1
|
Shi Z, Huang J, Chen C, Zhang X, Ma Z, Liu Q. Lipid nanoparticles encapsulating curcumin for imaging and stabilization of vulnerable atherosclerotic plaques via phagocytic "eat-me" signals. J Control Release 2024; 373:265-276. [PMID: 39019087 DOI: 10.1016/j.jconrel.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Curcumin potentiates the stabilization of atherosclerotic plaques by polarizing macrophages, but its non-specific targeting hinders its clinical application. We aim to harness multifunctional lipid nanoparticles (MLNPs) to facilitate the imaging and targeted delivery of curcumin specifically to inflammatory macrophages, counteracting vulnerable plaques and mitigating the risk of ischemic events. Cholesteryl-9-carboxynonanoate-(125I‑iron oxide nanoparticle/Curcumin)-lipid-coated nanoparticles [9-CCN-(125I-ION/Cur)-LNPs], namely MLNPs, are designed to carry hybrid imaging agents. These agents combine 125I-ION with lipids containing phagocytic 'eat-me' signals, inducing macrophages to engulf the MLNPs. Our research demonstrates that the designed MLNPs accurately accumulate at unstable plaques and are precisely visualized and highlighted by both SPECT and MRI. Furthermore, MLNPs achieve high efficiency in delivering 125I-ION and curcumin to macrophages, ultimately leading to significant M1-to-M2 macrophage polarization. These real-time imaging and polarization capabilities of plaques have immediate clinical applicability and may pave the way for novel therapies to stabilize unstable atherosclerotic plaques.
Collapse
Affiliation(s)
- Zhang Shi
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China; Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Huang
- Department of Radiology, The Second Naval Hospital of Southern Theater Command of PLA, Sanya, China
| | - Chao Chen
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xuefeng Zhang
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhiqiang Ma
- Pharmacy School, Naval Medical University, Shanghai, China.
| | - Qi Liu
- Department of Radiology, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
2
|
Gawne PJ, Ferreira M, Papaluca M, Grimm J, Decuzzi P. New Opportunities and Old Challenges in the Clinical translation of Nanotheranostics. NATURE REVIEWS. MATERIALS 2023; 8:783-798. [PMID: 39022623 PMCID: PMC11251001 DOI: 10.1038/s41578-023-00581-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/20/2024]
Abstract
Nanoparticle-based systems imbued with both diagnostic and therapeutic functions, known as nanotheranostics, have enabled remarkable progress in guiding focal therapy, inducing active responses to endogenous and exogenous biophysical stimuli, and stratifying patients for optimal treatment. However, although in recent years more nanotechnological platforms and techniques have been implemented in the clinic, several important challenges remain that are specific to nanotheranostics. In this Review, we first discuss some of the many ways of 'constructing' nanotheranostics, focusing on the different imaging modalities and therapeutic strategies. We then outline nanotheranostics that are currently used in humans at different stages of clinical development, identifying specific advantages and opportunities. Finally, we define critical steps along the winding road of preclinical and clinical development and suggest actions to overcome technical, manufacturing, regulatory and economical challenges for the safe and effective clinical translation of nanotheranostics.
Collapse
Affiliation(s)
- Peter J. Gawne
- UCL Cancer Institute, University College London, London, UK
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary, University of London, London, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Miguel Ferreira
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Marisa Papaluca
- School of Public Health, Imperial College of London, South Kensington CampusLondon, UK
| | - Jan Grimm
- Molecular Pharmacology Program and Department of Radiology, Memorial Sloan-Kettering Cancer, Center, New York, NY, USA
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via, Morego 30, 16163, Genoa, IT
| |
Collapse
|
3
|
Xu S, Gu Z, Lu H, Guan P, Liu Z. Leveraging Macrophage-Mediated Cancer Immunotherapy via a Cascading Effect Induced by a Molecularly Imprinted Nanocoordinator. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37267068 DOI: 10.1021/acsami.3c03950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Reprogramming tumor-associated macrophages (TAMs) has emerged as a promising strategy in cancer immunotherapy. Targeted therapeutics integrating multiple functions to fully leverage the antitumor immune functions of macrophages without affecting systemic or tissue-resident macrophages are crucial for TAM reprogramming. Herein, by integrating molecular imprinting and nanotechnology, we rationally designed and engineered an unprecedented nanocoordinator for targeted remolding of TAMs to fully leverage the antitumor efficacy of macrophages by inducing a cascade effect. The nanocoordinator features a magnetic iron oxide nanoinner core and sialic acid-imprinted shell. Intravenously administered into systemic circulation, the nanocoordinator can rapidly accumulate at the tumor site in response to an external magnet. Then, by specifically binding to sialic acid overexpressed on tumor cells, the nanocoordinator anchors at the tumor site with prolonged retention time. Via binding with the nanocoordinator, tumor cells are tagged with a foreign substance, which promotes the intrinsic phagocytosis of macrophages. Subsequently, the nanocoordinator taken up by macrophages effectively promotes the polarization of macrophages toward the M1 phenotype, thus activating the immunotherapeutic efficacy of macrophages. Synergized by the cascade effect, this nanocoordinator effectively harnesses TAMs for macrophage-mediated immunotherapy. This study offers new TAM-targeted therapeutics that allows us to fully leverage the antitumor immune functions of macrophages without affecting the normal tissue.
Collapse
Affiliation(s)
- Shuxin Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zikuan Gu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Haifeng Lu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Peixin Guan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| |
Collapse
|
4
|
Unterweger H, Janko C, Folk T, Cicha I, Kovács N, Gyebnár G, Horváth I, Máthé D, Zheng KH, Coolen BF, Stroes E, Szebeni J, Alexiou C, Dézsi L, Lyer S. Comparative in vitro and in vivo Evaluation of Different Iron Oxide-Based Contrast Agents to Promote Clinical Translation in Compliance with Patient Safety. Int J Nanomedicine 2023; 18:2071-2086. [PMID: 37113796 PMCID: PMC10128873 DOI: 10.2147/ijn.s402320] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/14/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction One of the major challenges in the clinical translation of nanoparticles is the development of formulations combining favorable efficacy and optimal safety. In the past, iron oxide nanoparticles have been introduced as an alternative for gadolinium-containing contrast agents; however, candidates available at the time were not free from adverse effects. Methods Following the development of a potent iron oxide-based contrast agent SPIONDex, we now performed a systematic comparison of this formulation with the conventional contrast agent ferucarbotran and with ferumoxytol, taking into consideration their physicochemical characteristics, bio- and hemocompatibility in vitro and in vivo, as well as their liver imaging properties in rats. Results The results demonstrated superior in vitro cyto-, hemo- and immunocompatibility of SPIONDex in comparison to the other two formulations. Intravenous administration of ferucarbotran or ferumoxytol induced strong complement activation-related pseudoallergy in pigs. In contrast, SPIONDex did not elicit any hypersensitivity reactions in the experimental animals. In a rat model, comparable liver imaging properties, but a faster clearance was demonstrated for SPIONDex. Conclusion The results indicate that SPIONDex possess an exceptional safety compared to the other two formulations, making them a promising candidate for further clinical translation.
Collapse
Affiliation(s)
- Harald Unterweger
- ENT-Department, Section of Experimental Oncology und Nanomedicine (SEON), Universitätsklinikum Erlangen, Erlangen, Germany
- Correspondence: Harald Unterweger, Universitätsklinikum Erlangen, Glueckstr. 10a, Erlangen, 91054, Germany, Tel +49 9131 85-33142, Fax +49 9131 85-34828, Email
| | - Christina Janko
- ENT-Department, Section of Experimental Oncology und Nanomedicine (SEON), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tamara Folk
- ENT-Department, Section of Experimental Oncology und Nanomedicine (SEON), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iwona Cicha
- ENT-Department, Section of Experimental Oncology und Nanomedicine (SEON), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Noémi Kovács
- Hungarian Centre of Excellence for Molecular Medicine, Semmelweis University, Budapest, Hungary
| | - Gyula Gyebnár
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Ildikó Horváth
- Department Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Hungarian Centre of Excellence for Molecular Medicine, Semmelweis University, Budapest, Hungary
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Kang H Zheng
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Bram F Coolen
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Erik Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience Ltd, Budapest, Hungary
| | - Christoph Alexiou
- ENT-Department, Section of Experimental Oncology und Nanomedicine (SEON), Universitätsklinikum Erlangen, Erlangen, Germany
| | - László Dézsi
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience Ltd, Budapest, Hungary
| | - Stefan Lyer
- ENT-Department, Section of Experimental Oncology und Nanomedicine (SEON), Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
5
|
Zschiesche L, Janko C, Friedrich B, Frey B, Band J, Lyer S, Alexiou C, Unterweger H. Biocompatibility of Dextran-Coated 30 nm and 80 nm Sized SPIONs towards Monocytes, Dendritic Cells and Lymphocytes. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 13:14. [PMID: 36615924 PMCID: PMC9823599 DOI: 10.3390/nano13010014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
Dextran-coated superparamagnetic iron oxide nanoparticles (SPIONDex) of various sizes can be used as contrast agents in magnetic resonance imaging (MRI) of different tissues, e.g., liver or atherosclerotic plaques, after intravenous injection. In previous studies, the blood compatibility and the absence of immunogenicity of SPIONDex was demonstrated. The investigation of the interference of SPIONDex with stimulated immune cell activation is the aim of this study. For this purpose, sterile and endotoxin-free SPIONDex with different hydrodynamic sizes (30 and 80 nm) were investigated for their effect on monocytes, dendritic cells (DC) and lymphocytes in concentrations up to 200 µg/mL, which would be administered for use as an imaging agent. The cells were analyzed using flow cytometry and brightfield microscopy. We found that SPIONDex were hardly taken up by THP-1 monocytes and did not reduce cell viability. In the presence of SPIONDex, the phagocytosis of zymosan and E. coli by THP-1 was dose-dependently reduced. SPIONDex neither induced the maturation of DCs nor interfered with their stimulated maturation. The particles did not induce lymphocyte proliferation or interfere with lymphocyte proliferation after stimulation. Since SPIONDex rapidly distribute via the blood circulation in vivo, high concentrations were only reached locally at the injection site immediately after application and only for a very limited time. Thus, SPIONDex can be considered immune compatible in doses required for use as an MRI contrast agent.
Collapse
Affiliation(s)
- Lisa Zschiesche
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Julia Band
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Controlled Nanomaterials, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), Professorship for AI-Controlled Nanomaterials, Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
6
|
Emerging trends in the nanomedicine applications of functionalized magnetic nanoparticles as novel therapies for acute and chronic diseases. J Nanobiotechnology 2022; 20:393. [PMID: 36045375 PMCID: PMC9428876 DOI: 10.1186/s12951-022-01595-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
High-quality point-of-care is critical for timely decision of disease diagnosis and healthcare management. In this regard, biosensors have revolutionized the field of rapid testing and screening, however, are confounded by several technical challenges including material cost, half-life, stability, site-specific targeting, analytes specificity, and detection sensitivity that affect the overall diagnostic potential and therapeutic profile. Despite their advances in point-of-care testing, very few classical biosensors have proven effective and commercially viable in situations of healthcare emergency including the recent COVID-19 pandemic. To overcome these challenges functionalized magnetic nanoparticles (MNPs) have emerged as key players in advancing the biomedical and healthcare sector with promising applications during the ongoing healthcare crises. This critical review focus on understanding recent developments in theranostic applications of functionalized magnetic nanoparticles (MNPs). Given the profound global economic and health burden, we discuss the therapeutic impact of functionalized MNPs in acute and chronic diseases like small RNA therapeutics, vascular diseases, neurological disorders, and cancer, as well as for COVID-19 testing. Lastly, we culminate with a futuristic perspective on the scope of this field and provide an insight into the emerging opportunities whose impact is anticipated to disrupt the healthcare industry.
Collapse
|
7
|
Liu J, Liu Z, Pang Y, Zhou H. The interaction between nanoparticles and immune system: application in the treatment of inflammatory diseases. J Nanobiotechnology 2022; 20:127. [PMID: 35279135 PMCID: PMC8917374 DOI: 10.1186/s12951-022-01343-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Nanoparticle (NP) is an emerging tool applied in the biomedical field. With combination of different materials and adjustment of their physical and chemical properties, nanoparticles can have diverse effects on the organism and may change the treating paradigm of multiple diseases in the future. More and more results show that nanoparticles can function as immunomodulators and some formulas have been approved for the treatment of inflammation-related diseases. However, our current understanding of the mechanisms that nanoparticles can influence immune responses is still limited, and systemic clinical trials are necessary for the evaluation of their security and long-term effects. This review provides an overview of the recent advances in nanoparticles that can interact with different cellular and molecular components of the immune system and their application in the management of inflammatory diseases, which are caused by abnormal immune reactions. This article focuses on the mechanisms of interaction between nanoparticles and the immune system and tries to provide a reference for the future design of nanotechnology for the treatment of inflammatory diseases.
Collapse
|
8
|
Rahiman N, Mohammadi M, Alavizadeh SH, Arabi L, Badiee A, Jaafari MR. Recent advancements in nanoparticle-mediated approaches for restoration of multiple sclerosis. J Control Release 2022; 343:620-644. [PMID: 35176392 DOI: 10.1016/j.jconrel.2022.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022]
Abstract
Multiple Sclerosis (MS) is an autoimmune disease with complicated immunopathology which necessitates considering multifactorial aspects for its management. Nano-sized pharmaceutical carriers named nanoparticles (NPs) can support impressive management of disease not only in early detection and prognosis level but also in a therapeutic manner. The most prominent initiator of MS is the domination of cellular immunity to humoral immunity and increment of inflammatory cytokines. The administration of several platforms of NPs for MS management holds great promise so far. The efforts for MS management through in vitro and in vivo (experimental animal models) evaluations, pave a new way to a highly efficient therapeutic means and aiding its translation to the clinic in the near future.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of pharmaceutics, School of pharmacy, Mashhad University of Medical sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Abstract
Mesenchymal stem cells (MSCs) exhibit regenerative and reparative properties. However, most MSC-related studies remain to be translated for regular clinical usage, partly due to challenges in pre-transplantation cell labelling and post-transplantation cell tracking. Amidst this, there are growing concerns over the toxicity of commonly used gadolinium-based contrast agents that mediate in-vivo cell detection via MRI. This urges to search for equally effective but less toxic alternatives that would facilitate and enhance MSC detection post-administration and provide therapeutic benefits in-vivo. MSCs labelled with iron oxide nanoparticles (IONPs) have shown promising results in-vitro and in-vivo. Thus, it would be useful to revisit these studies before inventing new labelling approaches. Aiming to inform regenerative medicine and augment clinical applications of IONP-labelled MSCs, this review collates and critically evaluates the utility of IONPs in enhancing MSC detection and therapeutics. It explains the rationale, principle, and advantages of labelling MSCs with IONPs, and describes IONP-induced intracellular alterations and consequent cellular manifestations. By exemplifying clinical pathologies, it examines contextual in-vitro, animal, and clinical studies that used IONP-labelled bone marrow-, umbilical cord-, adipose tissue- and dental pulp-derived MSCs. It compiles and discusses studies involving MSC-labelling of IONPs in combinations with carbohydrates (Venofer, ferumoxytol, dextran, glucosamine), non-carbohydrate polymers [poly(L-lysine), poly(lactide-co-glycolide), poly(L-lactide), polydopamine], elements (ruthenium, selenium, gold, zinc), compounds/stains (silica, polyethylene glycol, fluorophore, rhodamine B, DAPI, Prussian blue), DNA, Fibroblast growth Factor-2 and the drug doxorubicin. Furthermore, IONP-labelling of MSC exosomes is reviewed. Also, limitations of IONP-labelling are addressed and methods of tackling those challenges are suggested.
Collapse
|
10
|
Helfer BM, Ponomarev V, Patrick PS, Blower PJ, Feitel A, Fruhwirth GO, Jackman S, Pereira Mouriès L, Park MVDZ, Srinivas M, Stuckey DJ, Thu MS, van den Hoorn T, Herberts CA, Shingleton WD. Options for imaging cellular therapeutics in vivo: a multi-stakeholder perspective. Cytotherapy 2021; 23:757-773. [PMID: 33832818 PMCID: PMC9344904 DOI: 10.1016/j.jcyt.2021.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/01/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Cell-based therapies have been making great advances toward clinical reality. Despite the increase in trial activity, few therapies have successfully navigated late-phase clinical trials and received market authorization. One possible explanation for this is that additional tools and technologies to enable their development have only recently become available. To support the safety evaluation of cell therapies, the Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee, a multisector collaborative committee, polled the attendees of the 2017 International Society for Cell & Gene Therapy conference in London, UK, to understand the gaps and needs that cell therapy developers have encountered regarding safety evaluations in vivo. The goal of the survey was to collect information to inform stakeholders of areas of interest that can help ensure the safe use of cellular therapeutics in the clinic. This review is a response to the cellular imaging interests of those respondents. The authors offer a brief overview of available technologies and then highlight the areas of interest from the survey by describing how imaging technologies can meet those needs. The areas of interest include imaging of cells over time, sensitivity of imaging modalities, ability to quantify cells, imaging cellular survival and differentiation and safety concerns around adding imaging agents to cellular therapy protocols. The Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee believes that the ability to understand therapeutic cell fate is vital for determining and understanding cell therapy efficacy and safety and offers this review to aid in those needs. An aim of this article is to share the available imaging technologies with the cell therapy community to demonstrate how these technologies can accomplish unmet needs throughout the translational process and strengthen the understanding of cellular therapeutics.
Collapse
Affiliation(s)
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - P Stephen Patrick
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Alexandra Feitel
- Formerly, Health and Environmental Sciences Institute, US Environmental Protection Agency, Washington, DC, USA
| | - Gilbert O Fruhwirth
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Shawna Jackman
- Charles River Laboratories, Shrewsbury, Massachusetts, USA
| | | | - Margriet V D Z Park
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, the Netherlands; Cenya Imaging BV, Amsterdam, the Netherlands
| | - Daniel J Stuckey
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Mya S Thu
- Visicell Medical Inc, La Jolla, California, USA
| | | | | | | |
Collapse
|
11
|
Glover JC, Aswendt M, Boulland JL, Lojk J, Stamenković S, Andjus P, Fiori F, Hoehn M, Mitrecic D, Pavlin M, Cavalli S, Frati C, Quaini F. In vivo Cell Tracking Using Non-invasive Imaging of Iron Oxide-Based Particles with Particular Relevance for Stem Cell-Based Treatments of Neurological and Cardiac Disease. Mol Imaging Biol 2021; 22:1469-1488. [PMID: 31802361 DOI: 10.1007/s11307-019-01440-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell-based therapeutics is a rapidly developing field associated with a number of clinical challenges. One such challenge lies in the implementation of methods to track stem cells and stem cell-derived cells in experimental animal models and in the living patient. Here, we provide an overview of cell tracking in the context of cardiac and neurological disease, focusing on the use of iron oxide-based particles (IOPs) visualized in vivo using magnetic resonance imaging (MRI). We discuss the types of IOPs available for such tracking, their advantages and limitations, approaches for labeling cells with IOPs, biological interactions and effects of IOPs at the molecular and cellular levels, and MRI-based and associated approaches for in vivo and histological visualization. We conclude with reviews of the literature on IOP-based cell tracking in cardiac and neurological disease, covering both preclinical and clinical studies.
Collapse
Affiliation(s)
- Joel C Glover
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway. .,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway.
| | - Markus Aswendt
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Jean-Luc Boulland
- Laboratory for Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, PB 1105, Blindern, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
| | - Jasna Lojk
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia
| | - Stefan Stamenković
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Pavle Andjus
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, PB 52, 10001 Belgrade, Serbia
| | - Fabrizio Fiori
- Department of Applied Physics, Università Politecnica delle Marche - Di.S.C.O., Via Brecce Bianche, 60131, Ancona, Italy
| | - Mathias Hoehn
- Institut für Neurowissenschaften und Medizin, Forschungszentrum Jülich, Leo-Brandt-Str. 5, 52425, Jülich, Germany
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mojca Pavlin
- Group for Nano and Biotechnological Applications, Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, Ljubljana, Slovenia.,Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Stefano Cavalli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | |
Collapse
|
12
|
The effect of magnetic guiding BMSCs on hypoxic-ischemic brain damage via magnetic resonance imaging evaluation. Magn Reson Imaging 2021; 79:59-65. [PMID: 33727146 DOI: 10.1016/j.mri.2021.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/16/2020] [Accepted: 03/10/2021] [Indexed: 11/20/2022]
Abstract
Hypoxic-ischemic brain damage (HIBD) is a critical disease in pediatric neurosurgery with high mortality rate and frequently leads to neurological sequelae. The role of bone marrow mesenchymal stem cells (BMSCs) in neuroprotection has been recognized. However, using the imaging methods to dynamically assess the neuroprotective effects of BMSCs is rarely reported. In this study, BMSCs were isolated, cultured and identified. Flow cytometry assay had shown the specific surface molecular markers of BMSCs, which indicated that the cultivated cells were purified BMSCs. The results demonstrated that CD29 and CD90 were highly expressed, whilst CD45 and CD11b were negatively expressed. Further, BMSCs were transplanted into Sprague Dawley (SD) rats established HIBD via three ways, including lateral ventricle (LV) injection, tail vein (TV) injection, and LV injection with magnetic guiding. Magnetic resonance imaging (MRI) was used to monitor and assess the treatment effect of super paramagnetic iron oxide (SPIO)-labeled BMSCs. The mean kurtosis (MK) values from diffusion kurtosis imaging (DKI) exhibited the significant differences. It was found that the MK value of HIBD group increased compared with that in Sham. At the meantime, the MK values of LV + HIBD, TV + HIBD and Magnetic+LV + HIBD groups decreased compared with that in HIBD group. Among these, the MK value reduced most significantly in Magnetic+LV + HIBD group. MRI illustrated that the treatment effect of Magnetic+LV + HIBD group was best. In addition, HE staining and TUNEL assay measured the pathological changes and apoptosis of brain tissues, which further verified the MRI results. All data suggest that magnetic guiding BMSCs, a targeted delivery way, is a new strategic theory for HIBD treatment. The DKI technology of MRI can dynamically evaluate the neuroprotective effects of transplanted BMSCs in HIBD.
Collapse
|
13
|
Sheng J, Shi C, Gu N. Clinical trials of MRI-based immune cell imaging: challenges and perspectives. Sci Bull (Beijing) 2021; 66:303-306. [PMID: 36654405 DOI: 10.1016/j.scib.2020.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Jingyi Sheng
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215213, China
| | - Chu Shi
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215213, China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China; Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou 215213, China.
| |
Collapse
|
14
|
Helfer BM, Bulte JW. Cell Surveillance Using Magnetic Resonance Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00042-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
15
|
Vakili-Ghartavol R, Momtazi-Borojeni AA, Vakili-Ghartavol Z, Aiyelabegan HT, Jaafari MR, Rezayat SM, Arbabi Bidgoli S. Toxicity assessment of superparamagnetic iron oxide nanoparticles in different tissues. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:443-451. [PMID: 32024389 DOI: 10.1080/21691401.2019.1709855] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) have been employed in several biomedical applications where they facilitate both diagnostic and therapeutic aims. Although the potential benefits of SPIONs with different surface chemistry and conjugated targeting ligands/proteins are considerable, complicated interactions between these nanoparticles (NPs) and cells leading to toxic impacts could limit their clinical applications. Hence, elevation of our knowledge regarding the SPION-related toxicity is necessary. Here, the present review article will consider current studies and compare the potential toxic effect of SPIONs with or without identical surface chemistries on different cell lines. It centers on cellular and molecular mechanisms underlying toxicity of SPIONs. Likewise, emphasis is being dedicated for toxicity of SPIONs in various cell lines, in vitro and animal models, in vivo.
Collapse
Affiliation(s)
- Roghayyeh Vakili-Ghartavol
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Abbas Momtazi-Borojeni
- Halal Research Center of IRI, FDA, Tehran, Iran.,Nanotechnology Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeynab Vakili-Ghartavol
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hammed Tanimowo Aiyelabegan
- Department of Medical Biochemistry and Pharmacology, College of Pure and Applied Sciences, Kwara State University Malete, Nigeria
| | - Mahmoud Reza Jaafari
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology-Pharmacology, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Sepideh Arbabi Bidgoli
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University (IAUPS), Tehran, Iran
| |
Collapse
|
16
|
Chrishtop VV, Mironov VA, Prilepskii AY, Nikonorova VG, Vinogradov VV. Organ-specific toxicity of magnetic iron oxide-based nanoparticles. Nanotoxicology 2020; 15:167-204. [PMID: 33216662 DOI: 10.1080/17435390.2020.1842934] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The unique properties of magnetic iron oxide nanoparticles determined their widespread use in medical applications, the food industry, textile industry, which in turn led to environmental pollution. These factors determine the long-term nature of the effect of iron oxide nanoparticles on the body. However, studies in the field of chronic nanotoxicology of magnetic iron particles are insufficient and scattered. Studies show that toxicity may be increased depending on oral and inhalation routes of administration rather than injection. The sensory nerve pathway can produce a number of specific effects not seen with other routes of administration. Organ systems showing potential toxic effects when injected with iron oxide nanoparticles include the nervous system, heart and lungs, the thyroid gland, and organs of the mononuclear phagocytic system (MPS). A special place is occupied by the reproductive system and the effect of nanoparticles on the health of the first and second generations of individuals exposed to the toxic effects of iron oxide nanoparticles. This knowledge should be taken into account for subsequent studies of the toxicity of iron oxide nanoparticles. Particular attention should be paid to tests conducted on animals with pathologies representing human chronic socially significant diseases. This part of preclinical studies is almost in its infancy but of great importance for further medical translation on nanomaterials to practice.
Collapse
Affiliation(s)
| | | | | | - Varvara G Nikonorova
- Ivanovo State Agricultural Academy named after D.K. Belyaev, Peterburg, Russian Federation
| | | |
Collapse
|
17
|
Miller MR, Poland CA. Nanotoxicology: The Need for a Human Touch? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2001516. [PMID: 32697439 DOI: 10.1002/smll.202001516] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/28/2020] [Indexed: 06/11/2023]
Abstract
With the ever-expanding number of manufactured nanomaterials (MNMs) under development there is a vital need for nanotoxicology studies that test the potential for MNMs to cause harm to health. An extensive body of work in cell cultures and animal models is vital to understanding the physicochemical characteristics of MNMs and the biological mechanisms that underlie any detrimental actions to cells and organs. In human subjects, exposure monitoring is combined with measurement of selected health parameters in small panel studies, especially in occupational settings. However, the availability of further in vivo human data would greatly assist the risk assessment of MNMs. Here, the potential for controlled inhalation exposures of MNMs in human subjects is discussed. Controlled exposures to carbon, gold, aluminum, and zinc nanoparticles in humans have already set a precedence to demonstrate the feasibility of this approach. These studies have provided considerable insight into the potential (or not) of nanoparticles to induce inflammation, alter lung function, affect the vasculature, reach the systemic circulation, and accumulate in other organs. The need for further controlled exposures of MNMs in human volunteers - to establish no-effect limits, biological mechanisms, and provide vital data for the risk assessment of MNMs - is advocated.
Collapse
Affiliation(s)
- Mark R Miller
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Craig A Poland
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
18
|
Sriramvenugopal M, Pacak CA. Noninvasive Tracking of Implanted Cells: Superparamagnetic Iron Oxide Nanoparticles as a Long-Term, Multimodal Imaging Label. Methods Mol Biol 2020; 2126:167-175. [PMID: 32112388 DOI: 10.1007/978-1-0716-0364-2_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Superparamagnetic iron oxide (SPIO) nanoparticles can function as specific, long-term multimodal contrast agents for noninvasive imaging studies. Here we describe how to achieve high-resolution, long-term, serial images of single-label transplanted cells through two complementary imaging techniques: magnetic resonance imaging (MRI) and microcomputed tomography (μCT).
Collapse
Affiliation(s)
| | - Christina A Pacak
- Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
19
|
Bio-application of Inorganic Nanomaterials in Tissue Engineering. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1249:115-130. [PMID: 32602094 DOI: 10.1007/978-981-15-3258-0_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inorganic nanomaterials or nanoparticles (INPs) have drawn high attention for their usage in the biomedical field. In addition to the facile synthetic and modifiable property of INPs, INPs have various unique properties that originate from the components of the INPs, such as metal ions that are essential for the human body. Apart from their roles as components of the human body, inorganic materials have unique properties, such as magnetic, antibacterial, and piezoelectric, so that INPs have been widely used as either carriers or inducers. However, most of the bio-applicable INPs, especially those consisting of metal, can cause cytotoxicity. Therefore, INPs require modification to alleviate the harmful effect toward the cells by controlling the release of metal ions from INPs. Even though many attempts have been made to modify INPs, many things, including the side effects of INPs, still remain as obstacles in the bio-application, which need to be elucidated. In this chapter, we introduce novel INPs in terms of their synthetic method and bio-application in tissue engineering.
Collapse
|
20
|
In Vivo MRI Tracking of Mesenchymal Stromal Cells Labeled with Ultrasmall Paramagnetic Iron Oxide Particles after Intramyocardial Transplantation in Patients with Chronic Ischemic Heart Disease. Stem Cells Int 2019; 2019:2754927. [PMID: 31814830 PMCID: PMC6877937 DOI: 10.1155/2019/2754927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 09/28/2019] [Indexed: 01/17/2023] Open
Abstract
Background While regenerative stem cell therapy for ischemic heart disease has moved into phase 3 studies, little is still known about retention and migration of cell posttransplantation. In human studies, the ability to track transplanted cells has been limited to labeling with radioisotopes and tracking using nuclear imaging. This method is limited by low resolution and short half-lives of available radioisotopes. Longitudinal tracking using magnetic resonance imaging (MRI) of myocardial injected cells labeled with iron oxide nanoparticles has shown promising results in numerous preclinical studies but has yet to be evaluated in human studies. We aimed to evaluate MRI tracking of mesenchymal stromal cells (MSCs) labeled with ultrasmall paramagnetic iron oxide (USPIO) nanoparticles after intramyocardial transplantation in patients with ischemic heart disease (IHD). Methods Five no-option patients with chronic symptomatic IHD underwent NOGA-guided intramyocardial transplantation of USPIO-labeled MSCs. Serial MRI scans were performed to track labeled cells both visually and using semiautomated T2∗ relaxation time analysis. For safety, we followed symptoms, quality of life, and myocardial function for 6 months. Results USPIO-labeled MSCs were tracked for up to 14 days after transplantation at injection sites both visually and using semiautomated regional T2∗ relaxation time analysis. Labeling of MSCs did not impair long-term safety of treatment. Conclusion This was a first-in-man clinical experience aimed at evaluating the utility of MRI tracking of USPIO-labeled bone marrow-derived autologous MSCs after intramyocardial injection in patients with chronic IHD. The treatment was safe, and cells were detectable at injection sites up to 14 days after transplantation. Further studies are needed to clarify if MSCs migrate out of the injection area into other areas of the myocardium or if injected cells are washed out into the peripheral circulation. The trial is registered with ClinicalTrials.gov NCT03651791.
Collapse
|
21
|
Zhang X, Wu B, Guo Z, Gao Y, Xi W, Yu H, Feng G, Zhang J, Shen W, Chen J. Determination of portal vein tumor thrombus blood supply using in vivo cellular magnetic resonance imaging in a rabbit model. Cancer Manag Res 2019; 11:5523-5529. [PMID: 31417305 PMCID: PMC6592059 DOI: 10.2147/cmar.s197231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/05/2019] [Indexed: 01/10/2023] Open
Abstract
Objective: This study aimed to investigate the anatomic configuration of the blood vessels that contribute to portal vein tumor thrombus (PVTT), a common complication of hepatocellular carcinoma, in VX2 rabbits. Materials and methods: Peripheral blood mononuclear cells (MNCs) were isolated and labeled using superparamagnetic iron oxide particles in vitro. Twenty-four rabbits were injected with the VX2 tumor via the portal vein to establish the PVTT model. The rabbits (n=6/treatment group) were randomly assigned into four groups. Rabbits of groups A, B and C received an infusion of iron-labeled MNCs via the hepatic artery, the portal vein or the auricular vein, respectively, whereas rabbits of group D received an injection of normal saline via the auricular vein 7 days after the injection of VX2 tumors. MRI was performed, and the signal intensity (SI) of the PVTTs was measured on T2-weighted images (T2WIs) 1 day after the transfusion of iron-labeled cells. Results: The SI of PVTTs, as measured on T2WIs, in rabbits of groups A, B, C and D was 241.400 (172.350, 364.825), 221.150 (203.775, 318.225), 590.200 (363.325, 728.875) and 568.050 (474.725, 705.150), respectively. Our data showed a significant decrease in the SI of PVTTs in rabbits of groups A and B compared with rabbits of groups C and D (group A vs group C, U=4.000, p=0.025; group A vs group D, U=2.000, p=0.010; group B vs group C, U=4.000, p=0.025; group B vs group D, U=1.000, p=0.006). There was no significant difference in the SI of PVTTs in rabbits of group A and B. Conclusion: Our results indicated that the portal vein and the hepatic artery supplied blood flow to the PVTT in rabbits.
Collapse
Affiliation(s)
- Xiuming Zhang
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Bei Wu
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Zhen Guo
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Yang Gao
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Wei Xi
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Hui Yu
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Guodong Feng
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Jingyuan Zhang
- Department of Pathology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Wenrong Shen
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| | - Jun Chen
- Department of Radiology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and Cancer Hospital of Nanjing Medical University (NMU), Nanjing 210009, People's Republic of China
| |
Collapse
|
22
|
Affiliation(s)
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh, Scotland
| |
Collapse
|
23
|
Bulte JWM, Daldrup-Link HE. Clinical Tracking of Cell Transfer and Cell Transplantation: Trials and Tribulations. Radiology 2018; 289:604-615. [PMID: 30299232 PMCID: PMC6276076 DOI: 10.1148/radiol.2018180449] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 12/29/2022]
Abstract
Cell therapy has provided unprecedented opportunities for tissue repair and cancer therapy. Imaging tools for in vivo tracking of therapeutic cells have entered the clinic to evaluate therapeutic cell delivery and retention in patients. Thus far, clinical cell tracking studies have been a mere proof of principle of the feasibility of cell detection. This review centers around the main clinical queries associated with cell therapy: Have cells been delivered correctly at the targeted site of injection? Are cells still alive, and, if so, how many? Are cells being rejected by the host, and, if so, how severe is the immune response? For stem cell therapeutics, have cells differentiated into downstream cell lineages? Is there cell proliferation including tumor formation? At present, clinical cell tracking trials have only provided information on immediate cell delivery and short-term cell retention. The next big question is if these cell tracking tools can improve the clinical management of the patients and, if so, by how much, for how many, and for whom; in addition, it must be determined whether tracking therapeutic cells in every patient is needed. To become clinically relevant, it must now be demonstrated how cell tracking techniques can inform patient treatment and affect clinical outcomes.
Collapse
Affiliation(s)
- Jeff W. M. Bulte
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Departments of Chemical & Biomolecular Engineering, Biomedical Engineering, and Oncology, The Johns Hopkins University School of Medicine, 217 Traylor Bldg, 720 Rutland Ave, Baltimore, MD 21205 (J.W.M.B.); and Departments of Radiology, Molecular Imaging Program at Stanford (MIPS) and Pediatrics, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, Calif (H.E.D.L.)
| | - Heike E. Daldrup-Link
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Departments of Chemical & Biomolecular Engineering, Biomedical Engineering, and Oncology, The Johns Hopkins University School of Medicine, 217 Traylor Bldg, 720 Rutland Ave, Baltimore, MD 21205 (J.W.M.B.); and Departments of Radiology, Molecular Imaging Program at Stanford (MIPS) and Pediatrics, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, Calif (H.E.D.L.)
| |
Collapse
|
24
|
Ojha S, Kumar B. A review on nanotechnology based innovations in diagnosis and treatment of multiple sclerosis. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.jocit.2017.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Matuszak J, Dörfler P, Lyer S, Unterweger H, Juenet M, Chauvierre C, Alaarg A, Franke D, Almer G, Texier I, Metselaar JM, Prassl R, Alexiou C, Mangge H, Letourneur D, Cicha I. Comparative analysis of nanosystems’ effects on human endothelial and monocytic cell functions. Nanotoxicology 2018; 12:957-974. [DOI: 10.1080/17435390.2018.1502375] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jasmin Matuszak
- Section of Experimental Oncology and Nanomedicine (SEON), ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Philipp Dörfler
- Section of Experimental Oncology and Nanomedicine (SEON), ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Stefan Lyer
- Section of Experimental Oncology and Nanomedicine (SEON), ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Harald Unterweger
- Section of Experimental Oncology and Nanomedicine (SEON), ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Maya Juenet
- INSERM, U1148, LVTS, Paris Diderot University, X Bichat Hospital, Paris, France
| | - Cédric Chauvierre
- INSERM, U1148, LVTS, Paris Diderot University, X Bichat Hospital, Paris, France
| | - Amr Alaarg
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | - Gunter Almer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Isabelle Texier
- Grenoble Alpes Université, CEA-LETI MINATEC Campus, Grenoble, France
| | - Josbert M. Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
- Department of Experimental Molecular Imaging, RWTH University Clinic Aachen, Aachen, Germany
| | - Ruth Prassl
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - Christoph Alexiou
- Section of Experimental Oncology and Nanomedicine (SEON), ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Didier Letourneur
- INSERM, U1148, LVTS, Paris Diderot University, X Bichat Hospital, Paris, France
| | - Iwona Cicha
- Section of Experimental Oncology and Nanomedicine (SEON), ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| |
Collapse
|
26
|
Chandrasekharan P, Tay ZW, Zhou XY, Yu E, Orendorff R, Hensley D, Huynh Q, Fung KLB, VanHook CC, Goodwill P, Zheng B, Conolly S. A perspective on a rapid and radiation-free tracer imaging modality, magnetic particle imaging, with promise for clinical translation. Br J Radiol 2018; 91:20180326. [PMID: 29888968 DOI: 10.1259/bjr.20180326] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Magnetic particle imaging (MPI), introduced at the beginning of the twenty-first century, is emerging as a promising diagnostic tool in addition to the current repertoire of medical imaging modalities. Using superparamagnetic iron oxide nanoparticles (SPIOs), that are available for clinical use, MPI produces high contrast and highly sensitive tomographic images with absolute quantitation, no tissue attenuation at-depth, and there are no view limitations. The MPI signal is governed by the Brownian and Néel relaxation behavior of the particles. The relaxation time constants of these particles can be utilized to map information relating to the local microenvironment, such as viscosity and temperature. Proof-of-concept pre-clinical studies have shown favourable applications of MPI for better understanding the pathophysiology associated with vascular defects, tracking cell-based therapies and nanotheranostics. Functional imaging techniques using MPI will be useful for studying the pathology related to viscosity changes such as in vascular plaques and in determining cell viability of superparamagnetic iron oxide nanoparticle labeled cells. In this review article, an overview of MPI is provided with discussions mainly focusing on MPI tracers, applications of translational capabilities ranging from diagnostics to theranostics and finally outline a promising path towards clinical translation.
Collapse
Affiliation(s)
| | - Zhi Wei Tay
- 1 Department of Bioengineering, University of California , Berkeley, CA , USA
| | - Xinyi Yedda Zhou
- 1 Department of Bioengineering, University of California , Berkeley, CA , USA
| | - Elaine Yu
- 2 Magnetic Insight Inc , Alameda, CA , USA
| | | | | | - Quincy Huynh
- 1 Department of Bioengineering, University of California , Berkeley, CA , USA
| | - K L Barry Fung
- 1 Department of Bioengineering, University of California , Berkeley, CA , USA
| | | | | | - Bo Zheng
- 1 Department of Bioengineering, University of California , Berkeley, CA , USA
| | - Steven Conolly
- 1 Department of Bioengineering, University of California , Berkeley, CA , USA.,3 Department of Electrical Engineering and Computer Sciences, University of California , Berkeley, CA , USA
| |
Collapse
|
27
|
Ugga L, Romeo V, Tedeschi E, Brunetti A, Quarantelli M. Superparamagnetic iron oxide nanocolloids in MRI studies of neuroinflammation. J Neurosci Methods 2018; 310:12-23. [PMID: 29913184 DOI: 10.1016/j.jneumeth.2018.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
Abstract
Iron oxide (IO) nanocolloids are being increasingly used to image cellular contribution to neuroinflammation using MRI, as these particles are capable of labeling circulating cells with phagocytic activity, allowing to assess cell trafficking from the blood to neuroinflammation sites. The use of IOs relies on the natural phagocytic properties of immune cells, allowing their labeling either in vitro or directly in vivo, following intravenous injection. Despite concerns on the specificity of the latter approach, the widespread availability and relatively low cost of these techniques, coupled to a sensitivity that allows to reach single cell detection, have promoted their use in several preclinical and clinical studies. In this review, we discuss the results of currently available preclinical and clinical IO-enhanced MRI studies of immune cell trafficking in neuroinflammation, examining the specificity of the existing findings, in view of the different possible mechanisms underlying IO accumulation in the brain. From this standpoint, we assess the implications of the temporal and spatial differences in the enhancement pattern of IOs, compared to gadolinium-based contrast agents, a clinically established MRI marker blood-brain barrier breakdown. While concerns on the specificity of cell labeling obtained using the in-vivo labeling approach still need to be fully addressed, these techniques have indeed proved able to provide additional information on neuroinflammatory phenomena, as compared to conventional Gadolinium-enhanced MRI.
Collapse
Affiliation(s)
- Lorenzo Ugga
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Enrico Tedeschi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Mario Quarantelli
- Biostructure and Bioimaging Institute, National Research Council, Naples, Italy.
| |
Collapse
|
28
|
Brisset JC, Gazeau F, Corot C, Nighoghossian N, Berthezène Y, Canet-Soulas E, Wiart M. INFLAM – INFLAMmation in Brain and Vessels with Iron Nanoparticles and Cell Trafficking: A Multiscale Approach of Tissue Microenvironment, Iron Nanostructure and Iron Biotransformation. Ing Rech Biomed 2018. [DOI: 10.1016/j.irbm.2018.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
MacAskill MG, Saif J, Condie A, Jansen MA, MacGillivray TJ, Tavares AAS, Fleisinger L, Spencer HL, Besnier M, Martin E, Biglino G, Newby DE, Hadoke PWF, Mountford JC, Emanueli C, Baker AH. Robust Revascularization in Models of Limb Ischemia Using a Clinically Translatable Human Stem Cell-Derived Endothelial Cell Product. Mol Ther 2018; 26:1669-1684. [PMID: 29703701 PMCID: PMC6035339 DOI: 10.1016/j.ymthe.2018.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cell-derived differentiated endothelial cells offer high potential in regenerative medicine in the cardiovascular system. With the aim of translating the use of a human stem cell-derived endothelial cell product (hESC-ECP) for treatment of critical limb ischemia (CLI) in man, we report a good manufacturing practice (GMP)-compatible protocol and detailed cell tracking and efficacy data in multiple preclinical models. The clinical-grade cell line RC11 was used to generate hESC-ECP, which was identified as mostly endothelial (60% CD31+/CD144+), with the remainder of the subset expressing various pericyte/mesenchymal stem cell markers. Cell tracking using MRI, PET, and qPCR in a murine model of limb ischemia demonstrated that hESC-ECP was detectable up to day 7 following injection. Efficacy in several murine models of limb ischemia (immunocompromised/immunocompetent mice and mice with either type I/II diabetes mellitus) demonstrated significantly increased blood perfusion and capillary density. Overall, we demonstrate a GMP-compatible hESC-ECP that improved ischemic limb perfusion and increased local angiogenesis without engraftment, paving the way for translation of this therapy.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Jaimy Saif
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Alison Condie
- Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Maurits A Jansen
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | | | - Adriana A S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| | - Lucija Fleisinger
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Helen L Spencer
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Marie Besnier
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Ernesto Martin
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Giovanni Biglino
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Joanne C Mountford
- Scottish National Blood Transfusion Service, Edinburgh, UK; Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Costanza Emanueli
- Experimental Cardiovascular Medicine Division, Bristol Heart Institute, University of Bristol, Bristol, UK; National Heart and Lung Institute, Imperial College London, London, UK
| | - Andrew H Baker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
30
|
Unterweger H, Dézsi L, Matuszak J, Janko C, Poettler M, Jordan J, Bäuerle T, Szebeni J, Fey T, Boccaccini AR, Alexiou C, Cicha I. Dextran-coated superparamagnetic iron oxide nanoparticles for magnetic resonance imaging: evaluation of size-dependent imaging properties, storage stability and safety. Int J Nanomedicine 2018; 13:1899-1915. [PMID: 29636608 PMCID: PMC5880571 DOI: 10.2147/ijn.s156528] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Rising criticism of currently available contrast agents for magnetic resonance imaging, either due to their side effects or limited possibilities in terms of functional imaging, evoked the need for safer and more versatile agents. We previously demonstrated the suitability of novel dextran-coated superparamagnetic iron oxide nanoparticles (SPIONDex) for biomedical applications in terms of safety and biocompatibility. METHODS In the present study, we investigated the size-dependent cross-linking process of these particles as well as the size dependency of their imaging properties. For the latter purpose, we adopted a simple and easy-to-perform experiment to estimate the relaxivity of the particles. Furthermore, we performed an extensive analysis of the particles' storage stability under different temperature conditions, showing their superb stability and the lack of any signs of agglomeration or sedimentation during a 12 week period. RESULTS Independent of their size, SPIONDex displayed no irritation potential in a chick chorioallantoic membrane assay. Cell uptake studies of ultra-small (30 nm) SPIONDex confirmed their internalization by macrophages, but not by non-phagocytic cells. Additionally, complement activation-related pseudoallergy (CARPA) experiments in pigs treated with ultra-small SPIONDex indicated the absence of hypersensitivity reactions. CONCLUSION These results emphasize the exceptional safety of SPIONDex, setting them apart from the existing SPION-based contrast agents and making them a very promising candidate for further clinical development.
Collapse
Affiliation(s)
- Harald Unterweger
- ENT Department, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - László Dézsi
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
- SeroScience Ltd., Budapest, Hungary
| | - Jasmin Matuszak
- ENT Department, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christina Janko
- ENT Department, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Marina Poettler
- ENT Department, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jutta Jordan
- Institute of Radiology, Preclinical Imaging Platform Erlangen, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, Preclinical Imaging Platform Erlangen, Universitätsklinikum Erlangen, Erlangen, Germany
| | - János Szebeni
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
- SeroScience Ltd., Budapest, Hungary
| | - Tobias Fey
- Institute of Glass and Ceramics, Department of Materials Science and Engineering, University Erlangen-Nuremberg, Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Alexiou
- ENT Department, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Iwona Cicha
- ENT Department, Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung Professorship, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
31
|
Hu S, Zhou Y, Zhao Y, Xu Y, Zhang F, Gu N, Ma J, Reynolds MA, Xia Y, Xu HH. Enhanced bone regeneration and visual monitoring via superparamagnetic iron oxide nanoparticle scaffold in rats. J Tissue Eng Regen Med 2018; 12:e2085-e2098. [DOI: 10.1002/term.2641] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 11/28/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Shuying Hu
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
| | - Yi Zhou
- Yixing People's Hospital Yixing P. R. China
| | - Yantao Zhao
- Beijing Engineering Research Center of Orthopaedic ImplantsFirst Affiliated Hospital of CPLA General Hospital Beijing P. R. China
| | - Yang Xu
- Affiliated Stomatology Hospital of Soochow University Suzhou P. R. China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
| | - Ning Gu
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical EngineeringSoutheast University Nanjing P. R. China
- Suzhou Institute & Collaborative Innovation Center of Suzhou Nano Science and TechnologySoutheast University Suzhou P. R. China
| | - Junqing Ma
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
| | - Mark A. Reynolds
- Department of Advanced Oral Sciences & TherapeuticsUniversity of Maryland School of Dentistry Baltimore MD USA
| | - Yang Xia
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical University Nanjing P. R. China
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical EngineeringSoutheast University Nanjing P. R. China
- Department of Advanced Oral Sciences & TherapeuticsUniversity of Maryland School of Dentistry Baltimore MD USA
| | - Hockin H.K. Xu
- Department of Advanced Oral Sciences & TherapeuticsUniversity of Maryland School of Dentistry Baltimore MD USA
- Center for Stem Cell Biology & Regenerative MedicineUniversity of Maryland School of Medicine Baltimore MD USA
- Department of Mechanical EngineeringUniversity of Maryland Baltimore County Baltimore County MD USA
| |
Collapse
|
32
|
Namestnikova D, Gubskiy I, Kholodenko I, Melnikov P, Sukhinich K, Gabashvili A, Vishnevskiy D, Soloveva A, Abakumov M, Vakhrushev I, Lupatov A, Chekhonin V, Gubsky L, Yarygin K. Methodological aspects of MRI of transplanted superparamagnetic iron oxide-labeled mesenchymal stem cells in live rat brain. PLoS One 2017; 12:e0186717. [PMID: 29049361 PMCID: PMC5648235 DOI: 10.1371/journal.pone.0186717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/08/2017] [Indexed: 12/22/2022] Open
Abstract
In vivo tracking of transplanted mesenchymal stem cells (MSCs) migration and homing is vital for understanding the mechanisms of beneficial effects of MSCs transplantation in animal models of diseases and in clinical trials. Transplanted cells can be labeled with superparamagnetic iron oxide (SPIO) particles and visualized in vivo using a number of iron sensitive MRI techniques. However, the applicability of those techniques for SPIO-labeled MSCs tracking in live brain has not been sufficiently investigated. The goal of this study was to estimate the efficiency of various MRI techniques of SPIO-labeled cell tracing in the brain. To achieve that goal, the precision and specificity of T2WI, T2*WI and SWI (Susceptibility-Weighted Imaging) techniques of SPIO-labeled MSCs tracing in vitro and in live rat brain were for the first time compared in the same experiment. We have shown that SWI presents the most sensitive pulse sequence for SPIO-labeled MSCs MR visualization. After intracerebral administration due to limitations caused by local micro-hemorrhages the visualization threshold was 102 cells, while after intra-arterial transplantation SWI permitted detection of several cells or even single cells. There is just one publication claiming detection of individual SPIO-labeled MSCs in live brain, while the other state much lower sensitivity, describe detection of different cell types or high resolution tracing of MSCs in other tissues. This study confirms the possibility of single cell tracing in live brain and outlines the necessary conditions. SWI is a method convenient for the detection of single SPIO labeled MSCs and small groups of SPIO labeled MSCs in brain tissue and can be appropriate for monitoring migration and homing of transplanted cells in basic and translational neuroscience.
Collapse
Affiliation(s)
| | - Ilya Gubskiy
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Pavel Melnikov
- Pirogov Russian National Research Medical University, Moscow, Russia.,Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | | | | | | | | | - Maxim Abakumov
- Pirogov Russian National Research Medical University, Moscow, Russia.,National University of Science and Technology, Moscow, Russia
| | | | | | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University, Moscow, Russia.,Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Leonid Gubsky
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | |
Collapse
|
33
|
Alaarg A, Pérez-Medina C, Metselaar JM, Nahrendorf M, Fayad ZA, Storm G, Mulder WJM. Applying nanomedicine in maladaptive inflammation and angiogenesis. Adv Drug Deliv Rev 2017; 119:143-158. [PMID: 28506745 PMCID: PMC5682240 DOI: 10.1016/j.addr.2017.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
Inflammation and angiogenesis drive the development and progression of multiple devastating diseases such as atherosclerosis, cancer, rheumatoid arthritis, and inflammatory bowel disease. Though these diseases have very different phenotypic consequences, they possess several common pathophysiological features in which monocyte recruitment, macrophage polarization, and enhanced vascular permeability play critical roles. Thus, developing rational targeting strategies tailored to the different stages of the journey of monocytes, from bone marrow to local lesions, and their extravasation from the vasculature in diseased tissues will advance nanomedicine. The integration of in vivo imaging uniquely allows studying nanoparticle kinetics, accumulation, clearance, and biological activity, at levels ranging from subcellular to an entire organism, and will shed light on the fate of intravenously administered nanomedicines. We anticipate that convergence of nanomedicines, biomedical engineering, and life sciences will help to advance clinically relevant therapeutics and diagnostic agents for patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Amr Alaarg
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Institute for Experimental Molecular Imaging, University Clinic, Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
34
|
Unterweger H, Janko C, Schwarz M, Dézsi L, Urbanics R, Matuszak J, Őrfi E, Fülöp T, Bäuerle T, Szebeni J, Journé C, Boccaccini AR, Alexiou C, Lyer S, Cicha I. Non-immunogenic dextran-coated superparamagnetic iron oxide nanoparticles: a biocompatible, size-tunable contrast agent for magnetic resonance imaging. Int J Nanomedicine 2017; 12:5223-5238. [PMID: 28769560 PMCID: PMC5533574 DOI: 10.2147/ijn.s138108] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Iron oxide-based contrast agents have been in clinical use for magnetic resonance imaging (MRI) of lymph nodes, liver, intestines, and the cardiovascular system. Superparamagnetic iron oxide nanoparticles (SPIONs) have high potential as a contrast agent for MRI, but no intravenous iron oxide-containing agents are currently approved for clinical imaging. The aim of our work was to analyze the hemocompatibility and immuno-safety of a new type of dextran-coated SPIONs (SPIONdex) and to characterize these nanoparticles with ultra-high-field MRI. Key parameters related to nanoparticle hemocompatibility and immuno-safety were investigated in vitro and ex vivo. To address concerns associated with hypersensitivity reactions to injectable nanoparticulate agents, we analyzed complement activation-related pseudoallergy (CARPA) upon intravenous administration of SPIONdex in a pig model. Furthermore, the size-tunability of SPIONdex and the effects of size reduction on their biocompatibility were investigated. In vitro, SPIONdex did not induce hemolysis, complement or platelet activation, plasma coagulation, or leukocyte procoagulant activity, and had no relevant effect on endothelial cell viability or endothelial–monocytic cell interactions. Furthermore, SPIONdex did not induce CARPA even upon intravenous administration of 5 mg Fe/kg in pigs. Upon SPIONdex administration in mice, decreased liver signal intensity was observed after 15 minutes and was still detectable 24 h later. In addition, by changing synthesis parameters, a reduction in particle size <30 nm was achieved, without affecting their hemo- and biocompatibility. Our findings suggest that due to their excellent biocompatibility, safety upon intravenous administration and size-tunability, SPIONdex particles may represent a suitable candidate for a new-generation MRI contrast agent.
Collapse
Affiliation(s)
- Harald Unterweger
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universitaet Erlangen-Nuernberg
| | - Christina Janko
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universitaet Erlangen-Nuernberg
| | - Marc Schwarz
- Preclinical Imaging Platform Erlangen (PIPE), Institute of Radiology, University Hospital Erlangen, Erlangen, Germany
| | - László Dézsi
- Nanomedicine Research and Education Center, Semmelweis University
| | | | - Jasmin Matuszak
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universitaet Erlangen-Nuernberg
| | - Erik Őrfi
- Nanomedicine Research and Education Center, Semmelweis University
| | - Tamás Fülöp
- Nanomedicine Research and Education Center, Semmelweis University
| | - Tobias Bäuerle
- Preclinical Imaging Platform Erlangen (PIPE), Institute of Radiology, University Hospital Erlangen, Erlangen, Germany
| | - János Szebeni
- Nanomedicine Research and Education Center, Semmelweis University.,SeroScience Ltd., Budapest, Hungary
| | - Clément Journé
- Inserm U1148, Fédération de Recherche en Imagerie Multimodalités (FRIM), X Bichat Hospital, Paris Diderot University, Paris, France
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Alexiou
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universitaet Erlangen-Nuernberg
| | - Stefan Lyer
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universitaet Erlangen-Nuernberg
| | - Iwona Cicha
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology und Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universitaet Erlangen-Nuernberg
| |
Collapse
|
35
|
Singh R, Wieser A, Reakasame S, Detsch R, Dietel B, Alexiou C, Boccaccini AR, Cicha I. Cell specificity of magnetic cell seeding approach to hydrogel colonization. J Biomed Mater Res A 2017. [PMID: 28639348 DOI: 10.1002/jbm.a.36147] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tissue-engineered scaffolds require an effective colonization with cells. Superparamagnetic iron oxide nanoparticles (SPIONs) can enhance cell adhesion to matrices by magnetic cell seeding. We investigated the possibility of improving cell attachment and growth on different alginate-based hydrogels using fibroblasts and endothelial cells (ECs) loaded with SPIONs. Hydrogels containing pure alginate (Alg), alginate dialdehyde crosslinked with gelatin (ADA-G) and Alg blended with G or silk fibroin (SF) were prepared. Endothelial cells and fibroblasts loaded with SPIONs were seeded and grown on hydrogels for up to 7 days, in the presence of magnetic field during the first 24 h. Cell morphology (fluorescent staining) and metabolic activity (WST-8 assay) of magnetically-seeded versus conventionally seeded cells were compared. Magnetic seeding of ECs improved their initial attachment and further growth on Alg/G hydrogel surfaces. However, we did not achieve an efficient and stable colonization of ADA-G films with ECs even with magnetic cell seeding. Fibroblast showed good initial colonization and growth on ADA-G and on Alg/SF. This effect was further significantly enhanced by magnetic cell seeding. On pure Alg, initial attachment and spreading of magnetically-seeded cells was dramatically improved compared to conventionally-seeded cells, but the effect was transient and diminished gradually with the cessation of magnetic force. Our results demonstrate that magnetic seeding improves the strength and uniformity of initial cell attachment to hydrogel surface in cell-specific manner, which may play a decisive role for the outcome in tissue engineering applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2948-2956, 2017.
Collapse
Affiliation(s)
- Raminder Singh
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Cardiology and Angiology, University Hospital Erlangen, Erlangen, Germany
| | - Anna Wieser
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Supachai Reakasame
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Detsch
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Dietel
- Department of Cardiology and Angiology, University Hospital Erlangen, Erlangen, Germany
| | - Christoph Alexiou
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Iwona Cicha
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
36
|
MacAskill MG, Tavares AS, Wu J, Lucatelli C, Mountford JC, Baker AH, Newby DE, Hadoke PWF. PET Cell Tracking Using 18F-FLT is Not Limited by Local Reuptake of Free Radiotracer. Sci Rep 2017; 7:44233. [PMID: 28287126 PMCID: PMC5347009 DOI: 10.1038/srep44233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/06/2017] [Indexed: 12/27/2022] Open
Abstract
Assessing the retention of cell therapies following implantation is vital and often achieved by labelling cells with 2'-[18F]-fluoro-2'-deoxy-D-glucose (18F-FDG). However, this approach is limited by local retention of cell-effluxed radiotracer. Here, in a preclinical model of critical limb ischemia, we assessed a novel method of cell tracking using 3'-deoxy-3'-L-[18F]-fluorothymidine (18F-FLT); a clinically available radiotracer which we hypothesise will result in minimal local radiotracer reuptake and allow a more accurate estimation of cell retention. Human endothelial cells (HUVECs) were incubated with 18F-FDG or 18F-FLT and cell characteristics were evaluated. Dynamic positron emission tomography (PET) images were acquired post-injection of free 18F-FDG/18F-FLT or 18F-FDG/18F-FLT-labelled HUVECs, following the surgical induction of mouse hind-limb ischemia. In vitro, radiotracer incorporation and efflux was similar with no effect on cell viability, function or proliferation under optimised conditions (5 MBq/mL, 60 min). Injection of free radiotracer demonstrated a faster clearance of 18F-FLT from the injection site vs. 18F-FDG (p ≤ 0.001), indicating local cellular uptake. Using 18F-FLT-labelling, estimation of HUVEC retention within the engraftment site 4 hr post-administration was 24.5 ± 3.2%. PET cell tracking using 18F-FLT labelling is an improved approach vs. 18F-FDG as it is not susceptible to local host cell reuptake, resulting in a more accurate estimation of cell retention.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Adriana S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Junxi Wu
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Joanne C Mountford
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Andrew H Baker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
37
|
Sharkey J, Starkey Lewis PJ, Barrow M, Alwahsh SM, Noble J, Livingstone E, Lennen RJ, Jansen MA, Carrion JG, Liptrott N, Forbes S, Adams DJ, Chadwick AE, Forbes SJ, Murray P, Rosseinsky MJ, Goldring CE, Park BK. Functionalized superparamagnetic iron oxide nanoparticles provide highly efficient iron-labeling in macrophages for magnetic resonance-based detection in vivo. Cytotherapy 2017; 19:555-569. [PMID: 28214127 PMCID: PMC5357746 DOI: 10.1016/j.jcyt.2017.01.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/01/2016] [Accepted: 01/02/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND AIMS Tracking cells during regenerative cytotherapy is crucial for monitoring their safety and efficacy. Macrophages are an emerging cell-based regenerative therapy for liver disease and can be readily labeled for medical imaging. A reliable, clinically applicable cell-tracking agent would be a powerful tool to study cell biodistribution. METHODS Using a recently described chemical design, we set out to functionalize, optimize and characterize a new set of superparamagnetic iron oxide nanoparticles (SPIONs) to efficiently label macrophages for magnetic resonance imaging-based cell tracking in vivo. RESULTS A series of cell health and iron uptake assays determined that positively charged SPIONs (+16.8 mV) could safely label macrophages more efficiently than the formerly approved ferumoxide (-6.7 mV; Endorem) and at least 10 times more efficiently than the clinically approved SPION ferumoxytol (-24.2 mV; Rienso). An optimal labeling time of 4 h at 25 µg/mL was demonstrated to label macrophages of mouse and human origin without any adverse effects on cell viability whilst providing substantial iron uptake (>5 pg Fe/cell) that was retained for 7 days in vitro. SPION labeling caused no significant reduction in phagocytic activity and a shift toward a reversible M1-like phenotype in bone marrow-derived macrophages (BMDMs). Finally, we show that SPION-labeled BMDMs delivered via the hepatic portal vein to mice are localized in the hepatic parenchyma resulting in a 50% drop in T2* in the liver. Engraftment of exogenous cells was confirmed via immunohistochemistry up to 3 weeks posttransplantation. DISCUSSION A positively charged dextran-coated SPION is a promising tool to noninvasively track hepatic macrophage localization for therapeutic monitoring.
Collapse
Affiliation(s)
- Jack Sharkey
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom
| | - Philip J Starkey Lewis
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; MRC Centre for Regenerative Medicine, Little France Drive, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Barrow
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Salamah M Alwahsh
- MRC Centre for Regenerative Medicine, Little France Drive, University of Edinburgh, Edinburgh, United Kingdom
| | - June Noble
- Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Eilidh Livingstone
- MRC Centre for Regenerative Medicine, Little France Drive, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross J Lennen
- Edinburgh Preclinical Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | - Maurits A Jansen
- Edinburgh Preclinical Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Neill Liptrott
- MRC Centre for Drug Safety Science, Ashton Street, University of Liverpool, Liverpool, United Kingdom; European Nanomedicine Characterisation Laboratory (EU-NCL), Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Shareen Forbes
- Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Dave J Adams
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Amy E Chadwick
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; MRC Centre for Drug Safety Science, Ashton Street, University of Liverpool, Liverpool, United Kingdom
| | - Stuart J Forbes
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; MRC Centre for Regenerative Medicine, Little France Drive, University of Edinburgh, Edinburgh, United Kingdom
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom; UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom
| | - Matthew J Rosseinsky
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; Department of Chemistry, University of Liverpool, Liverpool, United Kingdom
| | - Christopher E Goldring
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; MRC Centre for Drug Safety Science, Ashton Street, University of Liverpool, Liverpool, United Kingdom.
| | - B Kevin Park
- UK Regenerative Medicine Platform Safety and Efficacy Hub, United Kingdom; MRC Centre for Drug Safety Science, Ashton Street, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
38
|
Extremely Small Pseudoparamagnetic Iron Oxide Nanoparticle as a Novel Blood Pool T1 Magnetic Resonance Contrast Agent for 3 T Whole-Heart Coronary Angiography in Canines. Invest Radiol 2017; 52:128-133. [DOI: 10.1097/rli.0000000000000321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Wáng YXJ, Idée JM. A comprehensive literatures update of clinical researches of superparamagnetic resonance iron oxide nanoparticles for magnetic resonance imaging. Quant Imaging Med Surg 2017; 7:88-122. [PMID: 28275562 DOI: 10.21037/qims.2017.02.09] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This paper aims to update the clinical researches using superparamagnetic iron oxide (SPIO) nanoparticles as magnetic resonance imaging (MRI) contrast agent published during the past five years. PubMed database was used for literature search, and the search terms were (SPIO OR superparamagnetic iron oxide OR Resovist OR Ferumoxytol OR Ferumoxtran-10) AND (MRI OR magnetic resonance imaging). The literature search results show clinical research on SPIO remains robust, particularly fuelled by the approval of ferumoxytol for intravenously administration. SPIOs have been tested on MR angiography, sentinel lymph node detection, lymph node metastasis evaluation; inflammation evaluation; blood volume measurement; as well as liver imaging. Two experimental SPIOs with unique potentials are also discussed in this review. A curcumin-conjugated SPIO can penetrate brain blood barrier (BBB) and bind to amyloid plaques in Alzheime's disease transgenic mice brain, and thereafter detectable by MRI. Another SPIO was fabricated with a core of Fe3O4 nanoparticle and a shell coating of concentrated hydrophilic polymer brushes and are almost not taken by peripheral macrophages as well as by mononuclear phagocytes and reticuloendothelial system (RES) due to the suppression of non-specific protein binding caused by their stealthy ''brush-afforded'' structure. This SPIO may offer potentials for the applications such as drug targeting and tissue or organ imaging other than liver and lymph nodes.
Collapse
Affiliation(s)
- Yì Xiáng J Wáng
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, New Territories, Hong Kong SAR, China
| | - Jean-Marc Idée
- Guerbet, Research and Innovation Division, Roissy-Charles de Gaulle, France
| |
Collapse
|
40
|
Zanganeh S, Hutter G, Spitler R, Lenkov O, Mahmoudi M, Shaw A, Pajarinen JS, Nejadnik H, Goodman S, Moseley M, Coussens LM, Daldrup-Link HE. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. NATURE NANOTECHNOLOGY 2016; 11:986-994. [PMID: 27668795 PMCID: PMC5198777 DOI: 10.1038/nnano.2016.168] [Citation(s) in RCA: 1068] [Impact Index Per Article: 133.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 08/11/2016] [Indexed: 05/13/2023]
Abstract
Until now, the Food and Drug Administration (FDA)-approved iron supplement ferumoxytol and other iron oxide nanoparticles have been used for treating iron deficiency, as contrast agents for magnetic resonance imaging and as drug carriers. Here, we show an intrinsic therapeutic effect of ferumoxytol on the growth of early mammary cancers, and lung cancer metastases in liver and lungs. In vitro, adenocarcinoma cells co-incubated with ferumoxytol and macrophages showed increased caspase-3 activity. Macrophages exposed to ferumoxytol displayed increased mRNA associated with pro-inflammatory Th1-type responses. In vivo, ferumoxytol significantly inhibited growth of subcutaneous adenocarcinomas in mice. In addition, intravenous ferumoxytol treatment before intravenous tumour cell challenge prevented development of liver metastasis. Fluorescence-activated cell sorting (FACS) and histopathology studies showed that the observed tumour growth inhibition was accompanied by increased presence of pro-inflammatory M1 macrophages in the tumour tissues. Our results suggest that ferumoxytol could be applied 'off label' to protect the liver from metastatic seeds and potentiate macrophage-modulating cancer immunotherapies.
Collapse
Affiliation(s)
- Saeid Zanganeh
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Road, Stanford, California 94305, USA
- Institute of Stem Cell Research and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
| | - Gregor Hutter
- Institute of Stem Cell Research and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Department of Neurosurgery, Stanford University, Stanford, California 94305, USA
| | - Ryan Spitler
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Road, Stanford, California 94305, USA
| | - Olga Lenkov
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Road, Stanford, California 94305, USA
- Institute of Stem Cell Research and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
| | - Morteza Mahmoudi
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, California 94305, USA
| | - Aubie Shaw
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Jukka Sakari Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University, Stanford, California 94305, USA
| | - Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Road, Stanford, California 94305, USA
- Institute of Stem Cell Research and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
| | - Stuart Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University, Stanford, California 94305, USA
| | - Michael Moseley
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Road, Stanford, California 94305, USA
| | - Lisa Marie Coussens
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Heike Elisabeth Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, 725 Welch Road, Stanford, California 94305, USA
- Institute of Stem Cell Research and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Department of Pediatrics, Stanford University, Stanford, California 94305, USA
- Correspondence and requests for materials should be addressed to H.E.D.-L.
| |
Collapse
|
41
|
Bietenbeck M, Florian A, Faber C, Sechtem U, Yilmaz A. Remote magnetic targeting of iron oxide nanoparticles for cardiovascular diagnosis and therapeutic drug delivery: where are we now? Int J Nanomedicine 2016; 11:3191-203. [PMID: 27486321 PMCID: PMC4957681 DOI: 10.2147/ijn.s110542] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Magnetic resonance imaging (MRI) allows for an accurate assessment of both functional and structural cardiac parameters, and thereby appropriate diagnosis and validation of cardiovascular diseases. The diagnostic yield of cardiovascular MRI examinations is often increased by the use of contrast agents that are almost exclusively based on gadolinium compounds. Another clinically approved contrast medium is composed of superparamagnetic iron oxide nanoparticles (IONs). These particles may expand the field of contrast-enhanced cardiovascular MRI as recently shown in clinical studies focusing on acute myocardial infarction (AMI) and atherosclerosis. Furthermore, IONs open up new research opportunities such as remote magnetic drug targeting (MDT). The approach of MDT relies on the coupling of bioactive molecules and magnetic nanoparticles to form an injectable complex. This complex, in turn, can be attracted to and retained at a desired target inside the body with the help of applied magnetic fields. In comparison to common systemic drug applications, MDT techniques promise both higher concentrations at the target site and lower concentrations elsewhere in the body. Moreover, concurrent or subsequent MRI can be used for noninvasive monitoring of drug distribution and successful delivery to the desired organ in vivo. This review does not only illustrate the basic conceptual and biophysical principles of IONs, but also focuses on new research activities and achievements in the cardiovascular field, mainly in the management of AMI. Based on the presentation of successful MDT applications in preclinical models of AMI, novel approaches and the translational potential of MDT are discussed.
Collapse
Affiliation(s)
| | | | - Cornelius Faber
- Department of Clinical Radiology, University Hospital Münster, Münster
| | - Udo Sechtem
- Division of Cardiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | | |
Collapse
|
42
|
Pereira SM, Herrmann A, Moss D, Poptani H, Williams SR, Murray P, Taylor A. Evaluating the effectiveness of transferrin receptor-1 (TfR1) as a magnetic resonance reporter gene. CONTRAST MEDIA & MOLECULAR IMAGING 2016; 11:236-44. [PMID: 26929139 PMCID: PMC4981909 DOI: 10.1002/cmmi.1686] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/06/2016] [Accepted: 01/08/2016] [Indexed: 12/13/2022]
Abstract
Magnetic resonance (MR) reporter genes have the potential for tracking the biodistribution and fate of cells in vivo, thus allowing the safety, efficacy and mechanisms of action of cell-based therapies to be comprehensively assessed. In this study, we evaluate the effectiveness of the iron importer transferrin receptor-1 (TfR1) as an MR reporter gene in the model cell line CHO-K1. Overexpression of the TfR1 transgene led to a reduction in the levels of endogenous TfR1 mRNA, but to a 60-fold increase in total TfR1 protein levels. Although the mRNA levels of ferritin heavy chain-1 (Fth1) did not change, Fth1 protein levels increased 13-fold. The concentration of intracellular iron increased significantly, even when cells were cultured in medium that was not supplemented with iron and the amount of iron in the extracellular environment was thus at physiological levels. However, we found that, by supplementing the cell culture medium with ferric citrate, a comparable degree of iron uptake and MR contrast could be achieved in control cells that did not express the TfR1 transgene. Sufficient MR contrast to enable the cells to be detected in vivo following their administration into the midbrain of chick embryos was obtained irrespective of the reporter gene. We conclude that TfR1 is not an effective reporter and that, to track the biodistribution of cells with MR imaging in the short term, it is sufficient to simply culture cells in the presence of ferric citrate. Copyright © 2016 The Authors Contrast Media & Molecular Imaging Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Sofia M Pereira
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Anne Herrmann
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Diana Moss
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Harish Poptani
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Steve R Williams
- Centre for Imaging Sciences, Oxford Road, University of Manchester, Manchester, UK
| | - Patricia Murray
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Arthur Taylor
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
43
|
Connell JJ, Patrick PS, Yu Y, Lythgoe MF, Kalber TL. Advanced cell therapies: targeting, tracking and actuation of cells with magnetic particles. Regen Med 2015; 10:757-72. [PMID: 26390317 DOI: 10.2217/rme.15.36] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Regenerative medicine would greatly benefit from a new platform technology that enabled measurable, controllable and targeting of stem cells to a site of disease or injury in the body. Superparamagnetic iron-oxide nanoparticles offer attractive possibilities in biomedicine and can be incorporated into cells, affording a safe and reliable means of tagging. This review describes three current and emerging methods to enhance regenerative medicine using magnetic particles to guide therapeutic cells to a target organ; track the cells using MRI and assess their spatial localization with high precision and influence the behavior of the cell using magnetic actuation. This approach is complementary to the systemic injection of cell therapies, thus expanding the horizon of stem cell therapeutics.
Collapse
Affiliation(s)
- John J Connell
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - P Stephen Patrick
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Yichao Yu
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Mark F Lythgoe
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| | - Tammy L Kalber
- UCL Centre of Advanced Biomedical Imaging, Division of Medicine, University College London, Paul O'Gorman Building, 72 Huntley Street, London, WC1E 6DD, UK
| |
Collapse
|
44
|
Alam SR, Stirrat C, Richards J, Mirsadraee S, Semple SIK, Tse G, Henriksen P, Newby DE. Vascular and plaque imaging with ultrasmall superparamagnetic particles of iron oxide. J Cardiovasc Magn Reson 2015; 17:83. [PMID: 26381872 PMCID: PMC4574723 DOI: 10.1186/s12968-015-0183-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 08/16/2015] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular Magnetic Resonance (CMR) has become a primary tool for non-invasive assessment of cardiovascular anatomy, pathology and function. Existing contrast agents have been utilised for the identification of infarction, fibrosis, perfusion deficits and for angiography. Novel ultrasmall superparamagnetic particles of iron oxide (USPIO) contrast agents that are taken up by inflammatory cells can detect cellular inflammation non-invasively using CMR, potentially aiding the diagnosis of inflammatory medical conditions, guiding their treatment and giving insight into their pathophysiology. In this review we describe the utilization of USPIO as a novel contrast agent in vascular disease.
Collapse
Affiliation(s)
- Shirjel R Alam
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Colin Stirrat
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Jennifer Richards
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
| | - Saeed Mirsadraee
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, EH16 5SA, UK.
- Department of Radiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Scott I K Semple
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - George Tse
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - Peter Henriksen
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| | - David E Newby
- Centre for Cardiovascular Science, The University of Edinburgh, The Chancellor's Building, Little France Crescent, Edinburgh, EH16 5SA, UK.
- Department of Cardiology, Royal Infirmary of Edinburgh, Edinburgh, EH16 5SA, UK.
| |
Collapse
|
45
|
MRI/MRS in neuroinflammation: methodology and applications. Clin Transl Imaging 2015; 3:475-489. [PMID: 26705534 PMCID: PMC4679099 DOI: 10.1007/s40336-015-0142-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/30/2015] [Indexed: 12/11/2022]
Abstract
Neuroinflammation encompasses a wide range of humoral and cellular responses, not only enabling the CNS to fight various noxious events, including infections and trauma, but also playing a critical role in autoimmune as well as in neurodegenerative diseases. The complex interactions of immune, endothelial, and neuronal cells that take place during inflammation require an equivalent complexity of imaging approaches to be appropriately explored in vivo. Magnetic Resonance provides several complementary techniques that allow to study most mechanisms underlying the brain/immune interaction. In this review, we discuss the MR approaches to the study of endothelial activation, blood-brain barrier permeability alterations, intercellular compartment modifications, immune cell trafficking, and of metabolic alterations linked to immune cell activity. The main advantages and limitations of these techniques are assessed, in view of their exploitation in the clinical arena, where the complementarity of the information that can be obtained has the potential to change our way of studying neuroinflammation, with implications for the management of several CNS diseases.
Collapse
|
46
|
Matuszak J, Dörfler P, Zaloga J, Unterweger H, Lyer S, Dietel B, Alexiou C, Cicha I. Shell matters: Magnetic targeting of SPIONs and in vitro effects on endothelial and monocytic cell function. Clin Hemorheol Microcirc 2015; 61:259-77. [DOI: 10.3233/ch-151998] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jasmin Matuszak
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| | - Philipp Dörfler
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| | - Jan Zaloga
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| | - Harald Unterweger
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| | - Stefan Lyer
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| | - Barbara Dietel
- Laboratory of Molecular Cardiology, Department of Cardiology and Angiology, University Hospital Erlangen, Germany
| | - Christoph Alexiou
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| | - Iwona Cicha
- Section of Experimental Oncology und Nanomedicine (SEON), ENT-Department, Erlangen, Germany
| |
Collapse
|
47
|
Lane LA, Qian X, Nie S. SERS Nanoparticles in Medicine: From Label-Free Detection to Spectroscopic Tagging. Chem Rev 2015; 115:10489-529. [DOI: 10.1021/acs.chemrev.5b00265] [Citation(s) in RCA: 607] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Lucas A. Lane
- Departments
of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Health Sciences Research Building,
Room E116, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| | - Ximei Qian
- Departments
of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Health Sciences Research Building,
Room E116, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| | - Shuming Nie
- Departments
of Biomedical Engineering and Chemistry, Emory University and Georgia Institute of Technology, Health Sciences Research Building,
Room E116, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
- College
of Engineering and Applied Sciences, Nanjing University, 22 Hankou
Road, Nanjing, Jiangsu Province 210093, China
| |
Collapse
|
48
|
Bernsen MR, Guenoun J, van Tiel ST, Krestin GP. Nanoparticles and clinically applicable cell tracking. Br J Radiol 2015; 88:20150375. [PMID: 26248872 DOI: 10.1259/bjr.20150375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In vivo cell tracking has emerged as a much sought after tool for design and monitoring of cell-based treatment strategies. Various techniques are available for pre-clinical animal studies, from which much has been learned and still can be learned. However, there is also a need for clinically translatable techniques. Central to in vivo cell imaging is labelling of cells with agents that can give rise to signals in vivo, that can be detected and measured non-invasively. The current imaging technology of choice for clinical translation is MRI in combination with labelling of cells with magnetic agents. The main challenge encountered during the cell labelling procedure is to efficiently incorporate the label into the cell, such that the labelled cells can be imaged at high sensitivity for prolonged periods of time, without the labelling process affecting the functionality of the cells. In this respect, nanoparticles offer attractive features since their structure and chemical properties can be modified to facilitate cellular incorporation and because they can carry a high payload of the relevant label into cells. While these technologies have already been applied in clinical trials and have increased the understanding of cell-based therapy mechanism, many challenges are still faced.
Collapse
Affiliation(s)
- Monique R Bernsen
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands.,2 Department of Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Jamal Guenoun
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands
| | | | | |
Collapse
|
49
|
Bietenbeck M, Florian A, Sechtem U, Yilmaz A. The diagnostic value of iron oxide nanoparticles for imaging of myocardial inflammation--quo vadis? J Cardiovasc Magn Reson 2015; 17:54. [PMID: 26152269 PMCID: PMC4495803 DOI: 10.1186/s12968-015-0165-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/24/2015] [Indexed: 01/23/2023] Open
Abstract
Cardiovascular magnetic resonance (CMR) is an integral part in the diagnostic work-up of cardiac inflammatory diseases. In this context, superparamagnetic iron oxide-based contrast agents can provide additional diagnostic information regarding the assessment of myocardial infarction and myocarditis. After intravenous administration, these nanoparticles are taken up by activated monocytes and macrophages, which predominantly accumulate in regions associated with inflammation as was successfully shown in recent preclinical studies. Furthermore, first clinical studies with a new iron oxide-complex that was clinically approved for the treatment of iron deficiency anaemia recently demonstrated a superior diagnostic value of iron oxide nanoparticles compared to gadolinium-based compounds for imaging of myocardial inflammation in patients with acute myocardial infarction. In this article, we outline the basic features of superparamagnetic iron oxide-based contrast agents and review recent studies using such nanoparticles for cardiac imaging in case of acute myocardial infarction as well as acute myocarditis. Moreover, we highlight the translational potential of these agents and possible research applications with regard to imaging and therapy.
Collapse
Affiliation(s)
- Michael Bietenbeck
- Department of Cardiology and Angiology, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany
| | - Anca Florian
- Department of Cardiology and Angiology, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany
| | - Udo Sechtem
- Division of Cardiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Ali Yilmaz
- Department of Cardiology and Angiology, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany.
| |
Collapse
|
50
|
Peng M, Li H, Luo Z, Kong J, Wan Y, Zheng L, Zhang Q, Niu H, Vermorken A, Van de Ven W, Chen C, Zhang X, Li F, Guo L, Cui Y. Dextran-coated superparamagnetic nanoparticles as potential cancer drug carriers in vivo. NANOSCALE 2015; 7:11155-11162. [PMID: 26062012 DOI: 10.1039/c5nr01382h] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Dextran-coated superparamagnetic iron oxide nanoparticles (DSPIONs) have gained considerable interest, because of their biocompatibility and biosafety in clinics. Doxorubicin (Dox), a widely used chemotherapeutic drug, always has limited applications in clinical therapy due to its serious side effects of dose-limiting irreversible cardiotoxicity and myelo suppression. Herein, DSPIONs were synthesized and developed as magnetic carriers for doxorubicin. The Dox-DSPION conjugates were evaluated in the in vitro test of Dox release, which showed pH-dependence with the highest release percentage of 50.3% at pH 5.0 and the lowest release percentage of 11.8% in a physiological environment. The cytotoxicity of DSPIONs and Dox-DSPIONs evaluated by the MTT assay indicated that DSPIONs had no cytotoxicity and the conjugates had significantly reduced the toxicity (IC50 = 1.36 μg mL(-1)) compared to free Dox (IC50 = 0.533 μg mL(-1)). Furthermore, confocal microscopic data of cell uptake suggest that less cytotoxicity of Dox-DSPIONs may be attributed to the cellular internalization of the conjugates and sustainable release of Dox from the formulation in the cytoplasm. More importantly, the results from the rabbit VX2 liver tumor model test under an external magnetic field showed that the conjugates had approximately twice the anti-tumor activity and two and a half times the animal survival rate, respectively, compared to free Dox. Collectively, our data have demonstrated that Dox-DSPIONs have less toxicity with better antitumor effectiveness in in vitro and in vivo applications, suggesting that the conjugates have potential to be developed into chemo-therapeutic formulations.
Collapse
Affiliation(s)
- Mingli Peng
- Key Laboratory of Synthetic and Natural Functional Molecular Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, P. R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|