1
|
Gomez-Garcia MJ, Abdelkarim M, Cramb DT, Childs SJ, Rinker KD, Labouta HI. Blood vessel wall shear stress determines regions of liposome accumulation in angiogenic vasculature. Drug Deliv Transl Res 2024; 14:3608-3620. [PMID: 39042244 DOI: 10.1007/s13346-024-01671-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Nanoparticles used for drug delivery often require intravenous administration exposing them to fluid forces within the vasculature, yet the impact of blood flow on nanoparticle delivery remains incompletely understood. Here, we utilized transgenic zebrafish embryos to investigate the relationship between the accumulation of fluorescently labeled PEGylated liposomes and various hemodynamic factors (such as flow velocity, wall shear stress (WSS), and flow pattern) across a wide range of angiogenic blood vessels. We reconstructed 3D models of vascular structures from confocal images and used computational fluid dynamics to calculate local WSS, velocities, and define flow patterns. The spatial distribution of fluorescently labeled liposomes was subsequently mapped within the same 3D space and correlated with local hemodynamic parameters. Through the integration of computational fluid dynamics and in vivo experimentation, we show that liposomes accumulated in vessel regions with WSS between 0.1-0.8 Pa, displaying an inverse linear correlation (R2 > 0.85) between time-averaged wall shear stress and liposome localization in vivo. Interestingly, flow pattern did not appear to impact liposome accumulation. Collectively, our findings suggest the potential of stealth liposomes for passive targeting of low-flow vasculature, including capillaries and intricate angiogenic vasculature resembling that of tumor vessel networks.
Collapse
Affiliation(s)
- M Juliana Gomez-Garcia
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada
- Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Mahmoud Abdelkarim
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - David T Cramb
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University, Toronto, ON, Canada
| | - Sarah J Childs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kristina D Rinker
- Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada.
| | - Hagar I Labouta
- Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto, ON, Canada.
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [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: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
3
|
Jahromi BR, Zamotin V, Code C, Netti E, Lorey MB, Alitalo K, Öörni K, Laakso A, Tulamo R, Niemelä M. Immunoliposomes for detection of rupture-prone intracranial aneurysms. Acta Neurochir (Wien) 2023; 165:3353-3360. [PMID: 37749289 PMCID: PMC10624708 DOI: 10.1007/s00701-023-05770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/15/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND It is estimated that significant (3.2%) of population carries intracranial aneurysm (IA). An increasing number of imaging studies have caused that the chance of finding an incidental aneurysm is becoming more common. Since IA rupture causes subarachnoidal hemorrhage (SAH) and have significant mortality and morbidity prophylactic treatment should be considered when IA is detected. The benefit and risk of treatment of IA is based on epidemiological estimate which takes account patient and aneurysm characteristics. However we know that aneurysm rupture is biological process where inflammation of aneurysm wall is actively leading to degeneration of aneurysm wall and finally weakens it until it bursts. Until now, there have not been imaging method to detect inflammatory process of aneurysm wall METHODS: We created targeting immunoliposome for use in the imaging of aneurysm. Immunoliposome comprises antibodies against at least one vascular inflammatory marker associated with aneurysm inflammation and a label and/or a contrast agent. RESULTS Histological analysis of IAs where immunoliposome comprises antibodies against vascular inflammation with a label shows promising results for selectively detecting aneurysms inflammation. In magnetic resonance imaging (MRI) we were able to detect immunoliposomes carrying gadolinium. CONCLUSION Our work opens a new avenue for using contrast labeled immunoliposomes for detecting rupture-prone aneurysms. Immunoliposomes can cary gadolinium and selectively bind to inflammatory section of aneurysm that can be detected with MRI. Further research is needed to develop immunoliposomes to be used with MRI in humans to target treatment to those patients who benefit from it the most.
Collapse
Affiliation(s)
- Behnam Rezai Jahromi
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland.
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland.
| | - Vladimir Zamotin
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| | - Christian Code
- PHYLIFE: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Odense, Denmark
| | - Eliisa Netti
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| | - Martina B Lorey
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, Finland
| | - Kari Alitalo
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, Finland
| | - Katariina Öörni
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| | - Riikka Tulamo
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
- Department of Vascular Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mika Niemelä
- Department of Neurosurgery, Helsinki University Hospital and University of Helsinki, Topeliuksenkatu 5, 00260, Helsinki, Finland
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Dai Z, Zhang Y, Meng Y, Li S, Suonan Z, Sun Y, Ji J, Shen Q, Zheng H, Xue Y. Targeted delivery of nutraceuticals derived from food for the treatment of obesity and its related complications. Food Chem 2023; 418:135980. [PMID: 36989644 DOI: 10.1016/j.foodchem.2023.135980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023]
Abstract
Nutraceuticals which are abundant in foods have attracted much attention due to their bioactive activities of anti-obesity, anti-hyperlipidemia and anti-atherosclerosis. Unfortunately, the poor bioavailability severely undermines their envisioned benefits. Therefore, there is an urgent need to develop suitable delivery systems to promote the benefits of their biological activity. Targeted drug delivery system (TDDS) is a novel drug delivery system that can selectively concentrate drugs on targets in the body, improve the bioavailability of agents and reduce side effects. This emerging drug delivery system provides a new strategy for the treatment of obesity with nutraceuticals and would be a promising alternative to be widely used in the food field. This review summarizes the recent studies on the application in the targeted delivery of nutraceuticals for treating obesity and its related complications, especially the available receptors and their corresponding ligands for TDDS and the evaluation methods of the targeting ability.
Collapse
|
5
|
Poznyak AV, Sukhorukov VN, Eremin II, Nadelyaeva II, Orekhov AN. Diagnostics of atherosclerosis: Overview of the existing methods. Front Cardiovasc Med 2023; 10:1134097. [PMID: 37229223 PMCID: PMC10203409 DOI: 10.3389/fcvm.2023.1134097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/11/2023] [Indexed: 05/27/2023] Open
Abstract
Atherosclerosis was and remains an extremely common and serious health problem. Since the elderly are most at risk of cardiovascular risk, and the average life expectancy is increasing, the spread of atherosclerosis and its consequences increases as well. One of the features of atherosclerosis is its asymptomaticity. This factor makes it difficult to make a timely diagnosis. This entails the lack of timely treatment and even prevention. To date, in the arsenal of physicians, there is only a limited set of methods to suspect and fully diagnose atherosclerosis. In this review, we have tried to briefly describe the most common and effective methods for diagnosing atherosclerosis.
Collapse
|
6
|
Xu H, Li S, Liu YS. Nanoparticles in the diagnosis and treatment of vascular aging and related diseases. Signal Transduct Target Ther 2022; 7:231. [PMID: 35817770 PMCID: PMC9272665 DOI: 10.1038/s41392-022-01082-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 11/09/2022] Open
Abstract
Aging-induced alternations of vasculature structures, phenotypes, and functions are key in the occurrence and development of vascular aging-related diseases. Multiple molecular and cellular events, such as oxidative stress, mitochondrial dysfunction, vascular inflammation, cellular senescence, and epigenetic alterations are highly associated with vascular aging physiopathology. Advances in nanoparticles and nanotechnology, which can realize sensitive diagnostic modalities, efficient medical treatment, and better prognosis as well as less adverse effects on non-target tissues, provide an amazing window in the field of vascular aging and related diseases. Throughout this review, we presented current knowledge on classification of nanoparticles and the relationship between vascular aging and related diseases. Importantly, we comprehensively summarized the potential of nanoparticles-based diagnostic and therapeutic techniques in vascular aging and related diseases, including cardiovascular diseases, cerebrovascular diseases, as well as chronic kidney diseases, and discussed the advantages and limitations of their clinical applications.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.,Institute of Aging and Age-related Disease Research, Central South University, 410011, Changsha, Hunan, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.,Institute of Aging and Age-related Disease Research, Central South University, 410011, Changsha, Hunan, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China. .,Institute of Aging and Age-related Disease Research, Central South University, 410011, Changsha, Hunan, China.
| |
Collapse
|
7
|
Targeting vascular inflammation through emerging methods and drug carriers. Adv Drug Deliv Rev 2022; 184:114180. [PMID: 35271986 PMCID: PMC9035126 DOI: 10.1016/j.addr.2022.114180] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a common dangerous component of pathogenesis of many prevalent conditions with high morbidity and mortality including sepsis, thrombosis, acute respiratory distress syndrome (ARDS), COVID-19, myocardial and cerebral ischemia-reperfusion, infection, and trauma. Inflammatory changes of the vasculature and blood mediate the course and outcome of the pathology in the tissue site of insult, remote organs and systemically. Endothelial cells lining the luminal surface of the vasculature play the key regulatory functions in the body, distinct under normal vs. pathological conditions. In theory, pharmacological interventions in the endothelial cells might enable therapeutic correction of the overzealous damaging pro-inflammatory and pro-thrombotic changes in the vasculature. However, current agents and drug delivery systems (DDS) have inadequate pharmacokinetics and lack the spatiotemporal precision of vascular delivery in the context of acute inflammation. To attain this level of precision, many groups design DDS targeted to specific endothelial surface determinants. These DDS are able to provide specificity for desired tissues, organs, cells, and sub-cellular compartments needed for a particular intervention. We provide a brief overview of endothelial determinants, design of DDS targeted to these molecules, their performance in experimental models with focus on animal studies and appraisal of emerging new approaches. Particular attention is paid to challenges and perspectives of targeted therapeutics and nanomedicine for advanced management of acute inflammation.
Collapse
|
8
|
Naman S, Naryal S, Palliwal R, Paliwal SR, Baldi A. Combating atherosclerosis with nanodrug delivery approaches: from bench side to commercialization. DRUG DELIVERY SYSTEMS FOR METABOLIC DISORDERS 2022:97-136. [DOI: 10.1016/b978-0-323-99616-7.00021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
9
|
Marson D, Aulic S, Fermeglia A, Laurini E, Pricl S. Nanovesicles for the delivery of cardiovascular drugs. APPLICATIONS OF NANOVESICULAR DRUG DELIVERY 2022:341-369. [DOI: 10.1016/b978-0-323-91865-7.00009-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Onishchenko N, Tretiakova D, Vodovozova E. Spotlight on the protein corona of liposomes. Acta Biomater 2021; 134:57-78. [PMID: 34364016 DOI: 10.1016/j.actbio.2021.07.074] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/19/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Although an established drug delivery platform, liposomes have not fulfilled their true potential. In the body, interactions of liposomes are mediated by the layer of plasma proteins adsorbed on the surface, the protein corona. The review aims to collect the data of the last decade on liposome protein corona, tracing the path from interactions of individual proteins to the effects mediated by the protein corona in vivo. It offers a classification of the approaches to exploitation of the protein corona-rather than elimination thereof-based on the bilayer composition-corona composition-molecular interactions-biological performance framework. The multitude of factors that affect each level of this relationship urge to the widest implementation of bioinformatics tools to predict the most effective liposome compositions relying on the data on protein corona. Supplementing the picture with new pieces of accurately reported experimental data will contribute to the accuracy and efficiency of the predictions. STATEMENT OF SIGNIFICANCE: The review focuses on liposomes as an established nanomedicine platform and analyzes the available data on how the protein corona formed on liposome surface in biological fluids affects performance of the liposomes. The review offers a rigorous account of existing literature and critical analysis of methodology currently applied to the assessment of liposome-plasma protein interactions. It introduces a classification of the approaches to exploitation of the protein corona and tailoring liposome carriers to advance the field of nanoparticulate drug delivery systems for the benefit of patients.
Collapse
|
11
|
Zia A, Wu Y, Nguyen T, Wang X, Peter K, Ta HT. The choice of targets and ligands for site-specific delivery of nanomedicine to atherosclerosis. Cardiovasc Res 2021; 116:2055-2068. [PMID: 32077918 DOI: 10.1093/cvr/cvaa047] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
As nanotechnologies advance into clinical medicine, novel methods for applying nanomedicine to cardiovascular diseases are emerging. Extensive research has been undertaken to unlock the complex pathogenesis of atherosclerosis. However, this complexity presents challenges to develop effective imaging and therapeutic modalities for early diagnosis and acute intervention. The choice of ligand-receptor system vastly influences the effectiveness of nanomedicine. This review collates current ligand-receptor systems used in targeting functionalized nanoparticles for diagnosis and treatment of atherosclerosis. Our focus is on the binding affinity and selectivity of ligand-receptor systems, as well as the relative abundance of targets throughout the development and progression of atherosclerosis. Antibody-based targeting systems are currently the most commonly researched due to their high binding affinities when compared with other ligands, such as antibody fragments, peptides, and other small molecules. However, antibodies tend to be immunogenic due to their size. Engineering antibody fragments can address this issue but will compromise their binding affinity. Peptides are promising ligands due to their synthetic flexibility and low production costs. Alongside the aforementioned binding affinity of ligands, the choice of target and its abundance throughout distinct stages of atherosclerosis and thrombosis is relevant to the intended purpose of the nanomedicine. Further studies to investigate the components of atherosclerotic plaques are required as their cellular and molecular profile shifts over time.
Collapse
Affiliation(s)
- Adil Zia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yuao Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Tuan Nguyen
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
12
|
New Approaches in Nanomedicine for Ischemic Stroke. Pharmaceutics 2021; 13:pharmaceutics13050757. [PMID: 34065179 PMCID: PMC8161190 DOI: 10.3390/pharmaceutics13050757] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke, caused by the interruption of blood flow to the brain and subsequent neuronal death, represents one of the main causes of disability in developed countries. Therapeutic methods such as recanalization approaches, neuroprotective drugs, or recovery strategies have been widely developed to improve the patient's outcome; however, important limitations such as a narrow therapeutic window, the ability to reach brain targets, or drug side effects constitute some of the main aspects that limit the clinical applicability of the current treatments. Nanotechnology has emerged as a promising tool to overcome many of these drug limitations and improve the efficacy of treatments for neurological diseases such as stroke. The use of nanoparticles as a contrast agent or as drug carriers to a specific target are some of the most common approaches developed in nanomedicine for stroke. Throughout this review, we have summarized our experience of using nanotechnology tools for the study of stroke and the search for novel therapies.
Collapse
|
13
|
Oh HJ, Kim J, Kim H, Choi N, Chung S. Microfluidic Reconstitution of Tumor Microenvironment for Nanomedical Applications. Adv Healthc Mater 2021; 10:e2002122. [PMID: 33576178 DOI: 10.1002/adhm.202002122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Indexed: 12/17/2022]
Abstract
Nanoparticles have an extensive range of diagnostic and therapeutic applications in cancer treatment. However, their current clinical translation is slow, mainly due to the failure to develop preclinical evaluation techniques that can draw similar conclusions to clinical outcomes by adequately mimicking nanoparticle behavior in complicated tumor microenvironments (TMEs). Microfluidic methods offer significant advantages over conventional in vitro methods to resolve these challenges by recapitulating physiological cues of the TME such as the extracellular matrix, shear stress, interstitial flow, soluble factors, oxygen, and nutrient gradients. The methods are capable of de-coupling microenvironmental features, spatiotemporal controlling of experimental sequences, and high throughput readouts in situ. This progress report highlights the recent achievements of microfluidic models to reconstitute the physiological microenvironment, especially for nanomedical tools for cancer treatment.
Collapse
Affiliation(s)
- Hyun Jeong Oh
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Jaehoon Kim
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Hyunho Kim
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- Division of Bio‐Medical Science & Technology KIST School Korea University of Science and Technology (UST) Seoul 34113 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| |
Collapse
|
14
|
Cheng YA, Wu TH, Wang YM, Cheng TL, Chen IJ, Lu YC, Chuang KH, Wang CK, Chen CY, Lin RA, Chen HJ, Liao TY, Liu ES, Chen FM. Humanized bispecific antibody (mPEG × HER2) rapidly confers PEGylated nanoparticles tumor specificity for multimodality imaging in breast cancer. J Nanobiotechnology 2020; 18:118. [PMID: 32854720 PMCID: PMC7457265 DOI: 10.1186/s12951-020-00680-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Developing a universal strategy to improve the specificity and sensitivity of PEGylated nanoaparticles (PEG-NPs) for assisting in the diagnosis of tumors is important in multimodality imaging. Here, we developed the anti-methoxypolyethylene glycol (mPEG) bispecific antibody (BsAb; mPEG × HER2), which has dual specificity for mPEG and human epidermal growth factor receptor 2 (HER2), with a diverse array of PEG-NPs to confer nanoparticles with HER2 specificity and stronger intensity. Result We used a one-step formulation to rapidly modify the nanoprobes with mPEG × HER2 and optimized the modified ratio of BsAbs on several PEG-NPs (Lipo-DiR, SPIO, Qdot and AuNP). The αHER2/PEG-NPs could specifically target MCF7/HER2 cells (HER2++) but not MCF7/neo1 cells (HER2+/−). The αHER2/Lipo-DiR and αHER2/SPIO could enhance the sensitivity of untargeted PEG-NPs on MCF7/HER2 (HER2++). In in vivo imaging, αHER2/Lipo-DiR and αHER2/SPIO increased the specific targeting and enhanced PEG-NPs accumulation at 175% and 187% on 24 h, respectively, in HER2-overexpressing tumors. Conclusion mPEG × HER2, therefore, provided a simple one-step formulation to confer HER2-specific targeting and enhanced sensitivity and contrast intensity on HER2 positive tumors for multimodality imaging. ![]()
Collapse
Affiliation(s)
- Yi-An Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Tung-Ho Wu
- Cardiovascular Division of Surgical Department, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd, Zuoying Dist, Kaohsiung, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, No. 1001, University Road, Hsinchu, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - I-Ju Chen
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Yun-Chi Lu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, No.172-1, Sec. 2, Keelung Rd, Taipei, Taiwan
| | - Chih-Kuang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Chiao-Yun Chen
- Department of Radiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan
| | - Rui-An Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Huei-Jen Chen
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Tzu-Yi Liao
- Graduate Institute of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan
| | - Fang-Ming Chen
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Road, Kaohsiung, Taiwan. .,Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, No.68, Jhonghua 3rd Rd, Cianjin District, Kaohsiung, Taiwan. .,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan. .,Drug Development and Value Creation Research Center, Kaohsiung Medical University, No.100, Shin-Chuan 1st Road, Sanmin Dist, Kaohsiung, Taiwan.
| |
Collapse
|
15
|
Hu B, Boakye‐Yiadom KO, Yu W, Yuan Z, Ho W, Xu X, Zhang X. Nanomedicine Approaches for Advanced Diagnosis and Treatment of Atherosclerosis and Related Ischemic Diseases. Adv Healthc Mater 2020; 9:e2000336. [PMID: 32597562 DOI: 10.1002/adhm.202000336] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/30/2020] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVDs) remain one of the major causes of mortality worldwide. In response to this and other worldwide health epidemics, nanomedicine has emerged as a rapidly evolving discipline that involves the development of innovative nanomaterials and nanotechnologies and their applications in therapy and diagnosis. Nanomedicine presents unique advantages over conventional medicines due to the superior properties intrinsic to nanoscopic therapies. Once used mainly for cancer therapies, recently, tremendous progress has been made in nanomedicine that has led to an overall improvement in the treatment and diagnosis of CVDs. This review elucidates the pathophysiology and potential targets of atherosclerosis and associated ischemic diseases. It may be fruitful to pursue future work in the nanomedicine-mediated treatment of CVDs based on these targets. A comprehensive overview is then provided featuring the latest preclinical and clinical outcomes in cardiovascular imaging, biomarker detection, tissue engineering, and nanoscale delivery, with specific emphasis on nanoparticles, nanostructured scaffolds, and nanosensors. Finally, the challenges and opportunities regarding the future development and clinical translation of nanomedicine in related fields are discussed. Overall, this review aims to provide a deep and thorough understanding of the design, application, and future development of nanomedicine for atherosclerosis and related ischemic diseases.
Collapse
Affiliation(s)
- Bin Hu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Kofi Oti Boakye‐Yiadom
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Wei Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Zi‐Wei Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| |
Collapse
|
16
|
Meghani NM, Amin H, Park C, Cui JH, Cao QR, Choi KH, Lee BJ. Combinatory interpretation of protein corona and shear stress for active cancer targeting of bioorthogonally clickable gelatin-oleic nanoparticles. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110760. [PMID: 32279783 DOI: 10.1016/j.msec.2020.110760] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 02/16/2020] [Accepted: 02/16/2020] [Indexed: 12/20/2022]
Abstract
Nanoparticle-protein interactions under conditions mimicking physiology determine how nanoparticles (NPs) will behave inside blood vessels and, therefore, the overall outcome of the drug-delivery system. Here, for the first time, we explore the effects of bio-mimicking shear stress and protein corona conditions on novel active targeting of clickable fattigation nanoparticles (NPs) for cancer therapy. Active targeting dibenzocyclooctyne-functionalized biocompatible gelatin-oleic NPs (GON-DBCOs) via a bioorthogonal click reaction were prepared by the desolvation method for delivery of docetaxel (DTX) to lung and breast cancer models. The effect of shear stress (5 dyne/cm2) and human serum albumin (HSA) protein corona on the cellular behavior of NPs was explored under a dynamic microfluidic system in lung (A549) and breast (MCF-7) cancer cell lines. The developed drug-loaded NPs had a particle size of 300 nm, a narrow size distribution, positive zeta potential, high encapsulation efficacy (72.4%), and spherical morphology. The particle size of the protein corona-coated NPs increased to 341 nm with a negative zeta potential. The inhibitory dose (IC50) increased approximately 3- and 42-fold in A549 and MCF-7 cells, respectively, under dynamic microfluidic conditions compared to static conditions. Cellular uptake was significantly decreased in the presence of shear stress and a protein corona, compared with static conditions, in both lung (A549, **p < 0.01) and breast (MCF-7, *p < 0.05) cancer cell lines. Clathrin-and energy-dependent pathways were found to be involved in the cellular uptake of NPs. This study could serve as a vital tool for the evaluation of NPs under aggressive bio-mimicking conditions comprising shear stress and a protein corona to predict the in vivo performance of NPs and support the preclinical and clinical translation of NP drug delivery systems.
Collapse
Affiliation(s)
- Nileshkumar M Meghani
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; Advanced Micro-Mechatronics Lab, Mechatronics Engineering, Jeju National University, Jeju City 63243, Republic of Korea.
| | - Hardik Amin
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Chulhun Park
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea.
| | - Jing-Hao Cui
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| | - Qing-Ri Cao
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China.
| | - Kyung Hyun Choi
- Advanced Micro-Mechatronics Lab, Mechatronics Engineering, Jeju National University, Jeju City 63243, Republic of Korea.
| | - Beom-Jin Lee
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
17
|
Deng Y, Zhang X, Shen H, He Q, Wu Z, Liao W, Yuan M. Application of the Nano-Drug Delivery System in Treatment of Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 7:489. [PMID: 32083068 PMCID: PMC7005934 DOI: 10.3389/fbioe.2019.00489] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) have become a serious threat to human life and health. Though many drugs acting via different mechanism of action are available in the market as conventional formulations for the treatment of CVDs, they are still far from satisfactory due to poor water solubility, low biological efficacy, non-targeting, and drug resistance. Nano-drug delivery systems (NDDSs) provide a new drug delivery method for the treatment of CVDs with the development of nanotechnology, demonstrating great advantages in solving the above problems. Nevertheless, there are some problems about NDDSs need to be addressed, such as cytotoxicity. In this review, the types and targeting strategies of NDDSs were summarized, and the new research progress in the diagnosis and therapy of CVDs in recent years was reviewed. Future prospective for nano-carriers in drug delivery for CVDs includes gene therapy, in order to provide more ideas for the improvement of cardiovascular drugs. In addition, its safety was also discussed in the review.
Collapse
Affiliation(s)
- Yudi Deng
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xudong Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haibin Shen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qiangnan He
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zijian Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
18
|
Wang X, Liang Q, Mao Y, Zhang R, Deng Q, Chen Y, Zhu R, Duan S, Yin L. Bioreducible, branched poly(β-amino ester)s mediate anti-inflammatory ICAM-1 siRNA delivery against myocardial ischemia reperfusion (IR) injury. Biomater Sci 2020; 8:3856-3870. [DOI: 10.1039/d0bm00631a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
ICAM-1 siRNA delivery mediated by bioreducible, branched BPAE-SS toward the anti-inflammatory treatment of myocardial IR injury.
Collapse
Affiliation(s)
- Xiao Wang
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Collaborative Innovation Center of Suzhou Nano Science & Technology
- Soochow University
- Suzhou 215123
| | - Qiujun Liang
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Collaborative Innovation Center of Suzhou Nano Science & Technology
- Soochow University
- Suzhou 215123
| | - Yiming Mao
- Department of Cardiothoracic Surgery
- the Second Affiliated Hospital of Soochow University
- Suzhou 214804
- China
- Department of Thoracic Surgery
| | - Rujing Zhang
- Department of Micro- and Nanotechnology
- DTU Nanotech
- Technical University of Denmark
- 2800 Kgs. Lyngby
- Denmark
| | - Qiurong Deng
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Collaborative Innovation Center of Suzhou Nano Science & Technology
- Soochow University
- Suzhou 215123
| | - Yongbing Chen
- Department of Cardiothoracic Surgery
- the Second Affiliated Hospital of Soochow University
- Suzhou 214804
- China
| | - Rongying Zhu
- Department of Cardiothoracic Surgery
- the Second Affiliated Hospital of Soochow University
- Suzhou 214804
- China
| | - Shanzhou Duan
- Department of Cardiothoracic Surgery
- the Second Affiliated Hospital of Soochow University
- Suzhou 214804
- China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices
- Collaborative Innovation Center of Suzhou Nano Science & Technology
- Soochow University
- Suzhou 215123
| |
Collapse
|
19
|
Kiaie N, Gorabi AM, Penson PE, Watts G, Johnston TP, Banach M, Sahebkar A. A new approach to the diagnosis and treatment of atherosclerosis: the era of the liposome. Drug Discov Today 2019; 25:58-72. [PMID: 31525463 DOI: 10.1016/j.drudis.2019.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/11/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
The consequences of atherosclerotic cardiovascular disease (ASCVD) include myocardial infarction, ischemic stroke, and angina pectoris, which are major causes of mortality and morbidity worldwide. Despite current therapeutic strategies to reduce risk, patients still experience the consequences of ASCVD. Consequently, a current goal is to enhance visualization of early atherosclerotic lesions to improve residual ASCVD risk. The uses of liposomes, in the context of ASCVD, can include as contrast agents for imaging techniques, as well as for the delivery of antiatherosclerotic drugs, genes, and cells to established sites of plaque. Additionally, liposomes have a role as vaccine adjuvants against mediators of atherosclerosis. Here. we review the scientific and clinical evidence relating to the use of liposomes in the diagnosis and management of ASCVD.
Collapse
Affiliation(s)
- Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Gerald Watts
- Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MI, USA
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Da Silva-Candal A, Brown T, Krishnan V, Lopez-Loureiro I, Ávila-Gómez P, Pusuluri A, Pérez-Díaz A, Correa-Paz C, Hervella P, Castillo J, Mitragotri S, Campos F. Shape effect in active targeting of nanoparticles to inflamed cerebral endothelium under static and flow conditions. J Control Release 2019; 309:94-105. [DOI: 10.1016/j.jconrel.2019.07.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
|
21
|
Tan A, Lam YY, Pacot O, Hawley A, Boyd BJ. Probing cell-nanoparticle (cubosome) interactions at the endothelial interface: do tissue dimension and flow matter? Biomater Sci 2019; 7:3460-3470. [PMID: 31268062 DOI: 10.1039/c9bm00243j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the research field of nanostructured systems for biomedical applications, increasing attention has been paid to using biomimetic, dynamic cellular models to adequately predict their bio-nano behaviours. This work specifically evaluates the biointeractions of nanostructured lipid-based particles (cubosomes) with human vascular cells from the aspects of tissue dimension (conventional 2D well plate versus 3D dynamic tubular vasculature) and shear flow effect (static, venous and arterial flow-mimicking conditions). A glass capillary-hosted, 3D tubular endothelial construct was coupled with circulating luminal fluid flow to simulate the human vascular systems. In the absence of fluid flow, the degree of cell-cubosome association was not significantly different between the 2D planar and the 3D tubular systems. Under flow conditions simulating venous (0.8 dynes per cm2) and arterial (10 dynes per cm2) shear stresses, the cell-cubosome association notably declined by 50% and 98%, respectively. This highlights the significance of shear-guided biointeractions of non-targeted nanoparticles in the circulation. Across all 2D and 3D cellular models with and without flow, cubosomes had little effect on the cell-cell contact based on the unchanged immunoexpression of the endothelial-specific intercellular junction marker PECAM-1. Interestingly, there were dissimilar nanoparticle distribution patterns between the 2D planar (showing discrete punctate staining) and the 3D tubular endothelium (with a more diffused, patchy fashion). Taken together, these findings highlight the importance of tissue dimension and shear flow in governing the magnitude and feature of cell-nanoparticle interactions.
Collapse
Affiliation(s)
- Angel Tan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 3052 Victoria, Australia. and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University (Parkville Campus), 3052 Victoria, Australia
| | - Yuen Yi Lam
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 3052 Victoria, Australia. and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University (Parkville Campus), 3052 Victoria, Australia
| | - Olivier Pacot
- Institute of Systems Engineering, School of Engineering, University of Applied Sciences and Arts Western Switzerland, 1950 Sion, Switzerland
| | - Adrian Hawley
- Australian Synchrotron, ANSTO, 3168 Victoria, Australia
| | - Ben J Boyd
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 3052 Victoria, Australia. and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University (Parkville Campus), 3052 Victoria, Australia
| |
Collapse
|
22
|
Ruzycka M, Cimpan MR, Rios-Mondragon I, Grudzinski IP. Microfluidics for studying metastatic patterns of lung cancer. J Nanobiotechnology 2019; 17:71. [PMID: 31133019 PMCID: PMC6537392 DOI: 10.1186/s12951-019-0492-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/04/2019] [Indexed: 01/09/2023] Open
Abstract
The incidence of lung cancer continues to rise worldwide. Because the aggressive metastasis of lung cancer cells is the major drawback of successful therapies, the crucial challenge of modern nanomedicine is to develop diagnostic tools to map the molecular mechanisms of metastasis in lung cancer patients. In recent years, microfluidic platforms have been given much attention as tools for novel point-of-care diagnostic, an important aspect being the reconstruction of the body organs and tissues mimicking the in vivo conditions in one simple microdevice. Herein, we present the first comprehensive overview of the microfluidic systems used as innovative tools in the studies of lung cancer metastasis including single cancer cell analysis, endothelial transmigration, distant niches migration and finally neoangiogenesis. The application of the microfluidic systems to study the intercellular crosstalk between lung cancer cells and surrounding tumor microenvironment and the connection with multiple molecular signals coming from the external cellular matrix are discussed. We also focus on recent breakthrough technologies regarding lab-on-chip devices that serve as tools for detecting circulating lung cancer cells. The superiority of microfluidic systems over traditional in vitro cell-based assays with regard to modern nanosafety studies and new cancer drug design and discovery is also addressed. Finally, the current progress and future challenges regarding printable and paper-based microfluidic devices for personalized nanomedicine are summarized.
Collapse
Affiliation(s)
- Monika Ruzycka
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland
| | - Mihaela R Cimpan
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ivan Rios-Mondragon
- Biomaterials - Department for Clinical Dentistry, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Ireneusz P Grudzinski
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097, Warsaw, Poland.
| |
Collapse
|
23
|
Ran R, Wang H, Hou F, Liu Y, Hui Y, Petrovsky N, Zhang F, Zhao C. A Microfluidic Tumor-on-a-Chip for Assessing Multifunctional Liposomes' Tumor Targeting and Anticancer Efficacy. Adv Healthc Mater 2019; 8:e1900015. [PMID: 30868753 DOI: 10.1002/adhm.201900015] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/03/2019] [Indexed: 01/04/2023]
Abstract
Two principal methods for cancer drug testing are widely used, namely, in vitro 2D cell monolayers and in vivo animal models. In vitro 2D culture systems are simple and convenient but are unable to capture the complexity of biological processes. Animal models are costly, time-consuming, and often fail to replicate human activity. Here a microfluidic tumor-on-a-chip (TOC) model designed for assessing multifunctional liposome cancer targeting and efficacy is presented. The TOC device contains three sets of hemispheric wells with different sizes for tumor spheroid formation and evaluation of liposomes under a controlled flow condition. There is good agreement between time-elapsed tumor targeting of fluorescent liposomes in the TOC model and in in vivo mouse models. Evaluation of the anticancer efficacy of four PTX-loaded liposome formulations shows that compared to 2D cell monolayers and 3D tumor spheroid models, the TOC model better predicts the in vivo anticancer efficacy of targeted liposomes. Lastly, the TOC model is used to assess the effects of flow rates and tumor size on treatment outcome. This study demonstrates that the TOC model provides a convenient and powerful platform for rapid and reliable cancer drug evaluation.
Collapse
Affiliation(s)
- Rui Ran
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St. Lucia QLD 4072 Australia
| | - Hao‐Fei Wang
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St. Lucia QLD 4072 Australia
| | - Fei Hou
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St. Lucia QLD 4072 Australia
| | - Yun Liu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St. Lucia QLD 4072 Australia
| | - Yue Hui
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St. Lucia QLD 4072 Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd Bedford Park SA 5042 Australia
- Department of Endocrinology Flinders University Bedford Park SA 5042 Australia
| | - Fan Zhang
- Department of Chemistry State Key Laboratory of Molecular Engineering of Polymers Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem Fudan University Shanghai 200438 China
| | - Chun‐Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St. Lucia QLD 4072 Australia
| |
Collapse
|
24
|
He Z, Ranganathan N, Li P. Evaluating nanomedicine with microfluidics. NANOTECHNOLOGY 2018; 29:492001. [PMID: 30215611 DOI: 10.1088/1361-6528/aae18a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanomedicines are engineered nanoscale structures that have an extensive range of application in the diagnosis and therapy of many diseases. Despite the rapid progress in and tremendous potential of nanomedicines, their clinical translational process is still slow, owing to the difficulty in understanding, evaluating, and predicting their behavior in complex living organisms. Microfluidic techniques offer a promising way to resolve these challenges. Carefully designed microfluidic chips enable in vivo microenvironment simulation and high-throughput analysis, thus providing robust platforms for nanomedicine evaluation. Here, we summarize the recent developments and achievements in microfluidic methods for nanomedicine evaluation, categorized into four sections based on their target systems: single cell, multicellular system, organ, and organism levels. Finally, we provide our perspectives on the challenges and future directions of microfluidics-based nanomedicine evaluation.
Collapse
Affiliation(s)
- Ziyi He
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV, United States of America
| | | | | |
Collapse
|
25
|
Gao X, Guo L, Li J, Thu HE, Hussain Z. Nanomedicines guided nanoimaging probes and nanotherapeutics for early detection of lung cancer and abolishing pulmonary metastasis: Critical appraisal of newer developments and challenges to clinical transition. J Control Release 2018; 292:29-57. [PMID: 30359665 DOI: 10.1016/j.jconrel.2018.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 01/13/2023]
Abstract
Lung cancer (LC) is the second most prevalent type of cancer and primary cause of mortality among both men and women, worldwide. The most commonly employed diagnostic modalities for LC include chest X-ray (CXR), magnetic-resonance-imaging (MRI), computed tomography (CT-scan), and fused-positron-emitting-tomography-CT (PET-CT). Owing to several limitations associated with the use of conventional diagnostic tools such as radiation burden to the patient, misleading diagnosis ("missed lung cancer"), false staging and low sensitivity and resolution, contemporary diagnostic regimen needed to be employed for screening of LC. In recent decades, nanotechnology-guided interventions have been transpired as emerging nanoimaging probes for detection of LC at advanced stages, while producing signal amplification, better resolution for surface and deep tissue imaging, and enhanced translocation and biodistribution of imaging probes within the cancerous tissues. Besides enormous potential of nanoimaging probes, nanotechnology-based advancements have also been evidenced for superior efficacy for treatment of LC and abolishing pulmonary metastasis (PM). The success of nanotherapeutics is due to their ability to maximise translocation and biodistribution of anti-neoplastic agents into the tumor tissues, improve pharmacokinetic profiles of anti-metastatic agents, optimise target-specific drug delivery, and control release kinetics of encapsulated moieties in target tissues. This review aims to overview and critically discuss the superiority of nanoimaging probes and nanotherapeutics over conventional regimen for early detection of LC and abolishing PM. Current challenges to clinical transition of nanoimaging probes and therapeutic viability of nanotherapeutics for treatment for LC and PM have also been pondered.
Collapse
Affiliation(s)
- Xiaoling Gao
- Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Lihua Guo
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jianqiang Li
- Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Hnin Ei Thu
- Department of Pharmacology and Dental Therapeutics, Faculty of Dentistry, Lincoln University College, Jalan Stadium, SS 7/15, Kelana Jaya, 47301 Petaling Jaya, Selangor, Malaysia
| | - Zahid Hussain
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
26
|
Effect of flow on targeting and penetration of angiopep-decorated nanoparticles in a microfluidic model blood-brain barrier. PLoS One 2018; 13:e0205158. [PMID: 30300391 PMCID: PMC6177192 DOI: 10.1371/journal.pone.0205158] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 09/20/2018] [Indexed: 12/27/2022] Open
Abstract
The blood-brain barrier (BBB) limits transport of nanoparticles from the circulation to the brain parenchyma. Angiopep-2, a peptide which functions as a brain transport vector, can be coupled to nanoparticles in order to facilitate binding and internalization by brain endothelial cells (ECs), and subsequent BBB penetration. This multi-step process may be affected by blood flow over brain ECs, as flow influences endothelial cell phenotype as well as interactions of nanoparticles with ECs. In the present study a microfluidic BBB model was constructed to evaluate binding and internalization by brain ECs, as well as BBB penetration of Angiopep-2 coupled liposomes (Ang2-Liposomes) in static and flow conditions. Ang2 conjugation to liposomes markedly improved binding relative to unconjugated liposomes. Ang2-Liposomes bound and were internalized efficiently by brain endothelial cells after static incubation or with 1 dyne/cm2 of fluid shear stress (FSS), while binding was reduced at a FSS of 6 dyne/cm2. Penetration of the model microfluidic BBB by Ang2-Liposomes was higher at a FSS of 1 dyne/cm2 and 6 dyne/cm2 than with static incubation. Analysis of barrier function and control experiments for receptor-mediated penetration provided insight into the magnitude of transcellular versus paracellular transport at each tested FSS. Overall, the results demonstrate that flow impacted the binding and BBB penetration of Ang2-functionalized nanoparticles. This highlights the relevance of the local flow environment for in vitro modeling of the performance of nanoparticles functionalized with BBB penetrating ligands.
Collapse
|
27
|
Zhang Q, Hu S, Wang K, Cui M, Li X, Wang M, Hu X. Engineering a yeast double-molecule carrier for drug screening. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:386-396. [PMID: 29611428 DOI: 10.1080/21691401.2018.1457539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
With the advantages of unicellular eukaryotic structure and easy manipulation, yeast becomes a popular tool for biochemical, genetic and medicinal studies. In order to construct an efficient anti-inflammatory drug screening platform, we engineered yeast as a double-molecule carrier, of which an inserted domain (I domain) of lymphocyte function-associated antigen 1 was displayed on yeast surface and a green fluorescent protein (GFP) was expressed inside cytosol. The I domain specifically targeted a surface marker of mammalian cells, intercellular adhesion molecule 1, whose number is correlated with the level of cellular inflammation. Examination of GFP intensity enables swift quantification of the yeast-mammalian cell binding and thus it reflects inflammatory potency, herein the inflammatory index, of a chemical imposed to cells. The inflammatory potency of a total of 1340 chemicals was indexed. Among them, 1 inflammation-inducing and 1 inflammation-reducing compounds were verified both in vitro and in vivo. Our method demonstrated a swift, facile and high-throughput screening platform at the protein level for inflammation and related diseases drug discovery without using sophisticated instruments.
Collapse
Affiliation(s)
- Qiyun Zhang
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| | - Sheng Hu
- d Hubei Cancer Hospital , Wuhan , China
| | - Ke Wang
- e State Key Laboratory of Agricultural Microbiology , HZAU , Wuhan , China.,f College of Veterinary Medicine , HZAU , Wuhan , China
| | - Min Cui
- e State Key Laboratory of Agricultural Microbiology , HZAU , Wuhan , China.,f College of Veterinary Medicine , HZAU , Wuhan , China
| | - Xiaohua Li
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| | - Mo Wang
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| | - Xuebo Hu
- a Laboratory of Drug Discovery and Molecular Engineering, Department of Medicinal Plants, College of Plant Science and Technology , Huazhong Agricultural University (HZAU) , Wuhan , China.,b National & Local Joint Engineering Research Center (Hubei) for Medicinal Plant Breeding and Cultivation , Wuhan , China.,c Hubei Provincial Engineering Research Center for Medicinal Plants , Wuhan , China
| |
Collapse
|
28
|
Nakhlband A, Eskandani M, Omidi Y, Saeedi N, Ghaffari S, Barar J, Garjani A. Combating atherosclerosis with targeted nanomedicines: recent advances and future prospective. ACTA ACUST UNITED AC 2018; 8:59-75. [PMID: 29713603 PMCID: PMC5915710 DOI: 10.15171/bi.2018.08] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/21/2018] [Indexed: 12/19/2022]
Abstract
![]()
Introduction:
Cardiovascular diseases (CVDs) is recognized as the leading cause of mortality worldwide. The increasing prevalence of such disease demands novel therapeutic and diagnostic approaches to overcome associated clinical/social issues. Recent advances in nanotechnology and biological sciences have provided intriguing insights to employ targeted Nanomachines to the desired location as imaging, diagnosis, and therapeutic modalities. Nanomedicines as novel tools for enhanced drug delivery, imaging, and diagnosis strategies have shown great promise to combat cardiovascular diseases.
Methods:
In the current study, we intend to review the most recent studies on the nano-based strategies for improved management of CVDs.
Results:
A cascade of events results in the formation of atheromatous plaque and arterial stenosis. Furthermore, recent studies have shown that nanomedicines have displayed unique functionalities and provided de novo applications in the diagnosis and treatment of atherosclerosis.
Conclusion:
Despite some limitations, nanomedicines hold considerable potential in the prevention, diagnosis, and treatment of various ailments including atherosclerosis. Fewer side effects, amenable physicochemical properties and multi-potential application of such nano-systems are recognized through various investigations. Therefore, it is strongly believed that with targeted drug delivery to atherosclerotic lesions and plaque, management of onset and progression of disease would be more efficient than classical treatment modalities.
Collapse
Affiliation(s)
- Ailar Nakhlband
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Dai Q, Bertleff‐Zieschang N, Braunger JA, Björnmalm M, Cortez‐Jugo C, Caruso F. Particle Targeting in Complex Biological Media. Adv Healthc Mater 2018; 7. [PMID: 28809092 DOI: 10.1002/adhm.201700575] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/04/2017] [Indexed: 12/22/2022]
Abstract
Over the past few decades, nanoengineered particles have gained increasing interest for applications in the biomedical realm, including diagnosis, imaging, and therapy. When functionalized with targeting ligands, these particles have the potential to interact with specific cells and tissues, and accumulate at desired target sites, reducing side effects and improve overall efficacy in applications such as vaccination and drug delivery. However, when targeted particles enter a complex biological environment, the adsorption of biomolecules and the formation of a surface coating (e.g., a protein corona) changes the properties of the carriers and can render their behavior unpredictable. For this reason, it is of importance to consider the potential challenges imposed by the biological environment at the early stages of particle design. This review describes parameters that affect the targeting ability of particulate drug carriers, with an emphasis on the effect of the protein corona. We highlight strategies for exploiting the protein corona to improve the targeting ability of particles. Finally, we provide suggestions for complementing current in vitro assays used for the evaluation of targeting and carrier efficacy with new and emerging techniques (e.g., 3D models and flow-based technologies) to advance fundamental understanding in bio-nano science and to accelerate the development of targeted particles for biomedical applications.
Collapse
Affiliation(s)
- Qiong Dai
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Nadja Bertleff‐Zieschang
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Julia A. Braunger
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Christina Cortez‐Jugo
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
30
|
Huang X, Xu MQ, Zhang W, Ma S, Guo W, Wang Y, Zhang Y, Gou T, Chen Y, Liang XJ, Cao F. ICAM-1-Targeted Liposomes Loaded with Liver X Receptor Agonists Suppress PDGF-Induced Proliferation of Vascular Smooth Muscle Cells. NANOSCALE RESEARCH LETTERS 2017; 12:322. [PMID: 28472871 PMCID: PMC5415450 DOI: 10.1186/s11671-017-2097-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/20/2017] [Indexed: 05/05/2023]
Abstract
The proliferation of vascular smooth muscle cells (VSMCs) is one of the key events during the progress of atherosclerosis. The activated liver X receptor (LXR) signalling pathway is demonstrated to inhibit platelet-derived growth factor BB (PDGF-BB)-induced VSMC proliferation. Notably, following PDGF-BB stimulation, the expression of intercellular adhesion molecule-1 (ICAM-1) by VSMCs increases significantly. In this study, anti-ICAM-1 antibody-conjugated liposomes were fabricated for targeted delivery of a water-insoluble LXR agonist (T0901317) to inhibit VSMC proliferation. The liposomes were prepared by filming-rehydration method with uniform size distribution and considerable drug entrapment efficiency. The targeting effect of the anti-ICAM-T0901317 liposomes was evaluated by confocal laser scanning microscope (CLSM) and flow cytometry. Anti-ICAM-T0901317 liposomes showed significantly higher inhibition effect of VSMC proliferation than free T0901317 by CCk8 proliferation assays and BrdU staining. Western blot assay further confirmed that anti-ICAM-T0901317 liposomes inhibited retinoblastoma (Rb) phosphorylation and MCM6 expression. In conclusion, this study identified anti-ICAM-T0901317 liposomes as a promising nanotherapeutic approach to overcome VSMC proliferation during atherosclerosis progression.
Collapse
Affiliation(s)
- Xu Huang
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Meng-Qi Xu
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Zhang
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Sai Ma
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weisheng Guo
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Yabin Wang
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Zhang
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Tiantian Gou
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yundai Chen
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xing-Jie Liang
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, China.
| | - Feng Cao
- Department of Cardiology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
31
|
Wang X, Peter K. Molecular Imaging of Atherothrombotic Diseases: Seeing Is Believing. Arterioscler Thromb Vasc Biol 2017; 37:1029-1040. [PMID: 28450298 DOI: 10.1161/atvbaha.116.306483] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/11/2017] [Indexed: 12/13/2022]
Abstract
Molecular imaging, with major advances in the development of both innovative targeted contrast agents/particles and radiotracers, as well as various imaging technologies, is a fascinating, rapidly growing field with many preclinical and clinical applications, particularly for personalized medicine. Thrombosis in either the venous or the arterial system, the latter typically caused by rupture of unstable atherosclerotic plaques, is a major determinant of mortality and morbidity in patients. However, imaging of the various thrombotic complications and the identification of plaques that are prone to rupture are at best indirect, mostly unreliable, or not available at all. The development of molecular imaging toward diagnosis and prevention of thrombotic disease holds promise for major advance in this clinically important field. Here, we review the medical need and clinical importance of direct molecular imaging of thrombi and unstable atherosclerotic plaques that are prone to rupture, thereby causing thrombotic complications such as myocardial infarction and ischemic stroke. We systematically compare the advantages/disadvantages of the various molecular imaging modalities, including X-ray computed tomography, magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, fluorescence imaging, and ultrasound. We further systematically discuss molecular targets specific for thrombi and those characterizing unstable, potentially thrombogenic atherosclerotic plaques. Finally, we provide examples for first theranostic approaches in thrombosis, combining diagnosis, targeted therapy, and monitoring of therapeutic success or failure. Overall, molecular imaging is a rapidly advancing field that holds promise of major benefits to many patients with atherothrombotic diseases.
Collapse
Affiliation(s)
- Xiaowei Wang
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute (X.W., K.P.), Departments of Medicine (X.W., K.P.), and Immunology (K.P.), Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute (X.W., K.P.), Departments of Medicine (X.W., K.P.), and Immunology (K.P.), Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
32
|
Atukorale PU, Covarrubias G, Bauer L, Karathanasis E. Vascular targeting of nanoparticles for molecular imaging of diseased endothelium. Adv Drug Deliv Rev 2017; 113:141-156. [PMID: 27639317 DOI: 10.1016/j.addr.2016.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 01/08/2023]
Abstract
This review seeks to highlight the enormous potential of targeted nanoparticles for molecular imaging applications. Being the closest point-of-contact, circulating nanoparticles can gain direct access to targetable molecular markers of disease that appear on the endothelium. Further, nanoparticles are ideally suitable to vascular targeting due to geometrically enhanced multivalent attachment on the vascular target. This natural synergy between nanoparticles, vascular targeting and molecular imaging can provide new avenues for diagnosis and prognosis of disease with quantitative precision. In addition to the obvious applications of targeting molecular signatures of vascular diseases (e.g., atherosclerosis), deep-tissue diseases often manifest themselves by continuously altering and remodeling their neighboring blood vessels (e.g., cancer). Thus, the remodeled endothelium provides a wide range of targets for nanoparticles and molecular imaging. To demonstrate the potential of molecular imaging, we present a variety of nanoparticles designed for molecular imaging of cancer or atherosclerosis using different imaging modalities.
Collapse
|
33
|
Khodabandehlou K, Masehi-Lano JJ, Poon C, Wang J, Chung EJ. Targeting cell adhesion molecules with nanoparticles using in vivo and flow-based in vitro models of atherosclerosis. Exp Biol Med (Maywood) 2017; 242:799-812. [PMID: 28195515 DOI: 10.1177/1535370217693116] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Atherosclerosis is a leading cause of death worldwide; in addition to lipid dysfunction, chronic arterial wall inflammation is a key component of atherosclerosis. Techniques that target cell adhesion molecules, which are overexpressed during inflammation, are effective methods to detect and treat atherosclerosis. Specifically, research groups have identified vascular cell adhesion molecule-1, intercellular adhesion molecule-1, platelet endothelial cell adhesion molecule, and selectins (E-selectin and P-selectin) as correlated to atherogenesis. In this review, we discuss recent strategies both in vivo and in vitro that target cell adhesion molecules. First, we discuss peptide-based and antibody (Ab)-based nanoparticles utilized in vivo for diagnostic, therapeutic, and theranostic applications. Second, we discuss flow-based in vitro models that serve to reduce the traditional disadvantages of in vivo studies such as variability, time to develop the disease, and ethical burden, but preserve physiological relevance. The knowledge gained from these targeting studies can be translated into clinical solutions for improved detection, prevention, and treatment of atherosclerosis. Impact statement As atherosclerosis remains the leading cause of death, there is an urgent need to develop better tools for treatment of the disease. The ability to improve current treatments relies on enhancing the accuracy of in vitro and in vivo atherosclerotic models. While in vivo models provide all the relevant testing parameters, variability between animals and among models used is a barrier to reproducible results and comparability of NP efficacy. In vitro cultures isolate cells into microenvironments that fail to take into account flow separation and shear stress, which are characteristics of atherosclerotic lesions. Flow-based in vitro models provide more physiologically relevant platforms, bridging the gap between in vivo and 2D in vitro models. This is the first review that presents recent advances regarding endothelial cell-targeting using adhesion molecules in light of in vivo and flow-based in vitro models, providing insights for future development of optimal strategies against atherosclerosis.
Collapse
Affiliation(s)
- Khosrow Khodabandehlou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Jacqueline J Masehi-Lano
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
34
|
Biocompatible and target specific hydrophobically modified glycol chitosan nanoparticles. Biointerphases 2016; 11:04B301. [PMID: 27126597 DOI: 10.1116/1.4948265] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in the United States. Atherosclerosis is a major cause for cardiovascular diseases. Drugs that treat atherosclerosis usually act nonspecifically. To enhance drug delivery specificity, the authors developed a hydrophobically modified glycol chitosan (HGC) nanoparticle that can specifically target activated endothelial cells. The biocompatibility of these nanoparticles toward red blood cells and platelets was evaluated through hemolysis, platelet activation, platelet thrombogenicity, and platelet aggregation assays. The biocompatibility of these nanoparticles toward vascular endothelial cells was evaluated by their effects on endothelial cell growth, metabolic activity, and activation. The results demonstrated that HGC nanoparticles did not cause hemolysis, or affect platelet activation, thrombogenicity, and aggregation capability in vitro. The nanoparticles did not impair vascular endothelial cell growth or metabolic activities in vitro, and did not cause cell activation either. When conjugated with intercellular adhesion molecular 1 antibodies, HGC nanoparticles showed a significantly increased targeting specificity toward activated endothelial cells. These results suggested that HGC nanoparticles are likely compatible toward red blood cells, platelets, and endothelial cells, and they can be potentially used to identify activated endothelial cells at atherosclerotic lesion areas within the vasculature, and deliver therapeutic drugs.
Collapse
|
35
|
Palekar RU, Jallouk AP, Lanza GM, Pan H, Wickline SA. Molecular imaging of atherosclerosis with nanoparticle-based fluorinated MRI contrast agents. Nanomedicine (Lond) 2016; 10:1817-32. [PMID: 26080701 DOI: 10.2217/nnm.15.26] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
As atherosclerosis remains one of the most prevalent causes of patient mortality, the ability to diagnose early signs of plaque rupture and thrombosis represents a significant clinical need. With recent advances in nanotechnology, it is now possible to image specific molecular processes noninvasively with MRI, using various types of nanoparticles as contrast agents. In the context of cardiovascular disease, it is possible to specifically deliver contrast agents to an epitope of interest for detecting vascular inflammatory processes, which serve as predecessors to atherosclerotic plaque development. Herein, we review various applications of nanotechnology in detecting atherosclerosis using MRI, with an emphasis on perfluorocarbon nanoparticles and fluorine imaging, along with theranostic prospects of nanotechnology in cardiovascular disease.
Collapse
Affiliation(s)
- Rohun U Palekar
- Department of Biomedical Engineering, Washington University, Whitaker Hall, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA
| | - Andrew P Jallouk
- Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| | - Gregory M Lanza
- Department of Biomedical Engineering, Washington University, Whitaker Hall, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA.,Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| | - Hua Pan
- Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| | - Samuel A Wickline
- Department of Biomedical Engineering, Washington University, Whitaker Hall, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA.,Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| |
Collapse
|
36
|
Yoon JH, Kim DK, Na M, Lee SY. Multi-ligand functionalized particle design for cell targeting and drug delivery. Biophys Chem 2016; 213:25-31. [PMID: 27100957 DOI: 10.1016/j.bpc.2016.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/23/2016] [Accepted: 03/27/2016] [Indexed: 11/29/2022]
Abstract
Particle-based delivery systems encompass some of the most promising techniques for therapeutic drug delivery. In particular, multi-functional nanovector systems permit diverse functions such as efficient drug/imaging agent loading and unloading and increased target specificity. To enhance the efficiency of delivery systems, particle size and shape can be altered and specific ligands can be conjugated to the particles to promote interactions with receptors expressed on target cells. Moreover, to maximize efficiency and specificity, multiple types of ligands can be conjugated to the particle surface. To analyze the multi-ligand-receptor mediated adhesion process, we developed a stochastic model considering diverse biophysical parameters, including non-specific interactions, ligand-receptor specific interactions, kinetic affinity between ligand and receptor, hydrodynamic force and particle size. The results demonstrate that limited contact area restricts the probability of adhesion such that multiple ligand-receptor pairs do not always show enhanced adhesion characteristics. To optimize the effect of multiple ligand-receptor pairs, biophysical parameters must be considered.
Collapse
Affiliation(s)
- Jung Hyun Yoon
- Department of Biomedical Engineering, Yonsei University, Wonju, Gangwon 220-170, Republic of Korea
| | - Dae Kyung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, Gangwon 220-170, Republic of Korea
| | - Miso Na
- Department of Biomedical Engineering, Yonsei University, Wonju, Gangwon 220-170, Republic of Korea
| | - Sei Young Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, Gangwon 220-170, Republic of Korea.
| |
Collapse
|
37
|
Yoon JH, Kim DK, Key J, Lee SW, Lee SY. Adhesion characteristics of nano/micro-sized particles with dual ligands with different interaction distances. RSC Adv 2016. [DOI: 10.1039/c6ra14974j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Dual ligand conjugated particle for targeted delivery. Rotational dislodging force on the ligand–receptor interaction. Optimal receptor and ligand ratio exist.
Collapse
Affiliation(s)
- J. H. Yoon
- Department of Biomedical Engineering
- Yonsei University
- Wonju
- Republic of Korea
| | - D. K. Kim
- Department of Biomedical Engineering
- Yonsei University
- Wonju
- Republic of Korea
| | - J. Key
- Department of Biomedical Engineering
- Yonsei University
- Wonju
- Republic of Korea
| | - S. W. Lee
- Department of Biomedical Engineering
- Yonsei University
- Wonju
- Republic of Korea
| | - S. Y. Lee
- Department of Biomedical Engineering
- Yonsei University
- Wonju
- Republic of Korea
| |
Collapse
|
38
|
Chung EJ, Tirrell M. Recent Advances in Targeted, Self-Assembling Nanoparticles to Address Vascular Damage Due to Atherosclerosis. Adv Healthc Mater 2015; 4:2408-22. [PMID: 26085109 PMCID: PMC4760622 DOI: 10.1002/adhm.201500126] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 03/31/2015] [Indexed: 01/03/2023]
Abstract
Self-assembling nanoparticles functionalized with targeting moieties have significant potential for atherosclerosis nanomedicine. While self-assembly allows the easy construction (and degradation) of nanoparticles with therapeutic or diagnostic functionality, or both, the targeting agent can direct them to a specific molecular marker within a given stage of the disease. Therefore, supramolecular nanoparticles have been investigated in the last decade as molecular imaging agents or explored as nanocarriers that can decrease the systemic toxicity of drugs by producing accumulation predominantly in specific tissues of interest. In this Progress Report, the pathogenesis of atherosclerosis and the damage caused to vascular tissue are described, as well as the current diagnostic and treatment options. An overview of targeted strategies using self-assembling nanoparticles is provided, including liposomes, high density lipoproteins, protein cages, micelles, proticles, and perfluorocarbon nanoparticles. Finally, an overview is given of current challenges, limitations, and future applications for personalized medicine in the context of atherosclerosis of self-assembling nanoparticles.
Collapse
Affiliation(s)
- Eun Ji Chung
- Institute for Molecular Engineering, University of Chicago, 5747 S.
Ellis Ave., Chicago, IL, 60637, USA
| | - Matthew Tirrell
- Institute for Molecular Engineering, University of Chicago, 5747 S.
Ellis Ave., Chicago, IL, 60637, USA
| |
Collapse
|
39
|
Godoy-Gallardo M, Ek PK, Jansman MMT, Wohl BM, Hosta-Rigau L. Interaction between drug delivery vehicles and cells under the effect of shear stress. BIOMICROFLUIDICS 2015; 9:052605. [PMID: 26180575 PMCID: PMC4491015 DOI: 10.1063/1.4923324] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/18/2015] [Indexed: 05/06/2023]
Abstract
Over the last decades, researchers have developed an ever greater and more ingenious variety of drug delivery vehicles (DDVs). This has made it possible to encapsulate a wide selection of therapeutic agents, ranging from proteins, enzymes, and peptides to hydrophilic and hydrophobic small drugs while, at the same time, allowing for drug release to be triggered through a diverse range of physical and chemical cues. While these advances are impressive, the field has been lacking behind in translating these systems into the clinic, mainly due to low predictability of in vitro and rodent in vivo models. An important factor within the complex and dynamic human in vivo environment is the shear flow observed within our circulatory system and many other tissues. Within this review, recent advances to leverage microfluidic devices to better mimic these conditions through novel in vitro assays are summarized. By grouping the discussion in three prominent classes of DDVs (lipidic and polymeric particles as well as inorganic nanoparticles), we hope to guide researchers within drug delivery into this exciting field and advance a further implementation of these assay systems within the development of DDVs.
Collapse
Affiliation(s)
- M Godoy-Gallardo
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - P K Ek
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - M M T Jansman
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - B M Wohl
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| | - L Hosta-Rigau
- Department of Micro-and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark , Building 423, 2800 Lyngby, Denmark
| |
Collapse
|
40
|
Setyawati MI, Tay CY, Docter D, Stauber RH, Leong DT. Understanding and exploiting nanoparticles' intimacy with the blood vessel and blood. Chem Soc Rev 2015; 44:8174-99. [PMID: 26239875 DOI: 10.1039/c5cs00499c] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While the blood vessel is seldom the target tissue, almost all nanomedicine will interact with blood vessels and blood at some point of time along its life cycle in the human body regardless of their intended destination. Despite its importance, many bionanotechnologists do not feature endothelial cells (ECs), the blood vessel cells, or consider blood effects in their studies. Including blood vessel cells in the study can greatly increase our understanding of the behavior of any given nanomedicine at the tissue of interest or to understand side effects that may occur in vivo. In this review, we will first describe the diversity of EC types found in the human body and their unique behaviors and possibly how these important differences can implicate nanomedicine behavior. Subsequently, we will discuss about the protein corona derived from blood with foci on the physiochemical aspects of nanoparticles (NPs) that dictate the protein corona characteristics. We would also discuss about how NPs characteristics can affect uptake by the endothelium. Subsequently, mechanisms of how NPs could cross the endothelium to access the tissue of interest. Throughout the paper, we will share some novel nanomedicine related ideas and insights that were derived from the understanding of the NPs' interaction with the ECs. This review will inspire more exciting nanotechnologies that had accounted for the complexities of the real human body.
Collapse
Affiliation(s)
- Magdiel Inggrid Setyawati
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | | | | | | | | |
Collapse
|
41
|
Bakermans AJ, Abdurrachim D, Moonen RPM, Motaal AG, Prompers JJ, Strijkers GJ, Vandoorne K, Nicolay K. Small animal cardiovascular MR imaging and spectroscopy. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2015; 88-89:1-47. [PMID: 26282195 DOI: 10.1016/j.pnmrs.2015.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/09/2015] [Accepted: 03/09/2015] [Indexed: 06/04/2023]
Abstract
The use of MR imaging and spectroscopy for studying cardiovascular disease processes in small animals has increased tremendously over the past decade. This is the result of the remarkable advances in MR technologies and the increased availability of genetically modified mice. MR techniques provide a window on the entire timeline of cardiovascular disease development, ranging from subtle early changes in myocardial metabolism that often mark disease onset to severe myocardial dysfunction associated with end-stage heart failure. MR imaging and spectroscopy techniques play an important role in basic cardiovascular research and in cardiovascular disease diagnosis and therapy follow-up. This is due to the broad range of functional, structural and metabolic parameters that can be quantified by MR under in vivo conditions non-invasively. This review describes the spectrum of MR techniques that are employed in small animal cardiovascular disease research and how the technological challenges resulting from the small dimensions of heart and blood vessels as well as high heart and respiratory rates, particularly in mice, are tackled.
Collapse
Affiliation(s)
- Adrianus J Bakermans
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rik P M Moonen
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abdallah G Motaal
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Gustav J Strijkers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katrien Vandoorne
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
42
|
Maritim S, Huang Y, Coman D, Hyder F. Characterization of a lanthanide complex encapsulated with MRI contrast agents into liposomes for biosensor imaging of redundant deviation in shifts (BIRDS). J Biol Inorg Chem 2014; 19:1385-98. [PMID: 25304046 DOI: 10.1007/s00775-014-1200-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 09/23/2014] [Indexed: 12/24/2022]
Abstract
Purposely designed magnetic resonance imaging (MRI) probes encapsulated in liposomes, which alter contrast by their paramagnetic effect on longitudinal (T₁) and transverse (T₂) relaxation times of tissue water, hold promise for molecular imaging. However, a challenge with liposomal MRI probes that are solely dependent on enhancement of water relaxation is lack of specific molecular readouts, especially in strong paramagnetic environments, thereby reducing the potential for monitoring disease treatment (e.g., cancer) beyond the generated MRI contrast. Previously, it has been shown that molecular imaging with magnetic resonance is also possible by detecting the signal of non-exchangeable protons emanating from paramagnetic lanthanide complexes themselves [e.g., TmDOTP⁵⁻, which is a Tm³⁺ -containing biosensor based on a macrocyclic chelate 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetrakis(methylene phosphonate), DOTP⁵⁻] with a method called biosensor imaging of redundant deviation in shifts (BIRDS). Here, we show that BIRDS is useful for molecular imaging with probes like TmDOTP⁵⁻ even when they are encapsulated inside liposomes with ultrastrong T₁and T₂contrast agents (e.g., Magnevist and Molday ION, respectively). We demonstrate that molecular readouts such as pH and temperature determined from probes like TmDOTP⁵⁻ are resilient, because the sensitivity of the chemical shifts to the probe's environment is not compromised by the presence of other paramagnetic agents contained within the same nanocarrier milieu. Because high liposomal encapsulation efficiency allows for robust MRI contrast and signal amplification for BIRDS, nanoengineered liposomal probes containing both monomers, TmDOTP⁵⁻ and paramagnetic contrast agents, could allow high spatial resolution imaging of disease diagnosis (with MRI) and status monitoring (with BIRDS).
Collapse
Affiliation(s)
- Samuel Maritim
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA,
| | | | | | | |
Collapse
|
43
|
Mudaliar H, Pollock C, Ma J, Wu H, Chadban S, Panchapakesan U. The role of TLR2 and 4-mediated inflammatory pathways in endothelial cells exposed to high glucose. PLoS One 2014; 9:e108844. [PMID: 25303153 PMCID: PMC4193767 DOI: 10.1371/journal.pone.0108844] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/05/2014] [Indexed: 12/22/2022] Open
Abstract
Postprandial hyperglycemia induces inflammation and endothelial dysfunction resulting in vascular complications in patients with diabetes. Toll-like receptors (TLRs) are central to the regulation of inflammatory responses through activation of nuclear factor-kappa B (NF-ĸB). This study examined the role of TLR2 and 4 in regulating inflammation and endothelial dysfunction when exposed to fluctuating glucose concentrations. HMEC-1 cells (a human microvascular endothelial cell line) were exposed to control (5 mM), 30 mM (high), fluctuating (5/30 mM) and 11.2 mM glucose (approximate glycaemic criteria for the diagnosis of diabetes mellitus) for 72 h. Cells were assessed for TLR2, 4, high mobility group box -1 (HMGB1), NF-ĸB, monocyte chemoattractant protein-1 (MCP-1), interleukin-8 (IL-8), intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). Fluctuating glucose concentrations maximally upregulated TLR4 but not TLR2 expression with increased NF-ĸB activation, IL-8 and ICAM-1 expression. HMGB1 was increased in the supernatants of cells exposed to 30 mM and 11.2 mM glucose compared to control. The addition of recombinant HMGB1 induced NF-ĸB activation and synthesis of proinflammatory cytokines and chemokines, which were prevented by TLR2 or 4 signalling inhibition. An additive effect when both TLR2 and 4 signalling pathways were inhibited was observed. However, only inhibition of TLR4 signalling suppressed the synthesis of MCP-1, IL-8 and ICAM-1. In vivo, streptozotocin-induced diabetic mice exhibited an increase in glomerular ICAM-1 which was not evident in TLR2-/- or TLR4-/- diabetic mice. Collectively, our results suggest that targeting the signalling pathway of TLR2 and 4 may be of therapeutic benefit in attenuating vascular inflammation in diabetic microangiopathy.
Collapse
Affiliation(s)
- Harshini Mudaliar
- Renal Research Group, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Carol Pollock
- Renal Research Group, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Jin Ma
- Renal Medicine Royal Prince Alfred Hospital and Collaborative Transplant Research Group, University of Sydney, Camperdown, New South Wales, Australia
| | - Huiling Wu
- Renal Medicine Royal Prince Alfred Hospital and Collaborative Transplant Research Group, University of Sydney, Camperdown, New South Wales, Australia
| | - Steven Chadban
- Renal Medicine Royal Prince Alfred Hospital and Collaborative Transplant Research Group, University of Sydney, Camperdown, New South Wales, Australia
| | - Usha Panchapakesan
- Renal Research Group, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- * E-mail:
| |
Collapse
|
44
|
Toy R, Bauer L, Hoimes C, Ghaghada KB, Karathanasis E. Targeted nanotechnology for cancer imaging. Adv Drug Deliv Rev 2014; 76:79-97. [PMID: 25116445 PMCID: PMC4169743 DOI: 10.1016/j.addr.2014.08.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/26/2014] [Accepted: 08/04/2014] [Indexed: 02/02/2023]
Abstract
Targeted nanoparticle imaging agents provide many benefits and new opportunities to facilitate accurate diagnosis of cancer and significantly impact patient outcome. Due to the highly engineerable nature of nanotechnology, targeted nanoparticles exhibit significant advantages including increased contrast sensitivity, binding avidity and targeting specificity. Considering the various nanoparticle designs and their adjustable ability to target a specific site and generate detectable signals, nanoparticles can be optimally designed in terms of biophysical interactions (i.e., intravascular and interstitial transport) and biochemical interactions (i.e., targeting avidity towards cancer-related biomarkers) for site-specific detection of very distinct microenvironments. This review seeks to illustrate that the design of a nanoparticle dictates its in vivo journey and targeting of hard-to-reach cancer sites, facilitating early and accurate diagnosis and interrogation of the most aggressive forms of cancer. We will report various targeted nanoparticles for cancer imaging using X-ray computed tomography, ultrasound, magnetic resonance imaging, nuclear imaging and optical imaging. Finally, to realize the full potential of targeted nanotechnology for cancer imaging, we will describe the challenges and opportunities for the clinical translation and widespread adaptation of targeted nanoparticles imaging agents.
Collapse
Affiliation(s)
- Randall Toy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lisa Bauer
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Physics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Christopher Hoimes
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Ketan B Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX 77030, USA; Department of Radiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
45
|
Bennett KM, Jo JI, Cabral H, Bakalova R, Aoki I. MR imaging techniques for nano-pathophysiology and theranostics. Adv Drug Deliv Rev 2014; 74:75-94. [PMID: 24787226 DOI: 10.1016/j.addr.2014.04.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 03/02/2014] [Accepted: 04/20/2014] [Indexed: 11/25/2022]
Abstract
The advent of nanoparticle DDSs (drug delivery systems, nano-DDSs) is opening new pathways to understanding physiology and pathophysiology at the nanometer scale. A nano-DDS can be used to deliver higher local concentrations of drugs to a target region and magnify therapeutic effects. However, interstitial cells or fibrosis in intractable tumors, as occurs in pancreatic or scirrhous stomach cancer, tend to impede nanoparticle delivery. Thus, it is critical to optimize the type and size of nanoparticles to reach the target. High-resolution 3D imaging provides a means of "seeing" the nanoparticle distribution and therapeutic effects. We introduce the concept of "nano-pathophysiological imaging" as a strategy for theranostics. The strategy consists of selecting an appropriate nano-DDS and rapidly evaluating drug effects in vivo to guide the next round of therapy. In this article we classify nano-DDSs by component carrier materials and present an overview of the significance of nano-pathophysiological MRI.
Collapse
|
46
|
Gharib A, Faezizadeh Z, Mesbah-Namin SAR, Saravani R. Preparation, characterization and in vitro efficacy of magnetic nanoliposomes containing the artemisinin and transferrin. ACTA ACUST UNITED AC 2014; 22:44. [PMID: 24887240 PMCID: PMC4053270 DOI: 10.1186/2008-2231-22-44] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 05/19/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Artemisinin is the major sesquiterpene lactones in sweet wormwood (Artemisia annua L.), and its combination with transferrin exhibits versatile anti-cancer activities. Their non-selective targeting for cancer cells, however, limits their application. The aim of this study was to prepare the artemisinin and transferrin-loaded magnetic nanoliposomes in thermosensitive and non-thermosensitive forms and evaluate their antiproliferative activity against MCF-7 and MDA-MB-231 cells for better tumor-targeted therapy. METHODS Artemisinin and transferrin-loaded magnetic nanoliposomes was prepared by extrusion method using various concentrations of lipids. These formulations were characterized for particle size, zeta potential, polydispersity index and shape morphology. The artemisinin and transferrin-loading efficiencies were determined using HPLC. The content of magnetic iron oxide in the nanoliposomes was analysed by spectrophotometry. The in vitro release of artemisinin, transferrin and magnetic iron oxide from vesicles was assessed by keeping of the nanoliposomes at 37°C for 12 h. The in vitro cytotoxicity of prepared nanoliposomes was investigated against MCF-7 and MDA-MB-231 cells using MTT assay. RESULTS The entrapment efficiencies of artemisinin, transferrin and magnetic iron oxide in the non-thermosensitive nanoliposomes were 89.11% ± 0.23, 85.09% ± 0.31 and 78.10% ± 0.24, respectively. Moreover, the thermosensitive formulation showed a suitable condition for thermal drug release at 42°C and exhibited high antiproliferative activity against MCF-7 and MDA-MB-231 cells in the presence of a magnetic field. CONCLUSIONS Our results showed that the thermosensitive artemisinin and transferrin-loaded magnetic nanoliposomes would be an effective choice for tumor-targeted therapy, due to its suitable stability and high effectiveness.
Collapse
Affiliation(s)
- Amir Gharib
- Department of Laboratory Sciences, Borujerd Branch, Islamic Azad University, Borujerd, Iran.
| | | | | | | |
Collapse
|
47
|
Nanomedicine-based strategies for treatment of atherosclerosis. Trends Mol Med 2014; 20:271-81. [PMID: 24594264 DOI: 10.1016/j.molmed.2013.12.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/02/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall that arises from an imbalanced lipid metabolism and a maladaptive inflammatory response. Despite intensive research on mechanisms underlying atherosclerotic lesion formation and progression during the past decade, translation of this knowledge into the clinic is scarce. Although developments have primarily been made in the area of antitumor therapy, recent advances have shown the potential of nanomedicine-based treatment strategies for atherosclerosis. Here we describe the features of currently available nanomedical formulations that have been optimized for atherosclerosis treatment, and we further describe how they can be instructed to target inflammatory processes in the arterial wall. Despite their limitations, nanomedical applications might hold promise for personalized medicine, and further efforts are needed to improve atherosclerosis-specific targeting.
Collapse
|
48
|
Deddens LH, van Tilborg GAF, van der Toorn A, van der Marel K, Paulis LEM, van Bloois L, Storm G, Strijkers GJ, Mulder WJM, de Vries HE, Dijkhuizen RM. MRI of ICAM-1 upregulation after stroke: the importance of choosing the appropriate target-specific particulate contrast agent. Mol Imaging Biol 2014; 15:411-22. [PMID: 23400400 DOI: 10.1007/s11307-013-0617-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Magnetic resonance imaging (MRI) with targeted contrast agents provides a promising means for diagnosis and treatment monitoring after cerebrovascular injury. Our goal was to demonstrate the feasibility of this approach to detect the neuroinflammatory biomarker intercellular adhesion molecule-1 (ICAM-1) after stroke and to establish a most efficient imaging procedure. PROCEDURES We compared two types of ICAM-1-functionalized contrast agent: T 1-shortening gadolinium chelate-containing liposomes and T2(*)-shortening micron-sized iron oxide particles (MPIO). Binding efficacy and MRI contrast effects were tested in cell cultures and a mouse stroke model. RESULTS Both ICAM-1-targeted agents bound effectively to activated cerebrovascular cells in vitro, generating significant MRI contrast-enhancing effects. Direct in vivo MRI-based detection after stroke was only achieved with ICAM-1-targeted MPIO, although both contrast agents showed similar target-specific vascular accumulation. CONCLUSIONS Our study demonstrates the potential of in vivo MRI of post-stroke ICAM-1 upregulation and signifies target-specific MPIO as most suitable contrast agent for molecular MRI of cerebrovascular inflammation.
Collapse
Affiliation(s)
- Lisette H Deddens
- Biomedical MR Imaging and Spectroscopy Group, Image Sciences Institute, University Medical Center Utrecht, Yalelaan 2, 3584 CM, Utrecht, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hosta-Rigau L, Schattling P, Teo BM, Lynge ME, Städler B. Recent progress of liposomes in nanomedicine. J Mater Chem B 2014; 2:6686-6691. [DOI: 10.1039/c4tb00825a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Liposome formulations are highlighted focusing on their chemical modification, interaction with cells, and use in substrate-mediated drug delivery and cell mimicry.
Collapse
Affiliation(s)
- Leticia Hosta-Rigau
- Interdisciplinary Nanoscience Centre (iNANO)
- Aarhus University
- Aarhus, Denmark
| | - Philipp Schattling
- Interdisciplinary Nanoscience Centre (iNANO)
- Aarhus University
- Aarhus, Denmark
| | - Boon M. Teo
- Interdisciplinary Nanoscience Centre (iNANO)
- Aarhus University
- Aarhus, Denmark
| | - Martin E. Lynge
- Interdisciplinary Nanoscience Centre (iNANO)
- Aarhus University
- Aarhus, Denmark
| | - Brigitte Städler
- Interdisciplinary Nanoscience Centre (iNANO)
- Aarhus University
- Aarhus, Denmark
| |
Collapse
|
50
|
Serres S, O'Brien ER, Sibson NR. Imaging angiogenesis, inflammation, and metastasis in the tumor microenvironment with magnetic resonance imaging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:263-83. [PMID: 24272363 DOI: 10.1007/978-1-4614-5915-6_12] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
With the development of new imaging techniques, the potential for probing the molecular, cellular, and structural components of the tumor microenvironment in situ has increased dramatically. A multitude of imaging modalities have been successfully employed to probe different aspects of the tumor microenvironment, including expression of molecules, cell motion, cellularity, vessel permeability, vascular perfusion, metabolic and physiological changes, apoptosis, and inflammation. This chapter focuses on the most recent advances in magnetic resonance imaging methods, which offer a number of advantages over other methodologies, including high spatial resolution and the use of nonionizing radiation, as well as the use of such methods in the context of primary and secondary brain tumors. It also highlights how they can be used to assess the molecular and cellular changes in the tumor microenvironment in response to therapy.
Collapse
Affiliation(s)
- Sébastien Serres
- CR-UK/MRC Gray Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, UK,
| | | | | |
Collapse
|