1
|
Ke Q, Qin Z, Yang X, Meng Q, Huang X, Kou X, Zhang Y. Mechanical properties of carriers based on natural polymers: Polysaccharides, proteins, and lipids as wall materials. Carbohydr Polym 2025; 362:123699. [PMID: 40409831 DOI: 10.1016/j.carbpol.2025.123699] [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: 02/27/2025] [Revised: 04/14/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025]
Abstract
Traditional synthetic polymer carriers are restricted due to microplastic pollution, whereas, natural polymer materials have gained widespread use as wall materials for carriers due to their biodegradability, availability, ease of modification, and biocompatibility. The mechanical properties of carriers are particularly crucial for formulation design, storage stability, and practical performance. However, there is currently a lack of reviews on the mechanical properties of natural polymer-based carriers (NPC). This paper delves into the mechanical properties of NPC from five aspects: First, natural polymer wall materials are classified into polysaccharide-based, protein-based, lipid-based, and composite materials, focusing on polysaccharide-dominated systems, and the mechanical properties of NPC constructed from materials of different origins are summarized. Second, various preparation techniques for NPC are introduced, summarizing the mechanical properties of carriers constructed by each method. The paper then examines regulation strategies of the mechanical properties of NPC, including modification techniques, encapsulated substances, morphology, and particle size. Next, methods for characterizing mechanical properties of NPC are introduced. Finally, there is a summary of the progress of NPCs with different mechanical properties in fields, highlighting the challenges faced and proposing future research directions. This review links mechanical optimization to performance, bridging research and applications with eco-friendly NPC strategies.
Collapse
Affiliation(s)
- Qinfei Ke
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhaoyuan Qin
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China
| | - Xingxing Yang
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China
| | - Qingran Meng
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China
| | - Xin Huang
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China
| | - Xingran Kou
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China.
| | - Yunchong Zhang
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology (Shanghai Research Institute of Fragrance & Flavour Industry), Shanghai Institute of Technology, Shanghai 201418, China.
| |
Collapse
|
2
|
Cunha J, Latocheski E, Fidalgo ACD, Gerola AP, Marin CFDF, Ribeiro AJ. Core-shell hybrid liposomes: Transforming imaging diagnostics and therapeutic strategies. Colloids Surf B Biointerfaces 2025; 251:114597. [PMID: 40043539 DOI: 10.1016/j.colsurfb.2025.114597] [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: 01/09/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 04/15/2025]
Abstract
For the last few years, researchers and industry have intensified efforts to develop a diverse array of diagnostic and therapeutic approaches to fight diseases such as cancer, diabetes, and viral infections. Among the emerging technologies, hybrid liposomes (HLs) stand out for their ability to address key limitations of conventional liposomes and deliver multifunctional solutions more effectively. While several novel nanosystems, including polymerlipid conjugates and inorganic nanoparticles (NPs), have shown great potential in the preclinical and clinical phases for the diagnosis and treatment of diseases, particularly cancer, HLs can integrate the best of both worlds, combining drug delivery properties with imaging capabilities. HLs, particularly those with core-shell structures, can surpass conventional liposomes by offering improved physicochemical properties, multifunctionality, and the capacity to overcome critical delivery challenges. The integration of natural and synthetic polymers has rapidly emerged as a preferred strategy in the development of HLs, providing significant advantages, such as enhanced stability, stimuli-responsive drug release, prolonged circulation, and improved therapeutic efficacy. Additionally, the customizable structure of HLs allows the incorporation of diverse materials, such as metals, ligands, and functional lipids, improving diagnosis and enhancing targeted delivery and cellular uptake far beyond what conventional liposomes offer. This review provides a critical and updated analysis of core-shell structure exhibiting HLs, with a focus on their preparation, characterization, and functional enhancements. We also examine in vitro/in vivo outcomes in imaging diagnosis and drug delivery while addressing the current barriers to clinical translation and future prospects for these versatile nanoplatforms.
Collapse
Affiliation(s)
- Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal
| | - Eloah Latocheski
- Department of Chemistry, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | | | | | | | - António José Ribeiro
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra 3000-548, Portugal; Group Genetics of Cognitive Dysfunction, I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4169-007, Portugal.
| |
Collapse
|
3
|
Zeng W, Huang Z, Huang Y, Xiong K, Sheng Y, Lin X, Zhong X, Ye J, Guo Y, Arkin G, Xu J, Fei H, Liu Y. Dual-targeted microbubbles for atherosclerosis therapy: Inducing M1 macrophage apoptosis by inhibiting telomerase activity. Mater Today Bio 2025; 32:101675. [PMID: 40225135 PMCID: PMC11986608 DOI: 10.1016/j.mtbio.2025.101675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 03/05/2025] [Accepted: 03/16/2025] [Indexed: 04/15/2025] Open
Abstract
The progression of atherosclerosis (AS) is closely associated with M1 macrophages. Although the activation of macrophage telomerase during plaque formation has been documented, targeted modulation strategies remain challenging. In this study, we developed a dual-target microbubble-delivery system (Ab-MMB1532) encapsulating BIBR1532, a telomerase inhibitor, for the targeted therapy of AS. This system exhibited remarkable targeting capabilities towards M1 macrophages, with its targeting advantage notably accentuated under high shear forces. Mechanistically, Ab-MMB1532 inhibited telomerase activity by downregulating telomerase reverse transcriptase (TERT) expression, subsequently inducing caspase-3-mediated apoptosis. Integrated multi-omics profiling revealed that the inhibition of the NF-κB pathway served as the central regulatory hub. In vivo studies further confirmed that Ab-MMB1532 effectively targets and accumulates within AS lesions, promoting M1 macrophage apoptosis through the inhibition of the TERT/NF-κB signaling axis, and significantly reducing plaque burden (25.4 % reduction vs. controls, p < 0.001). In summary, our findings suggest a novel approach for telomerase-targeted therapy in AS.
Collapse
Affiliation(s)
- Wei Zeng
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Zhengan Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 519041, China
| | - Yalan Huang
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
- Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, 510632, China
| | - Kaifen Xiong
- Department of Dermatology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Yuanyuan Sheng
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xiaoxuan Lin
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Xiaofang Zhong
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Jiayu Ye
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Yanbin Guo
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Gulzira Arkin
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Jinfeng Xu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Hongwen Fei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 519041, China
| | - Yingying Liu
- Shenzhen Medical Ultrasound Engineering Center, Department of Ultrasonography, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| |
Collapse
|
4
|
Fernandes DA. Comprehensive Review on Bubbles: Synthesis, Modification, Characterization and Biomedical Applications. Bioconjug Chem 2024; 35:1639-1686. [PMID: 39377727 DOI: 10.1021/acs.bioconjchem.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Accurate detection, treatment, and imaging of diseases are important for effective treatment outcomes in patients. In this regard, bubbles have gained much attention, due to their versatility. Bubbles usually 1 nm to 10 μm in size can be produced and loaded with a variety of lipids, polymers, proteins, and therapeutic and imaging agents. This review details the different production and loading methods for bubbles, for imaging and treatment of diseases/conditions such as cancer, tumor angiogenesis, thrombosis, and inflammation. Bubbles can also be used for perfusion measurements, important for diagnostic and therapeutic decision making in cardiac disease. The different factors important in the stability of bubbles and the different techniques for characterizing their physical and chemical properties are explained, for developing bubbles with advanced therapeutic and imaging features. Hence, the review provides important insights for researchers studying bubbles for biomedical applications.
Collapse
|
5
|
Zhang S, Fang H, Tian H. Recent Advances in Degradable Biomedical Polymers for Prevention, Diagnosis and Treatment of Diseases. Biomacromolecules 2024; 25:7015-7057. [PMID: 39420482 DOI: 10.1021/acs.biomac.4c01193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Biomedical polymers play a key role in preventing, diagnosing, and treating diseases, showcasing a wide range of applications. Their unique advantages, such as rich source, good biocompatibility, and excellent modifiability, make them ideal biomaterials for drug delivery, biomedical imaging, and tissue engineering. However, conventional biomedical polymers suffer from poor degradation in vivo, increasing the risks of bioaccumulation and potential toxicity. To address these issues, degradable biomedical polymers can serve as an alternative strategy in biomedicine. Degradable biomedical polymers can efficiently relieve bioaccumulation in vivo and effectively reduce patient burden in disease management. This review comprehensively introduces the classification and properties of biomedical polymers and the recent research progress of degradable biomedical polymers in various diseases. Through an in-depth analysis of their classification, properties, and applications, we aim to provide strong guidance for promoting basic research and clinical translation of degradable biomedical polymers.
Collapse
Affiliation(s)
- Siting Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
6
|
Qu Z, Luo J, Li Z, Yang R, Zhao J, Chen X, Yu S, Shu H. Advancements in strategies for overcoming the blood-brain barrier to deliver brain-targeted drugs. Front Aging Neurosci 2024; 16:1353003. [PMID: 39253614 PMCID: PMC11381257 DOI: 10.3389/fnagi.2024.1353003] [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: 12/09/2023] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier is known to consist of a variety of cells and complex inter-cellular junctions that protect the vulnerable brain from neurotoxic compounds; however, it also complicates the pharmacological treatment of central nervous system disorders as most drugs are unable to penetrate the blood-brain barrier on the basis of their own structural properties. This dramatically diminished the therapeutic effect of the drug and compromised its biosafety. In response, a number of drugs are often delivered to brain lesions in invasive ways that bypass the obstruction of the blood-brain barrier, such as subdural administration, intrathecal administration, and convection-enhanced delivery. Nevertheless, these intrusive strategies introduce the risk of brain injury, limiting their clinical application. In recent years, the intensive development of nanomaterials science and the interdisciplinary convergence of medical engineering have brought light to the penetration of the blood-brain barrier for brain-targeted drugs. In this paper, we extensively discuss the limitations of the blood-brain barrier on drug delivery and non-invasive brain-targeted strategies such as nanomedicine and blood-brain barrier disruption. In the meantime, we analyze their strengths and limitations and provide outlooks on the further development of brain-targeted drug delivery systems.
Collapse
Affiliation(s)
- Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, China
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Juan Luo
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zheng Li
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Yang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaxi Zhao
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
7
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
8
|
Lin J, Chen X, Li Y, Yu L, Chen Y, Zhang B. A dual-targeting therapeutic nanobubble for imaging-guided atherosclerosis treatment. Mater Today Bio 2024; 26:101037. [PMID: 38586870 PMCID: PMC10995877 DOI: 10.1016/j.mtbio.2024.101037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
Atherosclerosis is a cardiovascular disease that seriously endangers human health. Low shear stress (LSS) is recognized as a vital factor in causing chronic inflammatory and further inducing the occurrence and development of atherosclerosis. Targeting imaging and treatment are of substantial significance for the diagnosis and therapy of atherosclerosis. On this ground, a kind of ultrasound (US) imaging-guided therapeutic polymer nanobubbles (NBs) with dual targeting of magnetism and antibody was rationally designed and constructed for the efficiently treating LSS-mediated atherosclerosis. Under the combined targeting effect of an external magnetic field and antibodies, the drug-loaded therapeutic NBs can be effectively accumulated in the inflammatory area caused by LSS. Upon US irradiation, the NBs can be selectively disrupted, leading to the rapid release of the loaded drugs at the targeted site. Notably, the US irradiation generates a cavitation effect that induces repairable micro gaps in nearby cells, thereby enhancing the uptake of released drugs and further improving the therapeutic effect. The prominent US imaging, efficient anti-inflammatory effect and treatment outcome of LSS-mediated atherosclerosis had been verified in vivo on a surgically constructed LSS-atherosclerosis animal model. This work showcased the potential of the designed NBs with multifunctionality for in vivo imaging, dual-targeting, and drug delivery in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jie Lin
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Xiaoying Chen
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Yi Li
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Luodan Yu
- Department of Radiology, Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
- Shanghai Institute of Materdicine, Shanghai, 200051, PR China
| | - Bo Zhang
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| |
Collapse
|
9
|
Dong F, An J, Guo W, Dang J, Huang S, Feng F, Zhang J, Wang D, Yin J, Fang J, Cheng H, Zhang J. Programmable ultrasound imaging guided theranostic nanodroplet destruction for precision therapy of breast cancer. ULTRASONICS SONOCHEMISTRY 2024; 105:106854. [PMID: 38537562 PMCID: PMC11059134 DOI: 10.1016/j.ultsonch.2024.106854] [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: 06/02/2023] [Revised: 03/17/2024] [Accepted: 03/23/2024] [Indexed: 04/26/2024]
Abstract
Ultrasound-stimulated contrast agents have gained significant attention in the field of tumor treatment as drug delivery systems. However, their limited drug-loading efficiency and the issue of bulky, imprecise release have resulted in inadequate drug concentrations at targeted tissues. Herein, we developed a highly efficient approach for doxorubicin (DOX) precise release at tumor site and real-time feedback via an integrated strategy of "programmable ultrasonic imaging guided accurate nanodroplet destruction for drug release" (PND). We synthesized DOX-loaded nanodroplets (DOX-NDs) with improved loading efficiency (15 %) and smaller size (mean particle size: 358 nm). These DOX-NDs exhibited lower ultrasound activation thresholds (2.46 MPa). By utilizing a single diagnostic transducer for both ultrasound stimulation and imaging guidance, we successfully vaporized the DOX-NDs and released the drug at the tumor site in 4 T1 tumor-bearing mice. Remarkably, the PND group achieved similar tumor remission effects with less than half the dose of DOX required in conventional treatment. Furthermore, the ultrasound-mediated vaporization of DOX-NDs induced tumor cell apoptosis with minimal damage to surrounding normal tissues. In summary, our PND strategy offers a precise and programmable approach for drug delivery and therapy, combining ultrasound imaging guidance. This approach shows great potential in enhancing tumor treatment efficacy while minimizing harm to healthy tissues.
Collapse
Affiliation(s)
- Feihong Dong
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Jian An
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Wenyu Guo
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jie Dang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuo Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Feng Feng
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jiabin Zhang
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Di Wang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jingyi Yin
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing Fang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; College of Engineering, Peking University, Beijing 100871, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, National Biomedical Imaging Center, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; National Biomedical Imaging Center, Peking University, Beijing, 100871, China; Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational Medicine, Nanjing, 211899, China.
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; College of Engineering, Peking University, Beijing 100871, China; National Biomedical Imaging Center, Peking University, Beijing, 100871, China.
| |
Collapse
|
10
|
Su T, Zhao F, Ying Y, Li W, Li J, Zheng J, Qiao L, Che S, Yu J. Self-Monitoring Theranostic Nanomaterials: Emerging Visual Agents for Real-Time Monitoring of Tumor Treatment Processes. SMALL METHODS 2024; 8:e2301470. [PMID: 38044269 DOI: 10.1002/smtd.202301470] [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: 10/24/2023] [Revised: 11/14/2023] [Indexed: 12/05/2023]
Abstract
Self-monitoring in tumor therapy is a concept that allows for real-time monitoring of the location and state of applied nanomaterials. This monitoring relies on dynamic signals, such as wave or magnetic signals, which vary in response to changes in the location and state of nanomaterials. Dynamic changes in nanomaterials can be monitored using dynamic signals, making it possible to determine and control the treatment process. Theranostic nanomaterials, which possess unique physical and chemical properties, have recently been explored as a viable option for self-monitoring. With the help of self-monitoring, theranostic nanomaterials can guide themselves to achieve region-selective treatment with higher controllability and safety. In this review, self-monitoring theranostic nanomaterials will be introduced in three parts according to their roles during therapy: tumor accumulation, tumor therapy, and metabolism. The limitations and future challenges of current self-monitoring theranostic nanomaterials will also be discussed.
Collapse
Affiliation(s)
- Tuo Su
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Fan Zhao
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yao Ying
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Wangchang Li
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Juan Li
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jingwu Zheng
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Liang Qiao
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shenglei Che
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jing Yu
- College of Materials Science and Engineering, Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
11
|
Gusliakova OI, Kurochkin MA, Barmin RA, Prikhozhdenko ES, Estifeeva TM, Rudakovskaya PG, Sindeeva OA, Galushka VV, Vavaev ES, Komlev AS, Lyubin EV, Fedyanin AA, Dey KK, Gorin DA. Magnetically navigated microbubbles coated with albumin/polyarginine and superparamagnetic iron oxide nanoparticles. BIOMATERIALS ADVANCES 2024; 158:213759. [PMID: 38227987 DOI: 10.1016/j.bioadv.2024.213759] [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: 09/22/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
While microbubbles (MB) are routinely used for ultrasound (US) imaging, magnetic MB are increasingly explored as they can be guided to specific sites of interest by applied magnetic field gradient. This requires the MB shell composition tuning to prolong MB stability and provide functionalization capabilities with magnetic nanoparticles. Hence, we developed air-filled MB stabilized by a protein-polymer complex of bovine serum albumin (BSA) and poly-L-arginine (pArg) of different molecular weights, showing that pArg of moderate molecular weight distribution (15-70 kDa) enabled MB with greater stability and acoustic response while preserving MB narrow diameters and the relative viability of THP-1 cells after 48 h of incubation. After MB functionalization with superparamagnetic iron oxide nanoparticles (SPION), magnetic moment values provided by single MB confirmed the sufficient SPION deposition onto BSA + pArg MB shells. During MB magnetic navigation in a blood vessel mimicking phantom with magnetic tweezers and in a Petri dish with adherent mouse renal carcinoma cell line, we demonstrated the effectiveness of magnetic MB localization in the desired area by magnetic field gradient. Magnetic MB co-localization with cells was further exploited for effective doxorubicin delivery with drug-loaded MB. Taken together, these findings open new avenues in control over albumin MB properties and magnetic navigation of SPION-loaded MB, which can envisage their applications in diagnostic and therapeutic needs.
Collapse
Affiliation(s)
- Olga I Gusliakova
- Science Medical Center, Saratov State University, Saratov 410012, Russia; Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| | - Maxim A Kurochkin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Roman A Barmin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | | | - Tatyana M Estifeeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Polina G Rudakovskaya
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Olga A Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Victor V Galushka
- Education and Research Institute of Nanostructures and Biosystems, Saratov State University, Saratov 410012, Russia
| | - Evgeny S Vavaev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Aleksei S Komlev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Evgeny V Lyubin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Andrey A Fedyanin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Krishna Kanti Dey
- Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382055, India
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| |
Collapse
|
12
|
Abstract
In order to improve bioavailability, stability, control release, and target delivery of active pharmaceutical ingredients (APIs), as well as to mask their bitter taste, to increase their efficacy, and to minimize their side effects, a variety of microencapsulation (including nanoencapsulation, particle size <100 nm) technologies have been widely used in the pharmaceutical industry. Commonly used microencapsulation technologies are emulsion, coacervation, extrusion, spray drying, freeze-drying, molecular inclusion, microbubbles and microsponge, fluidized bed coating, supercritical fluid encapsulation, electro spinning/spray, and polymerization. In this review, APIs are categorized by their molecular complexity: small APIs (compounds with low molecular weight, like Aspirin, Ibuprofen, and Cannabidiol), medium APIs (compounds with medium molecular weight like insulin, peptides, and nucleic acids), and living microorganisms (such as probiotics, bacteria, and bacteriophages). This article provides an overview of these microencapsulation technologies including their processes, matrix, and their recent applications in microencapsulation of APIs. Furthermore, the advantages and disadvantages of these common microencapsulation technologies in terms of improving the efficacy of APIs for pharmaceutical treatments are comprehensively analyzed. The objective is to summarize the most recent progresses on microencapsulation of APIs for enhancing their bioavailability, control release, target delivery, masking their bitter taste and stability, and thus increasing their efficacy and minimizing their side effects. At the end, future perspectives on microencapsulation for pharmaceutical applications are highlighted.
Collapse
Affiliation(s)
- Cuie Yan
- Division of Encapsulation, Blue California, Rancho Santa Margarita, California 92688, United States
| | - Sang-Ryoung Kim
- Division of Encapsulation, Blue California, Rancho Santa Margarita, California 92688, United States
| |
Collapse
|
13
|
Jang Y, Park J, Kim P, Park EJ, Sun H, Baek Y, Jung J, Song TK, Doh J, Kim H. Development of exosome membrane materials-fused microbubbles for enhanced stability and efficient drug delivery of ultrasound contrast agent. Acta Pharm Sin B 2023; 13:4983-4998. [PMID: 38045059 PMCID: PMC10692476 DOI: 10.1016/j.apsb.2023.08.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 12/05/2023] Open
Abstract
Lipid-coated microbubbles are widely used as an ultrasound contrast agent, as well as drug delivery carriers. However, the two main limitations in ultrasound diagnosis and drug delivery using microbubbles are the short half-life in the blood system, and the difficulty of surface modification of microbubbles for active targeting. The exosome, a type of extracellular vesicle, has a preferentially targeting ability for its original cell. In this study, exosome-fused microbubbles (Exo-MBs) were developed by embedding the exosome membrane proteins into microbubbles. As a result, the stability of Exo-MBs is improved over the conventional microbubbles. On the same principle that under the exposure of ultrasound, microbubbles are cavitated and self-assembled into nano-sized particles, and Exo-MBs are self-assembled into exosome membrane proteins-embedded nanoparticles (Exo-NPs). The Exo-NPs showed favorable targeting properties to their original cells. A photosensitizer, chlorin e6, was loaded into Exo-MBs to evaluate therapeutic efficacy as a drug carrier. Much higher therapeutic efficacy of photodynamic therapy was confirmed, followed by cancer immunotherapy from immunogenic cell death. We have therefore developed a novel ultrasound image-guided drug delivery platform that overcomes the shortcomings of the conventional ultrasound contrast agent and is capable of simultaneous photodynamic therapy and cancer immunotherapy.
Collapse
Affiliation(s)
- Yongho Jang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Jeehun Park
- Research Institute of Advanced Materials (RIAM), Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea
| | - Pilsu Kim
- Department of Electronic Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Eun-Joo Park
- Biomedical Research Institute & Department of Radiology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Hyungjin Sun
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yujin Baek
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehun Jung
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Tai-kyong Song
- Department of Electronic Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Junsang Doh
- Research Institute of Advanced Materials (RIAM), Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Engineering Research, Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyuncheol Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Republic of Korea
| |
Collapse
|
14
|
Barmin RA, Moosavifar M, Dasgupta A, Herrmann A, Kiessling F, Pallares RM, Lammers T. Polymeric materials for ultrasound imaging and therapy. Chem Sci 2023; 14:11941-11954. [PMID: 37969594 PMCID: PMC10631124 DOI: 10.1039/d3sc04339h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/11/2023] [Indexed: 11/17/2023] Open
Abstract
Ultrasound (US) is routinely used for diagnostic imaging and increasingly employed for therapeutic applications. Materials that act as cavitation nuclei can improve the resolution of US imaging, and facilitate therapeutic US procedures by promoting local drug delivery or allowing temporary biological barrier opening at moderate acoustic powers. Polymeric materials offer a high degree of control over physicochemical features concerning responsiveness to US, e.g. via tuning chain composition, length and rigidity. This level of control cannot be achieved by materials made of lipids or proteins. In this perspective, we present key engineered polymeric materials that respond to US, including microbubbles, gas-stabilizing nanocups, microcapsules and gas-releasing nanoparticles, and discuss their formulation aspects as well as their principles of US responsiveness. Focusing on microbubbles as the most common US-responsive polymeric materials, we further evaluate the available chemical toolbox to engineer polymer shell properties and enhance their performance in US imaging and US-mediated drug delivery. Additionally, we summarize emerging applications of polymeric microbubbles in molecular imaging, sonopermeation, and gas and drug delivery, based on refinement of MB shell properties. Altogether, this manuscript provides new perspectives on US-responsive polymeric designs, envisaging their current and future applications in US imaging and therapy.
Collapse
Affiliation(s)
- Roman A Barmin
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - MirJavad Moosavifar
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Andreas Herrmann
- DWI - Leibniz Institute for Interactive Materials Aachen 52074 Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Aachen 52074 Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Roger M Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital Aachen 52074 Germany
| |
Collapse
|
15
|
Chang H, Wang Q, Liu T, Chen L, Hong J, Liu K, Li Y, Yang N, Han D, Mi X, Li X, Guo X, Li Y, Li Z. A Bibliometric Analysis for Low-Intensity Ultrasound Study Over the Past Three Decades. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2023; 42:2215-2232. [PMID: 37129170 DOI: 10.1002/jum.16245] [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: 08/29/2022] [Revised: 03/29/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
Low-intensity ultrasound (LI-US) is a non-invasive stimulation technique that has emerged in recent years and has been shown to have positive effects on neuromodulation, fracture healing, inflammation improvement, and metabolic regulation. This study reports the conclusions of a bibliometric analysis of LI-US. Input data for the period between 1995 and 2022, including 7209 related articles in the field of LI-US, were collected from the core library of the Web of Science (WOS) database. Using these data, a set of bibliometric indicators was obtained to gain knowledge on different aspects: global production, research areas, and sources analysis, contributions of countries and institutions, author analysis, citation analysis, and keyword analysis. This study combined the data analysis capabilities provided by the WOS database, making use of two bibliometric software tools: R software and VOS viewer to achieve analysis and data exploration visualization, and predicted the further development trends of LI-US. It turns out that the United States and China are co-leaders while Zhang ZG is the most significant author in LI-US. In the future, the hot spots of LI-US will continue to focus on parameter research, mechanism discussion, safety regulations, and neuromodulation applications.
Collapse
Affiliation(s)
- Huixian Chang
- School of Information Science and Engineering, Yanshan University, Qinhuangdao, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Yitong Li
- School of Information Science and Engineering, Yanshan University, Qinhuangdao, China
| | - Ning Yang
- School of Information Science and Engineering, Yanshan University, Qinhuangdao, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Xiaoli Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
- Beijing Center of Quality Control and Improvement on Clinical Anesthesia, Beijing, China
| | - Yingwei Li
- School of Information Science and Engineering, Yanshan University, Qinhuangdao, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
- Beijing Center of Quality Control and Improvement on Clinical Anesthesia, Beijing, China
| |
Collapse
|
16
|
Nistor M, Rugina D, Diaconeasa Z, Socaciu C, Socaciu MA. Pentacyclic Triterpenoid Phytochemicals with Anticancer Activity: Updated Studies on Mechanisms and Targeted Delivery. Int J Mol Sci 2023; 24:12923. [PMID: 37629103 PMCID: PMC10455110 DOI: 10.3390/ijms241612923] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pentacyclic triterpenoids (TTs) represent a unique family of phytochemicals with interesting properties and pharmacological effects, with some representatives, such as betulinic acid (BA) and betulin (B), being mainly investigated as potential anticancer molecules. Considering the recent scientific and preclinical investigations, a review of their anticancer mechanisms, structure-related activity, and efficiency improved by their insertion in nanolipid vehicles for targeted delivery is presented. A systematic literature study about their effects on tumor cells in vitro and in vivo, as free molecules or encapsulated in liposomes or nanolipids, is discussed. A special approach is given to liposome-TTs and nanolipid-TTs complexes to be linked to microbubbles, known as contrast agents in ultrasonography. The production of such supramolecular conjugates to deliver the drugs to target cells via sonoporation represents a new scientific and applicative direction to improve TT efficiency, considering that they have limited availability as lipophilic molecules. Relevant and recent examples of in vitro and in vivo studies, as well as the challenges for the next steps towards the application of these complex delivery systems to tumor cells, are discussed, as are the challenges for the next steps towards the application of targeted delivery to tumor cells, opening new directions for innovative nanotechnological solutions.
Collapse
Affiliation(s)
- Madalina Nistor
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Dumitrita Rugina
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Zorita Diaconeasa
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Carmen Socaciu
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Mihai Adrian Socaciu
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
- Department of Radiology, Imaging & Nuclear Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400347 Cluj-Napoca, Romania
| |
Collapse
|
17
|
Hong Park J, Lee S, Jeon H, Hoon Kim J, Jung Kim D, Im M, Chul Lee B. A novel convex acoustic lens-attached ultrasound drug delivery system and its testing in a murine melanoma subcutaneous modelo. Int J Pharm 2023:123118. [PMID: 37302671 DOI: 10.1016/j.ijpharm.2023.123118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/18/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Target-specific drug release is indispensable to improve chemotherapeutic efficacy as it enhances drug uptake and penetration into tumors. Sono-responsive drug-loaded nano-/micro-particles are a promising solution for achieving target specificity by exposing them to ultrasound near tumors. However, the complicated synthetic processes and limited ultrasound (US) exposure conditions, such as limited control of ultrasound focal depth and acoustic power, prevent the practical application of this approach in clinical practice. Here, we propose a convex acoustic lens-attached US (CALUS) as a simple, economic, and efficient alternative of focused US for drug delivery system (DDS) application. The CALUS was characterized both numerically and experimentally using a hydrophone. In vitro, microbubbles (MBs) inside microfluidic channels were destroyed using the CALUS with various acoustic parameters (acoustic pressure [P], pulse repetition frequency [PRF], and duty cycle) and flow velocity. In vivo, tumor inhibition was evaluated using melanoma-bearing mice by characterizing tumor growth rate, animal weight, and intratumoral drug concentration with/without CALUS DDS. US beams were measured to be efficiently converged by CALUS, which was consistent with our simulation results. The acoustic parameters were optimized through the CALUS-induced MB destruction test (P = 2.34 MPa, PRF = 100 kHz, and duty cycle = 9%); this optimal parameter combination successfully induced MB destruction inside the microfluidic channel with an average flow velocity of up to 9.6 cm/s. The CALUS also enhanced the therapeutic effects of an antitumor drug (doxorubicin) in vivo in a murine melanoma model. The combination of the doxorubicin and the CALUS inhibited tumor growth by ∼55% more than doxorubicin alone, clearly indicating synergistic antitumor efficacy. Our tumor growth inhibition performance was better than other methods based on drug carriers, even without a time-consuming and complicated chemical synthesis process. This result suggests that our novel, simple, economic, and efficient target-specific DDS may offer a transition from preclinical research to clinical trials and a potential treatment approach for patient-centered healthcare.
Collapse
Affiliation(s)
- Jun Hong Park
- Bionics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Seunghyun Lee
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Hoyoon Jeon
- Bionics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jung Hoon Kim
- Department of Radiology, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul 03080, Republic of Korea
| | - Da Jung Kim
- Metabolomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Maesoon Im
- Brain Science Institute, KIST, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science & Technology (UST), Seoul 02792, Republic of Korea
| | - Byung Chul Lee
- Bionics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science & Technology (UST), Seoul 02792, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
18
|
Huang D, Wang J, Song C, Zhao Y. Ultrasound-responsive matters for biomedical applications. Innovation (N Y) 2023; 4:100421. [PMID: 37192908 PMCID: PMC10182333 DOI: 10.1016/j.xinn.2023.100421] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/03/2023] [Indexed: 05/18/2023] Open
Abstract
Ultrasound (US) is a biofavorable mechanical wave that has shown practical significance in biomedical fields. Due to the cavitation effect, sonoluminescence, sonoporation, pyrolysis, and other biophysical and chemical effects, a wide range of matters have been elucidated to be responsive to the stimulus of US. This review addresses and discusses current developments in US-responsive matters, including US-breakable intermolecular conjugations, US-catalytic sonosensitizers, fluorocarbon compounds, microbubbles, and US-propelled micro- and nanorobots. Meanwhile, the interactions between US and advanced matters create various biochemical products and enhanced mechanical effects, leading to the exploration of potential biomedical applications, from US-facilitated biosensing and diagnostic imaging to US-induced therapeutic applications and clinical translations. Finally, the current challenges are summarized and future perspectives on US-responsive matters in biomedical applications and clinical translations are proposed.
Collapse
Affiliation(s)
- Danqing Huang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Jinglin Wang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Chuanhui Song
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China
| |
Collapse
|
19
|
Hu Y, Wei J, Shen Y, Chen S, Chen X. Barrier-breaking effects of ultrasonic cavitation for drug delivery and biomarker release. ULTRASONICS SONOCHEMISTRY 2023; 94:106346. [PMID: 36870921 PMCID: PMC10040969 DOI: 10.1016/j.ultsonch.2023.106346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 05/27/2023]
Abstract
Recently, emerging evidence has demonstrated that cavitation actually creates important bidirectional channels on biological barriers for both intratumoral drug delivery and extratumoral biomarker release. To promote the barrier-breaking effects of cavitation for both therapy and diagnosis, we first reviewed recent technical advances of ultrasound and its contrast agents (microbubbles, nanodroplets, and gas-stabilizing nanoparticles) and then reported the newly-revealed cavitation physical details. In particular, we summarized five types of cellular responses of cavitation in breaking the plasma membrane (membrane retraction, sonoporation, endocytosis/exocytosis, blebbing and apoptosis) and compared the vascular cavitation effects of three different types of ultrasound contrast agents in breaking the blood-tumor barrier and tumor microenvironment. Moreover, we highlighted the current achievements of the barrier-breaking effects of cavitation in mediating drug delivery and biomarker release. We emphasized that the precise induction of a specific cavitation effect for barrier-breaking was still challenged by the complex combination of multiple acoustic and non-acoustic cavitation parameters. Therefore, we provided the cutting-edge in-situ cavitation imaging and feedback control methods and suggested the development of an international cavitation quantification standard for the clinical guidance of cavitation-mediated barrier-breaking effects.
Collapse
Affiliation(s)
- Yaxin Hu
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Jianpeng Wei
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Yuanyuan Shen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Siping Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Xin Chen
- School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, Guangdong, 518060, PR China; National-regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong, 518060, PR China.
| |
Collapse
|
20
|
Li H, Lv W, Zhang Y, Feng Q, Wu H, Su C, Shu H, Nie F. PLGA-PEI nanobubbles carrying PDLIM5 siRNA inhibit EGFR-TKI-resistant NSCLC cell migration and invasion ability using UTND technology. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
|
21
|
Huang D, Wang J, Che J, Wen B, Kong W. Ultrasound-responsive microparticles from droplet microfluidics. BIOMEDICAL TECHNOLOGY 2023; 1:1-9. [DOI: 10.1016/j.bmt.2022.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
|
22
|
Moradi Kashkooli F, Jakhmola A, Hornsby TK, Tavakkoli JJ, Kolios MC. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions. J Control Release 2023; 355:552-578. [PMID: 36773959 DOI: 10.1016/j.jconrel.2023.02.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The application of biocompatible nanocarriers in medicine has provided several benefits over conventional treatment methods. However, achieving high treatment efficacy and deep penetration of nanocarriers in tumor tissue is still challenging. To address this, stimuli-responsive nano-sized drug delivery systems (DDSs) are an active area of investigation in delivering anticancer drugs. While ultrasound is mainly used for diagnostic purposes, it can also be applied to affect cellular function and the delivery/release of anticancer drugs. Therapeutic ultrasound (TUS) has shown potential as both a stand-alone anticancer treatment and a method to induce targeted drug release from nanocarrier systems. TUS approaches have been used to overcome various physiological obstacles, including endothelial barriers, the tumor microenvironment (TME), and immunological hurdles. Combining nanomedicine and ultrasound as a smart DDS can increase in situ drug delivery and improve access to impermeable tissues. Furthermore, smart DDSs can perform targeted drug release in response to distinctive TMEs, external triggers, or dual/multi-stimulus. This results in enhanced treatment efficacy and reduced damage to surrounding healthy tissue or organs at risk. Integrating DDSs and ultrasound is still in its early stages. More research and clinical trials are required to fully understand ultrasound's underlying physical mechanisms and interactions with various types of nanocarriers and different types of cells and tissues. In the present review, ultrasound-mediated nano-sized DDS, specifically focused on cancer treatment, is presented and discussed. Ultrasound interaction with nanoparticles (NPs), drug release mechanisms, and various types of ultrasound-sensitive NPs are examined. Additionally, in vitro, in vivo, and clinical applications of TUS are reviewed in light of the critical challenges that need to be considered to advance TUS toward an efficient, secure, straightforward, and accessible cancer treatment. This study also presents effective TUS parameters and safety considerations for this treatment modality and gives recommendations about system design and operation. Finally, future perspectives are considered, and different TUS approaches are examined and discussed in detail. This review investigates drug release and delivery through ultrasound-mediated nano-sized cancer treatment, both pre-clinically and clinically.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
23
|
Zeng W, Yue X, Dai Z. Ultrasound contrast agents from microbubbles to biogenic gas vesicles. MEDICAL REVIEW (2021) 2023; 3:31-48. [PMID: 37724107 PMCID: PMC10471104 DOI: 10.1515/mr-2022-0020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/11/2022] [Indexed: 09/20/2023]
Abstract
Microbubbles have been the earliest and most widely used ultrasound contrast agents by virtue of their unique features: such as non-toxicity, intravenous injectability, ability to cross the pulmonary capillary bed, and significant enhancement of echo signals for the duration of the examination, resulting in essential preclinical and clinical applications. The use of microbubbles functionalized with targeting ligands to bind to specific targets in the bloodstream has further enabled ultrasound molecular imaging. Nevertheless, it is very challenging to utilize targeted microbubbles for molecular imaging of extravascular targets due to their size. A series of acoustic nanomaterials have been developed for breaking free from this constraint. Especially, biogenic gas vesicles, gas-filled protein nanostructures from microorganisms, were engineered as the first biomolecular ultrasound contrast agents, opening the door for more direct visualization of cellular and molecular function by ultrasound imaging. The ordered protein shell structure and unique gas filling mechanism of biogenic gas vesicles endow them with excellent stability and attractive acoustic responses. What's more, their genetic encodability enables them to act as acoustic reporter genes. This article reviews the upgrading progresses of ultrasound contrast agents from microbubbles to biogenic gas vesicles, and the opportunities and challenges for the commercial and clinical translation of the nascent field of biomolecular ultrasound.
Collapse
Affiliation(s)
- Wenlong Zeng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Xiuli Yue
- School of Environment, Harbin Institute of Technology, Harbin, China
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| |
Collapse
|
24
|
Ultrasound-induced destruction of heparin-loaded microbubbles attenuates L-arginine-induced acute pancreatitis. Eur J Pharm Sci 2023; 180:106318. [PMID: 36332825 DOI: 10.1016/j.ejps.2022.106318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Acute pancreatitis (AP) involves sudden inflammation caused by abnormal activation of pancreatic enzymes. The mechanisms underlying AP include oxidative stress, high levels of inflammatory mediators and inflammatory cell infiltration. Heparin, a key therapeutic drug, exerts anti-inflammatory, antioxidative, and anticoagulative effects. However, safe and effective drug delivery remains an obstacle. This study is the first to investigate the therapeutic effects of heparin-loaded microbubbles (HPMB) combined with ultrasound (UHPMB) and the role of heparin in acoustic cavitation. METHODS The characteristics of the microbubbles, including particle size, concentration, release, stability, and development, were studied. Heparin concentration in the HPMB was measured, and heparin-induced anticoagulation was evaluated. Drug safety was explored using hemolysis and cell viability assessments. The ability of HPMB to alleviate oxidative stress and inflammation were investigated in vitro. L-arginine induces AP in vivo. UHPMB was used for AP treatment. Serum amylase levels were measured and pancreatic architecture and pathological features were evaluated to determine AP severity. In vivo efficacy was evaluated, and the underlying mechanism of heparin action during acoustic cavitation was explored. RESULTS HPMB was spherical and presented as an emulsion-like solution without aggregation. HPMB was visible and stable and effectively released the drug under ultrasound (US). HPMB and UHPMB led to lower AP severity than in the untreated group. US-targeted microbubble destruction (UTMD) enhanced the therapeutic effect by decreasing oxidative stress and inflammation in AP models without injuring vital organs. UHPMB regulated VEGF/Flt-1 and SOD-1 expression. HPMB can also mitigate oxidative stress and inflammation in H2O2-pretreated cells. CONCLUSION UHPMB exhibits a strong ability not only to selectively target pancreatic lesions and release heparin but also to provide efficient protection by inhibiting oxidative stress and inflammation.
Collapse
|
25
|
Alsaab HO, Alharbi FD, Alhibs AS, Alanazi NB, Alshehri BY, Saleh MA, Alshehri FS, Algarni MA, Almugaiteeb T, Uddin MN, Alzhrani RM. PLGA-Based Nanomedicine: History of Advancement and Development in Clinical Applications of Multiple Diseases. Pharmaceutics 2022; 14:pharmaceutics14122728. [PMID: 36559223 PMCID: PMC9786338 DOI: 10.3390/pharmaceutics14122728] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Research on the use of biodegradable polymers for drug delivery has been ongoing since they were first used as bioresorbable surgical devices in the 1980s. For tissue engineering and drug delivery, biodegradable polymer poly-lactic-co-glycolic acid (PLGA) has shown enormous promise among all biomaterials. PLGA are a family of FDA-approved biodegradable polymers that are physically strong and highly biocompatible and have been extensively studied as delivery vehicles of drugs, proteins, and macromolecules such as DNA and RNA. PLGA has a wide range of erosion times and mechanical properties that can be modified. Many innovative platforms have been widely studied and created for the development of methods for the controlled delivery of PLGA. In this paper, the various manufacturing processes and characteristics that impact their breakdown and drug release are explored in depth. Besides different PLGA-based nanoparticles, preclinical and clinical applications for different diseases and the PLGA platform types and their scale-up issues will be discussed.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
- Correspondence: ; Tel.: +966-556047523
| | - Fatima D. Alharbi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alanoud S. Alhibs
- Department of Pharmacy, King Fahad Medical City, Riyadh 11564, Saudi Arabia
| | - Nouf B. Alanazi
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bayan Y. Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa A. Saleh
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Fahad S. Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Majed A. Algarni
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Turki Almugaiteeb
- Taqnia-Research Products Development Company, Riyadh 13244, Saudi Arabia
| | | | - Rami M. Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| |
Collapse
|
26
|
He Y, Zhou M, Li S, Gong Z, Yan F, Liu H. Ultrasound Molecular Imaging of Neovascularization for Evaluation of Endometrial Receptivity Using Magnetic iRGD-Modified Lipid-Polymer Hybrid Microbubbles. Int J Nanomedicine 2022; 17:5869-5881. [PMID: 36483520 PMCID: PMC9726466 DOI: 10.2147/ijn.s359065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 06/01/2022] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Angiogenesis plays an important role in endometrial receptivity, determining the response of the endometrium to the blastocyst at the early stage of embryo implantation. During the application of assisted reproduction technologies, it is very important to evaluate the status of uterine angiogenesis before deciding on embryo implantation. Targeted microbubbles (MBs)-based ultrasound molecular imaging (UMI) can noninvasively detect the expression status of biomarkers at the molecular level, thereby being a potential diagnosis strategy for various diseases and their therapeutic evaluation. METHODS The iRGD-lipopeptide (DSPE-PEG2000-iRGD) conjugate was prepared with iRGD peptides and DSPE-PEG2000-maleimide through the Michael-type addition reaction. Then, the magnetic iRGD-modified lipid-polymer hybrid MBs (Mag-iLPMs) were prepared with the double-emulsification-solvent-evaporation method. Magnetic targeting of Mag-iLPMs was confirmed under the microscope, followed by a rectangular magnet. Next, the in vitro targeted binding of MBs to murine brain-derived endothelial cells.3 (bEnd.3) and human umbilical vein endothelial cells (HUVEC) were evaluated. The ratio of MBs binding to bEnd.3 and HUVEC at the same field was also compared. For in vivo studies, bolus injections of targeted or control MBs were randomly administrated to non-pregnant or pregnant rats on day 5. Then, the uteri were imaged using a VisualSonics Vevo 2100 ultrasound system (Fujifilm VisualSonics Inc., Ontario, Canada) equipped with a high-frequency transducer. Ultrasonic imaging signals were acquired from Mag-iLPMs, and compared with Mag-LPMs, iLPMs, and LPMs. RESULTS The Mag-iLPMs showed excellent performance in ultrasound contrast imaging and binding affinity to target cells. Using the magnetic field, 10.5- and 12.47-fold higher binding efficiency to bEnd.3 and HUVEC were achieved compared to non-magnetic iLPMs, respectively. Significantly enhanced UMI signals were also observed in the uteri of rats intravenously injected pregnant rats (6.58-fold higher than rats injected with iLPMs). CONCLUSION We provided a powerful ultrasonic molecular functional imaging tool for uterine angiogenesis evaluation before embryonic implantation.
Collapse
Affiliation(s)
- Yanni He
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| | - Meijun Zhou
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| | - Sushu Li
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| | - Zheli Gong
- Department of Ultrasound, The People’s Hospital of Hunan Province, Changsha, 410061, People’s Republic of China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People’s Republic of China
| | - Hongmei Liu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People’s Republic of China
| |
Collapse
|
27
|
Chapla R, Huynh KT, Schutt CE. Microbubble–Nanoparticle Complexes for Ultrasound-Enhanced Cargo Delivery. Pharmaceutics 2022; 14:pharmaceutics14112396. [PMID: 36365214 PMCID: PMC9698658 DOI: 10.3390/pharmaceutics14112396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble–nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid–polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin–avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble–nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.
Collapse
Affiliation(s)
- Rachel Chapla
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katherine T. Huynh
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
28
|
Wang J, Li Z, Pan M, Fiaz M, Hao Y, Yan Y, Sun L, Yan F. Ultrasound-mediated blood-brain barrier opening: An effective drug delivery system for theranostics of brain diseases. Adv Drug Deliv Rev 2022; 190:114539. [PMID: 36116720 DOI: 10.1016/j.addr.2022.114539] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Blood-brain barrier (BBB) remains a significant obstacle to drug therapy for brain diseases. Focused ultrasound (FUS) combined with microbubbles (MBs) can locally and transiently open the BBB, providing a potential strategy for drug delivery across the BBB into the brain. Nowadays, taking advantage of this technology, many therapeutic agents, such as antibodies, growth factors, and nanomedicine formulations, are intensively investigated across the BBB into specific brain regions for the treatment of various brain diseases. Several preliminary clinical trials also have demonstrated its safety and good tolerance in patients. This review gives an overview of the basic mechanisms, ultrasound contrast agents, evaluation or monitoring methods, and medical applications of FUS-mediated BBB opening in glioblastoma, Alzheimer's disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Jieqiong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 201206, China
| | - Zhenzhou Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, China
| | - Min Pan
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Muhammad Fiaz
- Department of Radiology, Azra Naheed Medical College, Lahore, Pakistan
| | - Yongsheng Hao
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yiran Yan
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
29
|
Zhao P, Zhao S, Zhang J, Lai M, Sun L, Yan F. Molecular Imaging of Steroid-Induced Osteonecrosis of the Femoral Head through iRGD-Targeted Microbubbles. Pharmaceutics 2022; 14:pharmaceutics14091898. [PMID: 36145646 PMCID: PMC9505504 DOI: 10.3390/pharmaceutics14091898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/15/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a disease that is commonly seen in the clinic, but its detection rate remains limited, especially at the early stage. We developed an ultrasound molecular imaging (UMI) approach for early diagnosis of ONFH by detecting the expression of integrin αvβ3 during the pathological changes in steroid-induced osteonecrosis of the femoral head (SIONFH) in rat models. The integrin αvβ3-targeted PLGA or lipid microbubbles modified with iRGD peptides were fabricated and characterized. Their adhesion efficiency to mouse brain microvascular endothelial cells in vitro was examined, and the better LIPOiRGD was used for further in vivo molecular imaging of SIONFH rats at 1, 3 and 5 weeks; revealing significantly higher UMI signals could be observed in the 3-week and 5-week SIONFH rats but not in the 1-week SIONFH rats in comparison with the non-targeted microbubbles (32.75 ± 0.95 vs. 0.17 ± 0.09 for 5 weeks, p < 0.05; 5.60 ± 1.31 dB vs. 0.94 ± 0.81 dB for 3 weeks, p < 0.01; 1.13 ± 0.13 dB vs. 0.73 ± 0.31 dB for 1 week, p > 0.05). These results were consistent with magnetic resonance imaging data and confirmed by immunofluorescence staining experiments. In conclusion, our study provides an alternative UMI approach to the early evaluation of ONFH.
Collapse
Affiliation(s)
- Ping Zhao
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Shuai Zhao
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
- Department of Ultrasound, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou 234000, China
| | - Jiaqi Zhang
- Department of Ultrasound, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Manlin Lai
- Department of Ultrasound, The Second People’s Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen 518061, China
| | - Litao Sun
- Cancer Center, Department of Ultrasound Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou 310014, China
- Correspondence: (L.S.); (F.Y.); Tel.: +86-755-8639-2284 (F.Y.); Fax: +86-755-9638-2299 (F.Y.)
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence: (L.S.); (F.Y.); Tel.: +86-755-8639-2284 (F.Y.); Fax: +86-755-9638-2299 (F.Y.)
| |
Collapse
|
30
|
Khalili L, Dehghan G, Sheibani N, Khataee A. Smart active-targeting of lipid-polymer hybrid nanoparticles for therapeutic applications: Recent advances and challenges. Int J Biol Macromol 2022; 213:166-194. [PMID: 35644315 DOI: 10.1016/j.ijbiomac.2022.05.156] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/24/2022]
Abstract
The advances in producing multifunctional lipid-polymer hybrid nanoparticles (LPHNs) by combining the biomimetic behavior of liposomes and architectural advantages of polymers have provided great opportunities for selective and efficient therapeutics delivery. The constructed LPHNs exhibit different therapeutic efficacies for special uses based on characteristics of different excipients. However, the high mechanical/structural stability of hybrid nano-systems could be viewed as both a negative property and a positive feature, where the concomitant release of drug molecules in a controllable manner is required. In addition, difficulties in scaling up the LPHNs production, due to involvement of several criteria, limit their application for biomedical fields, especially in monitoring, bioimaging, and drug delivery. To address these challenges bio-modifications have exhibited enormous potential to prepare reproducible LPHNs for site-specific therapeutics delivery, diagnostic and preventative applications. The ever-growing surface bio-functionality has provided continuous vitality to this biotechnology and has also posed desirable biosafety to nanoparticles (NPs). As a proof-of-concept, this manuscript provides a crucial review of coated lipid and polymer NPs displaying excellent surface functionality and architectural advantages. We also provide a description of structural classifications and production methodologies, as well as the biomedical possibilities and translational obstacles in the development of surface modified nanocarrier technology.
Collapse
Affiliation(s)
- Leila Khalili
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, 51666-16471 Tabriz, Iran.
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Cell and Regenerative Biology, and Biomedical Engineering, University of Wisconsin School of Medicine, Madison, WI, USA
| | - Alireza Khataee
- Research Laboratory of Advanced Water and Wastewater Treatment Processes, Department of Applied Chemistry, Faculty of Chemistry, University of Tabriz, 51666-16471 Tabriz, Iran; Department of Materials Science and Nanotechnology Engineering, Faculty of Engineering, Near East University, 99138 Nicosia, Mersin 10, Turkey.
| |
Collapse
|
31
|
Li N, Dong L, Shen Y, Wang Y, Chang L, Wu H, Chang Y, Li M, Li D, Li Z, He M, Li C, Wei Y, Xie H, Wang F. Therapeutic Effect of Ultrasound Combined With Porous Lipid Clioquinol/PLGA Microbubbles on Ferroptosis in HL-1 Cardiac Cell Induced by Isoproterenol Attack. Front Pharmacol 2022; 13:918292. [PMID: 35935822 PMCID: PMC9354950 DOI: 10.3389/fphar.2022.918292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, studies have shown a close relationship between cardiomyocyte death and ferroptosis. Clioquinol (CQ) can inhibit ferroptosis. Porous lipid-poly (lactic-co-glycolic acid) (PLGA) microbubbles (MBs) were prepared by double emulsification (W1/O/W2) using 1,2-dioctadecanoyl-sn-glycero-3-phophocholine and PLGA as raw materials. Porous lipid-PLGA MBs were used as carriers to prepare CQ/PLGA MBs containing CQ. CQ/PLGA had the advantages of high drug loading, good biocompatibility, and sustained release. Our results showed that CQ/PLGA improved the effect of CQ and reduced its cytotoxicity. Under low-frequency ultrasound with certain parameters, CQ/PLGA showed steady-state cavitation, which increased the membrane permeability of mouse cardiomyocyte HL-1 to a certain extent and further prevented the process of ferroptosis in mouse cardiomyocyte HL-1.
Collapse
Affiliation(s)
- Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lei Dong
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yuanyuan Shen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
- *Correspondence: Haiqin Xie, ; Yuanyuan Shen, ; Feng Wang,
| | - Yongling Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Liansheng Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hongwei Wu
- Department of Chemistry, Xinxiang Medical University, Xinxiang, China
| | - Yuqiao Chang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Menghao Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Dan Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhaoyi Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Mei He
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Cheng Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yao Wei
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Haiqin Xie
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Haiqin Xie, ; Yuanyuan Shen, ; Feng Wang,
| | - Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Haiqin Xie, ; Yuanyuan Shen, ; Feng Wang,
| |
Collapse
|
32
|
Li Z, Lai M, Zhao S, Zhou Y, Luo J, Hao Y, Xie L, Wang Y, Yan F. Ultrasound Molecular Imaging for Multiple Biomarkers by Serial Collapse of Targeting Microbubbles with Distinct Acoustic Pressures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2108040. [PMID: 35499188 DOI: 10.1002/smll.202108040] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ultrasound molecular imaging (UMI) has shown promise for assessing the expression levels of biomarkers for the early detection of various diseases. However, it remains difficult to simultaneously image multiple biomarkers in a single systemic administration, which is important for the accurate diagnosis of diseases and for understanding the dynamic intermolecular mechanisms that drive their malignant progression. The authors develop an ultrasound molecular imaging method by serial collapse of targeting microbubbles with distinct acoustic pressures for the simultaneous detection of two biomarkers. To test this, αv β3 -targeting lipid microbubbles (L-MBα ) and VEGFR2-targeting lipid-PLGA microbubbles (LP-MBv ) are fabricated and simultaneously injected into tumor-bearing mice at 7 and 14 days, followed by the low-intensity acoustic collapse of L-MBα and high-intensity acoustic collapse of LP-MBv . The UMI signals of L-MBα and LP-MBv are obtained by subtracting the first post-burst signals from the first pre-burst signals, and subtracting the second post-burst signals from the first post-burst signals, respectively. Interestingly, the signal intensities from UMI agree with the immunohistochemical staining results for αv β3 and VEGFR2. Importantly, they find a better fit for the invasive behavior of MDA-MB-231 breast tumors by analyzing the ratio of αv β3 integrin to VEGFR2, but not the single αv β3 or VEGFR2 levels.
Collapse
Affiliation(s)
- Zhenzhou Li
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518061, P. R. China
- Shenzhen University Health Science Center, Shenzhen, 518000, P. R. China
| | - Manlin Lai
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518061, P. R. China
- Shenzhen University Health Science Center, Shenzhen, 518000, P. R. China
| | - Shuai Zhao
- Department of Ultrasound, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, 234000, P. R. China
| | - Yi Zhou
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518061, P. R. China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, P. R. China
| | - Jingna Luo
- Department of Ultrasound, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518061, P. R. China
- Shenzhen University Health Science Center, Shenzhen, 518000, P. R. China
| | - Yongsheng Hao
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Liting Xie
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yaru Wang
- Department of Radiology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, 234000, P. R. China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| |
Collapse
|
33
|
Wu H, Zhou H, Zhang W, Jin P, Shi Q, Miao Z, Wang H, Zha Z. Three birds with one stone: co-encapsulation of diclofenac and DL-menthol for realizing enhanced energy deposition, glycolysis inhibition and anti-inflammation in HIFU surgery. J Nanobiotechnology 2022; 20:215. [PMID: 35524259 PMCID: PMC9074192 DOI: 10.1186/s12951-022-01437-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/25/2022] [Indexed: 01/12/2023] Open
Abstract
Despite attracting increasing attention in clinic, non-invasive high-intensity focused ultrasound (HIFU) surgery still commonly suffers from tumor recurrence and even matastasis due to the generation of thermo-resistance in non-apoptotic tumor cells and adverse therapy-induced inflammation with enhanced secretion of growth factors in irradiated region. In this work, inspired by the intrinsic property that the expression of thermo-resistant heat shock proteins (HSPs) is highly dependent with adenosine triphosphate (ATP), dual-functionalized diclofenac (DC) with anti-inflammation and glycolysis-inhibition abilities was successfully co-encapsulated with phase-change dl-menthol (DLM) in poly(lactic-co-glycolic acid) nanoparticles (DC/DLM@PLGA NPs) to realize improved HIFU surgery without causing adverse inflammation. Both in vitro and in vivo studies demonstrated the great potential of DC/DLM@PLGA NPs for serving as an efficient synergistic agent for HIFU surgery, which can not only amplify HIFU ablation efficacy through DLM vaporization-induced energy deposition but also simultaneously sensitize tumor cells to hyperthermia by glycolysis inhibition as well as diminished inflammation. Thus, our study provides an efficient strategy for simultaneously improving the curative efficiency and diminishing the harmful inflammatory responses of clinical HIFU surgery.
Collapse
Affiliation(s)
- Haitao Wu
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Hu Zhou
- Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, Guangdong, China
| | - Wenjie Zhang
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Ping Jin
- Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, Guangdong, China.
| | - Qianqian Shi
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Zhaohua Miao
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China
| | - Hua Wang
- Department of Oncology, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, School of Instrument Science and Opto-Electronics Engineering, Hefei University of Technology, Anhui, 230009, Hefei, China.
| |
Collapse
|
34
|
Zhang L, Lin Z, Zeng L, Zhang F, Sun L, Sun S, Wang P, Xu M, Zhang J, Liang X, Ge H. Ultrasound-induced biophysical effects in controlled drug delivery. SCIENCE CHINA. LIFE SCIENCES 2022; 65:896-908. [PMID: 34453275 DOI: 10.1007/s11427-021-1971-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/27/2021] [Indexed: 12/30/2022]
Abstract
Ultrasound is widely used in biomedical engineering and has applications in conventional diagnosis and drug delivery. Recent advances in ultrasound-induced drug delivery have been summarized previously in several reviews that have primarily focused on the fabrication of drug delivery carriers. This review discusses the mechanisms underlying ultrasound-induced drug delivery and factors affecting delivery efficiency, including the characteristics of drug delivery carriers and ultrasound parameters. Firstly, biophysical effects induced by ultrasound, namely thermal effects, cavitation effects, and acoustic radiation forces, are illustrated. Secondly, the use of these biophysical effects to enhance drug delivery by affecting drug carriers and corresponding tissues is clarified in detail. Thirdly, recent advances in ultrasound-triggered drug delivery are detailed. Safety issues and optimization strategies to improve therapeutic outcomes and reduce side effects are summarized. Finally, current progress and future directions are discussed.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Zhuohua Lin
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Lan Zeng
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Fan Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Lihong Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Ping Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Menghong Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Jinxia Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Huiyu Ge
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
35
|
Engineering of ultrasound contracts agents-focused cabazitaxel-loaded microbubbles nanomaterials induces cell proliferation and enhancing apoptosis in cancer cells. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-022-02376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Highlights in ultrasound-targeted microbubble destruction-mediated gene/drug delivery strategy for treatment of malignancies. Int J Pharm 2021; 613:121412. [PMID: 34942327 DOI: 10.1016/j.ijpharm.2021.121412] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/05/2023]
Abstract
Ultrasound is one of the safest and most advanced medical imaging technologies that is widely used in clinical practice. Ultrasound microbubbles, traditionally used for contrast-enhanced imaging, are increasingly applied in Ultrasound-targeted Microbubble Destruction (UTMD) technology which enhances tissue and cell membrane permeability through cavitation and sonoporation, to result in a promising therapeutic gene/drug delivery strategy. Here, we review recent developments in the application of UTMD-mediated gene and drug delivery in the diagnosis and treatment of tumors, including the concept, mechanism of action, clinical application status, and advantages of UTMD. Furthermore, the future perspectives that should be paid more attention to in this field are prospected.
Collapse
|
37
|
Yan Y, Chen Y, Liu Z, Cai F, Niu W, Song L, Liang H, Su Z, Yu B, Yan F. Brain Delivery of Curcumin Through Low-Intensity Ultrasound-Induced Blood-Brain Barrier Opening via Lipid-PLGA Nanobubbles. Int J Nanomedicine 2021; 16:7433-7447. [PMID: 34764649 PMCID: PMC8575349 DOI: 10.2147/ijn.s327737] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background Parkinson's disease (PD) is a progressive neurodegenerative disorder. Owing to the presence of blood-brain barrier (BBB), conventional pharmaceutical agents are difficult to the diseased nuclei and exert their action to inhibit or delay the progress of PD. Recent literatures have demonstrated that curcumin shows the great potential to treat PD. However, its applications are still difficult in vivo due to its poor druggability and low bioavailability through the BBB. Methods Melt-crystallization methods were used to improve the solubility of curcumin, and curcumin-loaded lipid-PLGA nanobubbles (Cur-NBs) were fabricated through encapsulating the curcumin into the cavity of lipid-PLGA nanobubbles. The bubble size, zeta potentials, ultrasound imaging capability and drug encapsulation efficiency of the Cur-NBs were characterized by a series of analytical methods. Low-intensity focused ultrasound (LIFU) combined with Cur-NB was used to open the BBB to facilitate curcumin delivery into the deep brain of PD mice, followed by behavioral evaluation for the treatment efficacy. Results The solubility of curcumin was improved by melt-crystallization methods, with 2627-fold higher than pure curcumin. The resulting Cur-NBs have a nanoscale size about 400 nm and show excellent contrast imaging performance. Curcumin drugs encapsulated into Cur-NBs could be effectively released when Cur-NBs were irradiated by LIFU at the optimized acoustic pressure, achieving 30% cumulative release rate within 6 h. Importantly, Cur-NBs combined with LIFU can open the BBB and locally deliver the curcumin into the deep-seated brain nuclei, significantly enhancing efficacy of curcumin in the Parkinson C57BL/6J mice model in comparison with only Cur-NBs and LIFU groups. Conclusion In this work, we greatly improved the solubility of curcumin and developed Cur-NBs for brain delivery of curcumin against PD through combining with LIFU-mediating BBB. Cur-NBs provide a platform for these potential drugs which are difficult to cross the BBB to treat PD disease or other central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Yiran Yan
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Yan Chen
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhongxun Liu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Feiyan Cai
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, People's Republic of China
| | - Wanting Niu
- VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Liming Song
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Haifeng Liang
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Zhiwen Su
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Bo Yu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, People's Republic of China
| |
Collapse
|
38
|
Li N, Duan S, Wang Y, Zhang L, Chen Y, Zhang J, Liu R, Li Y, Liu L, Ren S, Zhang Y, Guo Y, Ji Z, Zhang L. Preparation and evaluation of ultrasound-mediated dual-targeted theragnostic systems utilising phase-changeable polymeric nanodroplets on the integrin α ν β 3 overexpressed breast cancer. Clin Transl Med 2021; 11:e607. [PMID: 34709751 PMCID: PMC8516363 DOI: 10.1002/ctm2.607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/15/2021] [Accepted: 09/24/2021] [Indexed: 12/04/2022] Open
Affiliation(s)
- Na Li
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China.,Henan International Joint Laboratory for Gynecological Oncology and Nanomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Shaobo Duan
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yiwei Wang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Linlin Zhang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yongqing Chen
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Juan Zhang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Ruiqing Liu
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yaqiong Li
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Luwen Liu
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Shanshan Ren
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Ye Zhang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yuqi Guo
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China.,Henan International Joint Laboratory for Gynecological Oncology and Nanomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Zhenyu Ji
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Lianzhong Zhang
- Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
39
|
Applications of Ultrasound-Mediated Drug Delivery and Gene Therapy. Int J Mol Sci 2021; 22:ijms222111491. [PMID: 34768922 PMCID: PMC8583720 DOI: 10.3390/ijms222111491] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Gene therapy has continuously evolved throughout the years since its first proposal to develop more specific and effective transfection, capable of treating a myriad of health conditions. Viral vectors are some of the most common and most efficient vehicles for gene transfer. However, the safe and effective delivery of gene therapy remains a major obstacle. Ultrasound contrast agents in the form of microbubbles have provided a unique solution to fulfill the need to shield the vectors from the host immune system and the need for site specific targeted therapy. Since the discovery of the biophysical and biological effects of microbubble sonification, multiple developments have been made to enhance its applicability in targeted drug delivery. The concurrent development of viral vectors and recent research on dual vector strategies have shown promising results. This review will explore the mechanisms and recent advancements in the knowledge of ultrasound-mediated microbubbles in targeting gene and drug therapy.
Collapse
|
40
|
Li G, Li J, Hou Y, Xie S, Xu J, Yang M, Li D, Du Y. Levofloxacin-Loaded Nanosonosensitizer as a Highly Efficient Therapy for Bacillus Calmette-Guérin Infections Based on Bacteria-Specific Labeling and Sonotheranostic Strategy. Int J Nanomedicine 2021; 16:6553-6573. [PMID: 34602818 PMCID: PMC8478796 DOI: 10.2147/ijn.s321631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose The rapid emergence of multidrug-resistant Mycobacterium tuberculosis (MTB) poses a significant challenge to the treatment of tuberculosis (TB). Sonodynamic antibacterial chemotherapy (SACT) combined with sonosensitizer-loaded nanoparticles with targeted therapeutic function is highly expected to eliminate bacteria without fear of drug resistance. This study aimed to investigate the antibacterial effect and underlying mechanism of levofloxacin-loaded nanosonosensitizer with targeted therapeutic function against Bacillus Calmette-Guérin bacteria (BCG, an MTB model). Methods This study developed levofloxacin-loaded PLGA-PEG (poly lactide-co-glycolide-polyethylene glycol) nanoparticles with BM2 aptamer conjugation on its surface using the crosslinking agents EDC and NHS (BM2-LVFX-NPs). The average diameter, zeta potential, morphology, drug-loading properties, and drug release efficiency of the BM2-LVFX-NPs were investigated. In addition, the targeting and toxicity of BM2-LVFX-NPs in the subcutaneous BCG infection model were evaluated. The biosafety, reactive oxygen species (ROS) production, cellular phagocytic effect, and antibacterial effect of BM2-LVFX-NPs in the presence of ultrasound stimulations (42 kHz, 0.67 W/cm2, 5 min) were also systematically evaluated. Results BM2-LVFX-NPs not only specifically recognized BCG bacteria in vitro but also gathered accurately in the lesion tissues. Drugs loaded in BM2-LVFX-NPs with the ultrasound-responsive feature were effectively released compared to the natural state. In addition, BM2-LVFX-NPs exhibited significant SACT efficiency with higher ROS production levels than others, resulting in the effective elimination of bacteria in vitro. Meanwhile, in vivo experiments, compared with other options, BM2-LVFX-NPs also exhibited an excellent therapeutic effect in a rat model with BCG infection after exposure to ultrasound. Conclusion Our work demonstrated that a nanosonosensitizer formulation with LVFX could efficiently translocate therapeutic drugs into the cell and improve the bactericidal effects under ultrasound, which could be a promising strategy for targeted therapy for MTB infections with high biosafety.
Collapse
Affiliation(s)
- Gangjing Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jianhu Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yuru Hou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Shuang Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jieru Xu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Min Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dairong Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| |
Collapse
|
41
|
Wang F, Dong L, Wei X, Wang Y, Chang L, Wu H, Liu S, Chang Y, Yin Y, Luo X, Jia X, Yan F, Li N. Effect of Gambogic Acid-Loaded Porous-Lipid/PLGA Microbubbles in Combination With Ultrasound-Triggered Microbubble Destruction on Human Glioma. Front Bioeng Biotechnol 2021; 9:711787. [PMID: 34604184 PMCID: PMC8479098 DOI: 10.3389/fbioe.2021.711787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
Gambogic acid (GA) is a highly effective antitumor agent, and it is used for the treatment of a wide range of cancers. It is challenging to deliver drugs to the central nervous system due to the inability of GA to cross the blood-brain barrier (BBB). Studies have shown that ultrasound-targeted microbubble destruction can be used for transient and reversible BBB disruption, significantly facilitating intracerebral drug delivery. We first prepared GA-loaded porous-lipid microbubbles (GA porous-lipid/PLGA MBs), and an in vitro BBB model was established. The cell viability was detected by CCK-8 assay and flow cytometry. The results indicate that U251 human glioma cells were killed by focused ultrasound (FUS) combined with GA/PLGA microbubbles. FUS combined with GA/PLGA microbubbles was capable of locally and transiently enhancing the permeability of BBB under certain conditions. This conformational change allows the release of GA to extracellular space. This study provides novel targets for the treatment of glioma.
Collapse
Affiliation(s)
- Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lei Dong
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xixi Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yongling Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Liansheng Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Hongwei Wu
- Department of Chemistry, Xinxiang Medical University, Xinxiang, China
| | - Shuyuan Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, China
| | - Yuqiao Chang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaoqiu Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Xiaojian Jia
- Shenzhen Kangning Hospital and Shenzhen Mental Health Center, Shenzhen, China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
42
|
Xu L, Chen Y, Jin Q, Wu Y, Deng C, Zhong Y, Lin L, Chen L, Fu W, Yi L, Sun Z, Qin X, Li Y, Yang Y, Xie M. Biomimetic PLGA Microbubbles Coated with Platelet Membranes for Early Detection of Myocardial Ischaemia-Reperfusion Injury. Mol Pharm 2021; 18:2974-2985. [PMID: 34197128 DOI: 10.1021/acs.molpharmaceut.1c00145] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Early diagnosis of myocardial ischaemia-reperfusion (MI/R) injury is important for protecting the myocardium and improving patient prognoses. Fortunately, the platelet membrane possesses the ability to target the region of MI/R injury. Therefore, we hypothesized that platelet membrane-coated particles (PMPs) could be used to detect early MI/R injury by ultrasound imaging. We designed PMPs with a porous polylactic-co-glycolic acid (PLGA) core coated with a platelet membrane shell. Red blood cell membrane-coated particles (RMPs) were fabricated as controls. Transmission electron microscopy (TEM) and fluorescence microscopy were applied to confirm the membrane coatings of the PMPs and RMPs. In vitro imaging of the PMPs and RMPs was verified. Moreover, binding experiments were designed to examine the targeting ability of the PMPs. Finally, we assessed the signal intensity of the adherent PMPs in the risk area and remote area by ultrasound imaging based on an MI/R rat model. The platelet membrane equipped the PMPs with an accurate targeting ability. Compared with RMPs, PMPs showed significantly more adhesion to human umbilical vein endothelial cells and collagen IV in vitro. Both PMPs and RMPs exhibited good enhancement ability in vitro and in vivo. Furthermore, the signal intensity of PMPs in the risk area was significantly higher than that in remote areas. These results were further validated by an immunofluorescence assay and ex vivo fluorescence imaging. In summary, ultrasound imaging with PMPs can detect early MI/R injury in a noninvasive manner.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ya Wu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yi Zhong
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ling Lin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ling Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wenpei Fu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Luyang Yi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Zhenxing Sun
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiaojuan Qin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yuman Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
43
|
Ren WW, Xu SH, Sun LP, Zhang K. Ultrasound-Based Drug Delivery System. Curr Med Chem 2021; 29:1342-1351. [PMID: 34139971 DOI: 10.2174/0929867328666210617103905] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/25/2021] [Accepted: 05/01/2021] [Indexed: 12/07/2022]
Abstract
Cancer still represents a leading threat to human health worldwide. The effective usage of anti-cancer drugs can reduce patients' clinical symptoms and extend the life span. Current anti-cancer strategies include chemotherapy, traditional Chinese medicine, biopharmaceuticals, and the latest targeted therapy. However, due to the complexity and heterogeneity of tumors, serious side effects may result from the direct use of anti-cancer drugs. Besides, the current therapeutic strategies failed to effectively alleviate metastasized tumors. Recently, an ultrasound-mediated nano-drug delivery system has become an increasingly important treatment strategy. Due to its ability to enhance efficacy and reduce toxic side effects, it has become a research hotspot in the field of biomedicine. In this review, we introduced the latest research progress of the ultrasound-responsive nano-drug delivery systems and the possible mechanisms of ultrasound acting on the carrier to change the structure or conformation as well as to realize the controlled release. In addition, the progress in ultrasound responsive nano-drug delivery systems will also be briefly summarized.
Collapse
Affiliation(s)
- Wei-Wei Ren
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, Shanghai 200072, China
| | - Shi-Hao Xu
- Department of Ultrasound, The first affiliated hospital of Wenzhou Medical University, WenZhou, 325000, Zhejiang Province, China
| | - Li-Ping Sun
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, Shanghai 200072, China
| | - Kun Zhang
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
44
|
Ullah M, Kodam SP, Mu Q, Akbar A. Microbubbles versus Extracellular Vesicles as Therapeutic Cargo for Targeting Drug Delivery. ACS NANO 2021; 15:3612-3620. [PMID: 33666429 DOI: 10.1021/acsnano.0c10689] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Extracellular vesicles (EVs) and microbubbles are nanoparticles in drug-delivery systems that are both considered important for clinical translation. Current research has found that both microbubbles and EVs have the potential to be utilized as drug-delivery agents for therapeutic targets in various diseases. In combination with EVs, microbubbles are capable of delivering chemotherapeutic drugs to tumor sites and neighboring sites of damaged tissues. However, there are no standards to evaluate or to compare the benefits of EVs (natural carrier) versus microbubbles (synthetic carrier) as drug carriers. Both drug carriers are being investigated for release patterns and for pharmacokinetics; however, few researchers have focused on their targeted delivery or efficacy. In this Perspective, we compare EVs and microbubbles for a better understanding of their utility in terms of delivering drugs to their site of action and future clinical translation.
Collapse
Affiliation(s)
- Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, California 94304, United States
- Department of Molecular Medicine, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Sai Priyanka Kodam
- Department of Molecular Medicine, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Qian Mu
- Department of Molecular Medicine, School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Asma Akbar
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, California 94304, United States
| |
Collapse
|
45
|
Deng Z, Wang J, Xiao Y, Li F, Niu L, Liu X, Meng L, Zheng H. Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity. Am J Cancer Res 2021; 11:4351-4362. [PMID: 33754065 PMCID: PMC7977450 DOI: 10.7150/thno.52436] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Extracellular vesicles, including exosomes, are secreted by a variety of cell types in the central nervous system. Exosomes play a role in removing intracellular materials from the endosomal system. Alzheimer's disease (AD) is caused by an overproduction or reduced amyloid-beta (Aβ) peptide clearance. Increased Aβ levels in the brain may impair the exosome-mediated Aβ clearance pathway. Therapeutic ultrasound stimulation demonstrated its potential for promoting Aβ degradation efficiency in clinical trials. However, the underlying mechanism of ultrasound stimulation is still unclear. Methods: In this study, astrocytes, the most abundant glial cells in the brain, were used for exosome production. Post insonation, exosomes from ultrasound-stimulated HA cells (US-HA-Exo) were collected, nanoparticle tracking analysis and protein analysis were used to measure and characterize exosomes. Neuroprotective effect of US-HA-Exo in oligomeric Aβ42 toxicated SH-SY5Y cells was tested. Cellular uptake and distribution of exosomes were observed by flow cytometry and confocal laser scanning microscopy. Focused ultrasound (FUS) with microbubbles was employed for blood-brain-barrier opening to achieve brain-targeted exosome delivery. After US-HA-Exo/FUS treatment, amyloid-β plaque in APP/PS1 mice were evaluated by Aβ immunostaining and thioflavin-S staining. Results: We showed that ultrasound resulted in an almost 5-fold increase in the exosome release from human astrocytes. Exosomes were rapidly internalized in SH-SY5Y cells, and colocalized with FITC-Aβ42, causing a decreased uptake of FITC-Aβ42. CCk-8 test results showed that US-HA-Exo could mitigate Aβ toxicity to neurons in vitro. The therapeutic potential of US-HA-Exo/FUS delivery was demonstrated by a decrease in thioflavin-S-positive amyloid plaques and Aβ immuno-staining, a therapeutic target for AD in APP/PS1 transgenic mice. The iTRAQ-based proteomic quantification was performed to gain mechanistic insight into the ultrasound effect on astrocyte-derived exosomes and their ability to alleviate Aβ neurotoxicity. Conclusion: Our results imply that US-HA-Exo have the potential to provide neuroprotective effects to reverse oligomeric amyloid-β-induced cytotoxicity in vitro and, when combined with FUS-induced BBB opening, enable the clearance of amyloid-β plaques in vivo.
Collapse
|
46
|
Liu Y, Ma Y, Peng X, Wang L, Li H, Cheng W, Zheng X. Cetuximab-conjugated perfluorohexane/gold nanoparticles for low intensity focused ultrasound diagnosis ablation of thyroid cancer treatment. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2021; 21:856-866. [PMID: 33551680 PMCID: PMC7850351 DOI: 10.1080/14686996.2020.1855064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
We report the formulation of nanoassemblies (NAs) comprising C225 conjugates Au-PFH-NAs (C-Au-PFH-NAs) for low-intensity focused ultrasound diagnosis ablation of thyroid cancer. C-Au-PFH-NAs showed excellent stability in water, phosphate-buffered saline (PBS), and 20% rat serum. Transmission electron microscopy (TEM) images also revealed the effective construction of C-Au-PFH-NAs as common spherical assemblies. The incubation of C625 thyroid carcinoma with C-Au-PFH-NAs triggers apoptosis, as confirmed by flow cytometry analysis. The C-Au-PFH-NAs exhibited antitumour efficacy in human thyroid carcinoma xenografts, where histopathological results further confirmed these outcomes. Furthermore, we were able to use low-intensity focused ultrasound diagnosis imaging (LIFUS) to examine the efficiency of C-Au-PFH-NAs in thyroid carcinoma in vivo. These findings clearly show that the use of LIFUS agents with high-performance imaging in different therapeutic settings will have extensive potential for future biomedical applications.
Collapse
Affiliation(s)
- Ying Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
| | - Yue Ma
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
| | - Xiaoshan Peng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
| | - Lingling Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
| | - Haixia Li
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
| | - Xiulan Zheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, P.R. China
- CONTACT Xiulan Zheng No.150, Haping Road, Harbin150081, P.R. China
| |
Collapse
|
47
|
Lv W, Xia H, Zou L, Zhao M, Yang T, Jiang J, Chen Z, Liu S, Zhao Q. Yolk-shell structured Au nanorods@mesoporous silica for gas bubble driven drug release upon near-infrared light irradiation. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 32:102326. [PMID: 33166666 DOI: 10.1016/j.nano.2020.102326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 02/02/2023]
Abstract
Drug release systems co-encapusulated with ammonium bicarbonate (ABC) could facilitate drug release upon acidic or thermal stimulations to improve therapeutic effect. However, it is not easy to control drug release rate, owing to relative stable temperature and acidic condition in living body. Besides, the additional loaded ABC reduces drug loading capacity. Herein, a near-infrared light triggered rapid drug release system with high loading capacity was developed by loading ABC and doxorubicin into yolk-shell structured Au nanorods@mesoporous silica. Gas bubbles were generated from the thermolysis of ABC utilizing photothermal effect of Au nanorods to extrude drug molecules. The mesoporous silica shell was finally destroyed along with growing bubbles, resulting in burst drug release. The photothermal therapeutic effect of Au nanorods also contributed in tumor treatment. The excellent therapeutic effect was demonstrated in cancer cells and tumor-bearing mice, which provides a new reference to achieve controllable rapid drug release in cancer medicine.
Collapse
Affiliation(s)
- Wen Lv
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Huiting Xia
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Liang Zou
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Menglong Zhao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Tianshe Yang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Jianting Jiang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Zejing Chen
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China
| | - Shujuan Liu
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China.
| | - Qiang Zhao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications (NUPT), Nanjing, China.
| |
Collapse
|
48
|
Zhu Y, Sun Y, Liu W, Guan W, Liu H, Duan Y, Chen Y. Magnetic polymeric nanobubbles with optimized core size for MRI/ultrasound bimodal molecular imaging of prostate cancer. Nanomedicine (Lond) 2020; 15:2901-2916. [PMID: 33300812 DOI: 10.2217/nnm-2020-0188] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim: To design MRI/ultrasound (US) dual modality imaging probes with optimized size for prostate cancer imaging by targeting prostate-specific membrane antigen (PSMA). Materials & methods: The PSMA-targeting polypeptide-nanobubbles (PP-NBs) with core size of 400 and 700 nm were fabricated and evaluated. Results: With excellent physical property and specificity, PP-NBs of both core size could image PSMA expression in prostate cancer xenografts. Particularly, 400 nm PP-NBs generated higher PSMA-specific MRI/US dual modality contrast enhancement than 700 nm PP-NBs in correlation with histopathologic findings. Conclusion: Benefit from the smaller core size, 400 nm PP-NBs had higher permeability and specificity than 700 nm PP-NBs, hence producing better PSMA-specific MRI/US dual modality imaging.
Collapse
Affiliation(s)
- Yunkai Zhu
- Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Weiyong Liu
- Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Wenbin Guan
- Department of Pathology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, PR China
| | - Yaqing Chen
- Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| |
Collapse
|
49
|
Kumar SU, Telichko AV, Wang H, Hyun D, Johnson EG, Kent MS, Rebhun RB, Dahl JJ, Culp WTN, Paulmurugan R. Acoustically Driven Microbubbles Enable Targeted Delivery of microRNA-Loaded Nanoparticles to Spontaneous Hepatocellular Neoplasia in Canines. ADVANCED THERAPEUTICS 2020; 3:2000120. [PMID: 33415184 PMCID: PMC7784952 DOI: 10.1002/adtp.202000120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 01/16/2023]
Abstract
Spatially localized microbubble cavitation by ultrasound offers an effective means of altering permeability of natural barriers (i.e. blood vessel and cell membrane) in favor of nanomaterials accumulation in the target site. In this study, a clinically relevant, minimally invasive ultrasound guided therapeutic approach is investigated for targeted delivery of anticancer microRNA loaded PLGA-b-PEG nanoparticles to spontaneous hepatocellular neoplasia in a canine model. Quantitative assessment of the delivered microRNAs revealed prominent and consistent increase in miRNAs levels (1.5-to 2.3-fold increase (p<0.001)) in ultrasound treated tumor regions compared to untreated control regions. Immunohistology of ultrasound treated tumor tissue presented a clear evidence for higher amount of nanoparticles extravasation from the blood vessels. A distinct pattern of cytokine expression supporting CD8+ T cells mediated "cold-to-hot" tumor transition was evident in all patients. On the outset, proposed platform can enhance delivery of miRNA-loaded nanoparticles to deep seated tumors in large animals to enhance chemotherapy.
Collapse
Affiliation(s)
- Sukumar Uday Kumar
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Arsenii V Telichko
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Huaijun Wang
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Dongwoon Hyun
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Eric G Johnson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Robert B Rebhun
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Jeremy J Dahl
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - William T N Culp
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
50
|
Mohanty A, Uthaman S, Park IK. Utilization of Polymer-Lipid Hybrid Nanoparticles for Targeted Anti-Cancer Therapy. Molecules 2020; 25:E4377. [PMID: 32977707 PMCID: PMC7582728 DOI: 10.3390/molecules25194377] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer represents one of the most dangerous diseases, with 1.8 million deaths worldwide. Despite remarkable advances in conventional therapies, these treatments are not effective to completely eradicate cancer. Nanotechnology offers potential cancer treatment based on formulations of several nanoparticles (NPs). Liposomes and polymeric nanoparticle are the most investigated and effective drug delivery systems (DDS) for cancer treatment. Liposomes represent potential DDS due to their distinct properties, including high-drug entrapment efficacy, biocompatibility, low cost, and scalability. However, their use is restricted by susceptibility to lipid peroxidation, instability, burst release of drugs, and the limited surface modification. Similarly, polymeric nanoparticles show several chemical modifications with polymers, good stability, and controlled release, but their drawbacks for biological applications include limited drug loading, polymer toxicity, and difficulties in scaling up. Therefore, polymeric nanoparticles and liposomes are combined to form polymer-lipid hybrid nanoparticles (PLHNPs), with the positive attributes of both components such as high biocompatibility and stability, improved drug payload, controlled drug release, longer circulation time, and superior in vivo efficacy. In this review, we have focused on the prominent strategies used to develop tumor targeting PLHNPs and discuss their advantages and unique properties contributing to an ideal DDS.
Collapse
Affiliation(s)
- Ayeskanta Mohanty
- Department of Biomedical Sciences, Chonnam National University Medical School, 264, Seoyang-ro, Jeollanam-do 58128, Korea;
| | - Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseoung-gu, Daejeon 34134, Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 264, Seoyang-ro, Jeollanam-do 58128, Korea;
| |
Collapse
|