Pharmaceutics 2021; 13:pharmaceutics13101689. [PMID: 34683982 PMCID: PMC8537855 DOI: 10.3390/pharmaceutics13101689] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 01/08/2023] Open
Abstract
Various cell therapy strategies, including chimeric antigen receptor-expressing T or natural killer (NK) cells and cell-mediated drug delivery, have been developed for tumor eradication. However, the efficiency of these strategies against solid tumors remains unclear. We hypothesized that real-time control and visualization of therapeutic cells, such as NK cells, would improve their therapeutic efficacy against solid tumors. In this study, we engineered Sonazoid microbubble-conjugated NK (NK_Sona) cells and demonstrated that they were detectable by ultrasound imaging in real-time and maintained their functions. The Sonazoid microbubbles on the cell membrane did not affect the cytotoxicity and viability of the NK cells in vitro. Additionally, the NK_Sona cells could be visualized by ultrasound imaging and inhibited tumor growth in vivo. Taken together, our findings demonstrate the feasibility of this new approach in the use of therapeutic cells, such as NK cells, against solid tumors.
Collapse
Affiliation(s)
- Hyeong-Woo Song
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
| | - Han-Sol Lee
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Korea
| | - Seok-Jae Kim
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Korea
| | - Ho Yong Kim
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
| | - You Hee Choi
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
| | - Byungjeon Kang
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
- College of AI Convergence, Chonnam National University, Gwangju 61186, Korea
| | - Chang-Sei Kim
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Korea
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Korea
- Correspondence: (J.-O.P.); (E.C.)
| | - Eunpyo Choi
- Korea Institute of Medical Microrobotics, Gwangju 61011, Korea; (H.-W.S.); (H.-S.L.); (S.-J.K.); (H.Y.K.); (Y.H.C.); (B.K.); (C.-S.K.)
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Korea
- Correspondence: (J.-O.P.); (E.C.)
| |
Collapse
|
9
|
Ultrasound and Photoacoustic Imaging of Laser-Activated Phase-Change Perfluorocarbon Nanodroplets. PHOTONICS 2021. [DOI: 10.3390/photonics8100405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Laser-activated perfluorocarbon nanodroplets (PFCnDs) are emerging phase-change contrast agents that showed promising potential in ultrasound and photoacoustic (US/PA) imaging. Unlike monophase gaseous microbubbles, PFCnDs shift their state from liquid to gas via optical activation and can provide high US/PA contrast on demand. Depending on the choice of perfluorocarbon core, the vaporization and condensation dynamics of the PFCnDs are controllable. Therefore, these configurable properties of activation and deactivation of PFCnDs are employed to enable various imaging approaches, including contrast-enhanced imaging and super-resolution imaging. In addition, synchronous application of both acoustic and optical pulses showed a promising outcome vaporizing PFCnDs with lower activation thresholds. Furthermore, due to their sub-micrometer size, PFCnDs can be used for molecular imaging of extravascular tissue. PFCnDs can also be an effective therapeutic tool. As PFCnDs can carry therapeutic drugs or other particles, they can be used for drug delivery, as well as photothermal and photodynamic therapies. Blood barrier opening for neurological applications was recently demonstrated with optically-triggered PFCnDs. This paper specifically focuses on the activation and deactivation properties of laser-activated PFCnDs and associated US/PA imaging approaches, and briefly discusses their theranostic potential and future directions.
Collapse
|
10
|
Ohta S, Kikuchi E, Ishijima A, Azuma T, Sakuma I, Ito T. Investigating the optimum size of nanoparticles for their delivery into the brain assisted by focused ultrasound-induced blood-brain barrier opening. Sci Rep 2020; 10:18220. [PMID: 33106562 PMCID: PMC7588485 DOI: 10.1038/s41598-020-75253-9] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
The blood–brain barrier (BBB) has hampered the efficiency of nanoparticle delivery into the brain via conventional strategies. The widening of BBB tight junctions via focused ultrasound (FUS) offers a promising approach for enhancing the delivery of nanoparticles into the brain. However, there is currently an insufficient understanding of how nanoparticles pass through the opened BBB gaps. Here we investigated the size-dependence of nanoparticle delivery into the brain assisted by FUS-induced BBB opening, using gold nanoparticles (AuNPs) of 3, 15, and 120 nm diameter. For 3- and 15-nm AuNPs, FUS exposure significantly increased permeation across an in vitro BBB model by up to 9.5 times, and the permeability was higher with smaller diameter. However, in vivo transcranial FUS exposure in mice demonstrated that smaller particles were not necessarily better for delivery into the brain. Medium-sized (15 nm) AuNPs showed the highest delivery efficiency (0.22% ID), compared with 3- and 120-nm particles. A computational model suggested that this optimum size was determined by the competition between their permeation through opened BBB gaps and their excretion from blood. Our results would greatly contribute to designing nanoparticles for their delivery into the brain for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Seiichi Ohta
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan. .,Institute of Engineering Innovation, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| | - Emi Kikuchi
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ayumu Ishijima
- Department of Precision Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takashi Azuma
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ichiro Sakuma
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.,Department of Precision Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Taichi Ito
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
11
|
Ma Y, Han J, Jiang J, Zheng Z, Tan Y, Liu C, Zhao Y. Ultrasound targeting of microbubble-bound anti PD-L1 mAb to enhance anti-tumor effect of cisplatin in cervical cancer xenografts treatment. Life Sci 2020; 262:118565. [PMID: 33038371 DOI: 10.1016/j.lfs.2020.118565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
AIMS Anti-PD-L1 monoclonal antibody (mAb)-conjugated ultrasound (US) lipid-shelled microbubbles (PD-L1-MBs) were successfully synthesized to investigate whether that PD-L1-MBs could enhance anti-tumor effect in combination therapy with cisplatin (CDDP) under ultrasound mediation. MAIN METHODS Based on affinity between biotin and streptavidin, we prepared microbubbles conjugated with anti-PD-L1 mAb by membrane hydration and mechanical oscillation. A subcutaneous tumor model was established to test the anti-tumor effect and immunological activity of this combination therapy. Bax and Bcl-2 expression were detected by RT-qPCR and Immunohistochemistry. Cells undergoing apoptosis in tissue section were determined by TUNEL. Proliferation of splenocytes was analyzed by Flow cytometry. A cytotoxic T lymphocyte assay was performed by CTL. Expression of PD-L1 and CD8 in tissue section was examined by immunologfluorescence. Expression of IFN-γ, TNF-α, CD86 and CD80 was also detected by RT-qPCR. KEY FINDINGS We observed that the growth of the subcutaneous tumor was significantly slower in combined group than that in the group treated with either drug or microbubbles. Moreover, higher antitumor activity was observed in the combined group than that in cisplatin alone, which could be reflected by the number of apoptotic cells in tumor tissues and over expression of bax in the combined group. This combination treatment also exhibited a better immunological activity, increasing the infiltration of CD8+ T cells and the expression of several revelant cytokines. SIGNIFICANCE The ultrasound lipid-shelled PD-L1-MBs may enhance anti-tumor effects of cisplatin by blocking the PD-L1 site and improving immune function.
Collapse
Affiliation(s)
- Yao Ma
- Medical College of China Three Gorges University, Yichang, China; Department of Ultrasonography, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
| | - Jiaxuan Han
- Medical College of China Three Gorges University, Yichang, China
| | - Jinjun Jiang
- Medical College of China Three Gorges University, Yichang, China
| | - Zhiwei Zheng
- Medical College of China Three Gorges University, Yichang, China
| | - Yandi Tan
- Medical College of China Three Gorges University, Yichang, China
| | - Chaoqi Liu
- Medical College of China Three Gorges University, Yichang, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Yichang, China.
| | - Yun Zhao
- Medical College of China Three Gorges University, Yichang, China.
| |
Collapse
|
12
|
A review of ultrasound-mediated microbubbles technology for cancer therapy: a vehicle for chemotherapeutic drug delivery. JOURNAL OF RADIOTHERAPY IN PRACTICE 2020. [DOI: 10.1017/s1460396919000633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
AbstractBackground:The unique behaviour of microbubbles under ultrasound acoustic pressure makes them useful agents for drug and gene delivery. Several studies have demonstrated the potential application of microbubbles as a non-invasive, safe and effective technique for targeted delivery of drugs and genes. The drugs can be incorporated into the microbubbles in several different approaches and then carried to the site of interest where it can be released by destruction of the microbubbles using ultrasound to achieve the required therapeutic effect.Methods:The objective of this article is to report on a review of the recent advances of ultrasound-mediated microbubbles as a vehicle for delivering drugs and genes and its potential application for the treatment of cancer.Conclusion:Ultrasound-mediated microbubble technology has the potential to significantly improve chemotherapy drug delivery to treatment sites with minimal side effects. Moreover, the technology can induce temporary and reversible changes in the permeability of cells and vessels, thereby allowing for drug delivery in a spatially localised region which can improve the efficiency of drugs with poor bioavailability due to their poor absorption, rapid metabolism and rapid systemic elimination.
Collapse
|
13
|
Ishijima A, Yamaguchi S, Azuma T, Kobayashi E, Shibasaki Y, Nagamune T, Sakuma I. Selective intracellular delivery of perfluorocarbon nanodroplets for cytotoxicity threshold reduction on ultrasound-induced vaporization. Cancer Rep (Hoboken) 2020; 2:e1165. [PMID: 32721118 DOI: 10.1002/cnr2.1165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/21/2019] [Accepted: 01/25/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Phase-change nanodroplets (PCNDs), which are liquid perfluorocarbon nanoparticles, have garnered much attention as ultrasound-responsive nanomedicines. The vaporization phenomenon has been employed to treat tumors mechanically. However, the ultrasound pressure applied to induce vaporization must be low to avoid damage to nontarget tissues. AIMS Here, we report that the pressure threshold for vaporization to induce cytotoxicity can be significantly reduced by selective intracellular delivery of PCNDs into targeted tumors. METHODS AND RESULTS In vitro experiments revealed that selective intracellular delivery of PCNDs induced PCND aggregation specifically inside the targeted cells. This close-packed configuration decreased the pressure threshold for vaporization to induce cytotoxicity. Moreover, following ultrasound exposure, significant decrease was observed in the viability of cells that incorporated PCNDs (35%) but not in the viability of cells that did not incorporate PCNDs (88%). CONCLUSIONS Intracellular delivery of PCNDs reduced ultrasound pressure applied for vaporization to induce cytotoxicity. Confocal laser scanning microscopy and flow cytometry revealed that prolonged PCND-cell incubation increased PCND uptake and aggregation. This aggregation effect might have contributed to the cytotoxicity threshold reduction effect.
Collapse
Affiliation(s)
- Ayumu Ishijima
- Medical Device Development and Regulation Research Center, The University of Tokyo, Tokyo, Japan
| | - Satoshi Yamaguchi
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Takashi Azuma
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, Japan
| | - Etsuko Kobayashi
- Department of Precision Engineering, The University of Tokyo, Tokyo, Japan
| | - Yoshikazu Shibasaki
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Teruyuki Nagamune
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, The University of Tokyo, Tokyo, Japan
| | - Ichiro Sakuma
- Medical Device Development and Regulation Research Center, The University of Tokyo, Tokyo, Japan.,Department of Precision Engineering, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
14
|
Fernández-Toledano JC, Braeckeveldt B, Marengo M, De Coninck J. How Wettability Controls Nanoprinting. PHYSICAL REVIEW LETTERS 2020; 124:224503. [PMID: 32567897 DOI: 10.1103/physrevlett.124.224503] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
Using large scale molecular dynamics simulations, we study in detail the impact of nanometer droplets of low viscosity on flat substrates versus the wettability of the solid plate. The comparison between the molecular dynamics simulations and different macroscopic models reveals that most of these models do not correspond to the simulation results at the nanoscale, in particular for the maximal contact diameter during the nanodroplet impact (D_{max}). We have developed a new model for D_{max} that is in agreement with the simulation data and also takes into account the effects of the liquid-solid wettability. We also propose a new scaling for the time required to reach the maximal contact diameter t_{max} with respect to the impact velocity, which is also in agreement with the observations. With the new model for D_{max} plus the scaling found for t_{max}, we present a master curve collapsing the evolution of the nanometer drop contact diameter during impact for different wettabilities and different impact velocities. We believe our results may help in designing better nanoprinters since they provide an estimation of the maximum impact velocities required to obtain a smooth and homogenous coverage of the surfaces without dry spots.
Collapse
Affiliation(s)
| | - Bertrand Braeckeveldt
- Laboratory of Surface and Interfacial Physics (LPSI), Department of Physics, University of Mons, Mons 7000, Belgium
| | - Marco Marengo
- Advanced Engineering Centre, University of Brighton, Brighton BN2 4GJ, United Kingdom
| | - Joël De Coninck
- Laboratory of Surface and Interfacial Physics (LPSI), Department of Physics, University of Mons, Mons 7000, Belgium
| |
Collapse
|
15
|
Toumia Y, Cerroni B, Domenici F, Lange H, Bianchi L, Cociorb M, Brasili F, Chiessi E, D'Agostino E, Van Den Abeele K, Heymans SV, D'Hooge J, Paradossi G. Phase Change Ultrasound Contrast Agents with a Photopolymerized Diacetylene Shell. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10116-10127. [PMID: 31042396 DOI: 10.1021/acs.langmuir.9b01160] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Phase change contrast agents for ultrasound (US) imaging consist of nanodroplets (NDs) with a perfluorocarbon (PFC) liquid core stabilized with a lipid or a polymer shell. Liquid ↔ gas transition, occurring in the core, can be triggered by US to produce acoustically active microbubbles (MBs) in a process named acoustic droplet vaporization (ADV). MB shells containing polymerized diacetylene moiety were considered as a good trade off between the lipid MBs, showing optimal attenuation, and the polymeric ones, displaying enhanced stability. This work reports on novel perfluoropentane and perfluorobutane NDs stabilized with a monolayer of an amphiphilic fatty acid, i.e. 10,12-pentacosadiynoic acid (PCDA), cured with ultraviolet (UV) irradiation. The photopolymerization of the diacetylene groups, evidenced by the appearance of a blue color due to the conjugation of ene-yne sequences, exhibits a chromatic transition from the nonfluorescent blue color to a fluorescent red color when the NDs are heated or the pH of the suspension is basic. An estimate of the molecular weights reached by the polymerized PCDA in the shell, poly(PCDA), has been obtained using gel permeation chromatography and MALDI-TOF mass spectrometry. The poly(PCDA)/PFC NDs show good biocompatibility with fibroblast cells. ADV efficiency and acoustic properties before and after the transition were tested using a 1 MHz probe, revealing a resonance frequency between 1 and 2 MHz similar to other lipidic MBs. The surface of PCDA shelled NDs can be easily modified without influencing the stability and the acoustic performances of droplets. As a proof of concept we report on the conjugation of cyclic RGD and PEG chains of the particles to support targeting ability toward endothelial cells.
Collapse
Affiliation(s)
- Yosra Toumia
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Barbara Cerroni
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Fabio Domenici
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Heiko Lange
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Livia Bianchi
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Madalina Cociorb
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Francesco Brasili
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Ester Chiessi
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| | - Emiliano D'Agostino
- DoseVue NV , Philips Open Manufacturing Campus , Slachthuisstraat 96 , B-2300 Turnhout , Belgium
| | | | - Sophie V Heymans
- Department of Physics , KU Leuven , Kulak, 8500 Kortrijk , Belgium
| | - Jan D'Hooge
- Medical Center , KU Leuven , 3000 Leuven , Belgium
| | - Gaio Paradossi
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Via della Ricerca Scientifica 1 , 00133 , Rome , Italy
| |
Collapse
|
16
|
Tu J, Zhang H, Yu J, Liufu C, Chen Z. Ultrasound-mediated microbubble destruction: a new method in cancer immunotherapy. Onco Targets Ther 2018; 11:5763-5775. [PMID: 30254469 PMCID: PMC6140758 DOI: 10.2147/ott.s171019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immunotherapy provides a new treatment option for cancer. However, it may be therapeutically insufficient if only using the self-immune system alone to attack the tumor without any aiding methods. To overcome this drawback and improve the efficiency of therapy, new treatment methods are emerging. In recent years, ultrasound-mediated microbubble destruction (UMMD) has shown great potential in cancer immunotherapy. Using the combination of ultrasound and targeted microbubbles, molecules such as antigens or genes encoding antigens can be efficiently and specifically delivered into the tumor tissue. This review focuses on the recent progress in the application of UMMD in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiawei Tu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Hui Zhang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Jinsui Yu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Chun Liufu
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| | - Zhiyi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, People's Republic of China,
| |
Collapse
|
17
|
Sahle FF, Gulfam M, Lowe TL. Design strategies for physical-stimuli-responsive programmable nanotherapeutics. Drug Discov Today 2018; 23:992-1006. [PMID: 29653291 PMCID: PMC6195679 DOI: 10.1016/j.drudis.2018.04.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/31/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022]
Abstract
Nanomaterials that respond to externally applied physical stimuli such as temperature, light, ultrasound, magnetic field and electric field have shown great potential for controlled and targeted delivery of therapeutic agents. However, the body of literature on programming these stimuli-responsive nanomaterials to attain the desired level of pharmacologic responses is still fragmented and has not been systematically reviewed. The purpose of this review is to summarize and synthesize the literature on various design strategies for simple and sophisticated programmable physical-stimuli-responsive nanotherapeutics.
Collapse
Affiliation(s)
- Fitsum Feleke Sahle
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Muhammad Gulfam
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Tao L Lowe
- Department of Pharmaceutical Sciences, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA.
| |
Collapse
|