1
|
Wang J, Zhang H, Wan W, Yang H, Zhao J. Advances in nanotechnological approaches for the detection of early markers associated with severe cardiac ailments. Nanomedicine (Lond) 2024; 19:1487-1506. [PMID: 39121377 PMCID: PMC11318751 DOI: 10.1080/17435889.2024.2364581] [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: 03/01/2024] [Accepted: 05/31/2024] [Indexed: 08/11/2024] Open
Abstract
Mortality from cardiovascular disease (CVD) accounts for over 30% of all deaths globally, necessitating reliable diagnostic tools. Prompt identification and precise diagnosis are critical for effective personalized treatment. Nanotechnology offers promising applications in diagnostics, biosensing and drug delivery for prevalent cardiovascular diseases. Its integration into cardiovascular care enhances diagnostic accuracy, enabling early intervention and tailored treatment plans. By leveraging nanoscale innovations, healthcare professionals can address the complexities of CVD progression and customize interventions based on individual patient needs. Ongoing advancements in nanotechnology continue to shape the landscape of cardiovascular medicine, offering potential for improved patient outcomes and reduced mortality rates from these pervasive diseases.
Collapse
Affiliation(s)
- Jie Wang
- Department of Cardiac Care Unit, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Haifeng Zhang
- Department of Cardiology, Yantai Yeda Hospital, Yantai, Shangdong, 264006, China
| | - Weiping Wan
- Department of Ultrasound, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Haijiao Yang
- Department of Cardiac Care Unit, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Jing Zhao
- Department of Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong, 264003, China
| |
Collapse
|
2
|
Nuschke A, Sobey-Skelton C, Dawod B, Kelly B, Tremblay ML, Davis C, Rioux JA, Brewer K. Use of Magnetotactic Bacteria as an MRI Contrast Agent for In Vivo Tracking of Adoptively Transferred Immune Cells. Mol Imaging Biol 2023; 25:844-856. [PMID: 37715090 DOI: 10.1007/s11307-023-01849-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE In vivo immune cell tracking using MRI can be a valuable tool for studying the mechanisms underlying successful cancer therapies. Current cell labeling methods using superparamagnetic iron oxide (SPIO) lack the persistence to track the fate and location of transplanted cells long-term. Magnetospirillum magneticum is a commercially available, iron-producing bacterium that can be taken up by and live harmoniously within mammalian cells as magneto-endosymbionts (MEs). MEs have shown promise as labeling agents for in vivo stem and cancer cell tracking but have yet to be evaluated in immune cells. This pilot study examined ME labeling in myeloid-derived suppressor cells (MDSCs), cytotoxic T lymphocytes (CTLs), and dendritic cells (DCs) and its effects on cell purity, function, and MRI contrast. PROCEDURES MDSCs, CTLs, and DCs were incubated with MEs at various ME labeling ratios (MLR), and various biological metrics and iron uptake were assessed. For in vivo imaging, MDSCs were labeled overnight with either MEs or SPIO (Molday ION Rhodamine B) and injected into C3 tumor-bearing mice via tail vein injection 24 days post-implant and scanned daily with MRI for 1 week to assess cellular quantification. RESULTS Following incubations, MDSCs contained > 0.6 pg Fe/cell. CTLs achieved Fe loading of < 0.5 pg/cell, and DCs achieved Fe loading of ~ 1.4 pg/cell. The suppressive functionality of MDSCs at 1000 MLR was not affected by ME labeling but was affected at 2000 MLR. Markers of CTL dysfunction were not markedly affected by ME labeling nor were DC markers. In vivo data demonstrated that the MDSCs labeled with MEs generated sufficient contrast to be detectable using TurboSPI, similar to SPIO-labeled cells. CONCLUSIONS Cells can be labeled with sufficient numbers of MEs to be detectable with MRI without compromising cell viability. Care must be taken at higher concentrations of MEs, which may affect some cell types' functional activity and/or morphology. Immune cells with minimal phagocytic behavior have much lower iron content per cell after incubation with MEs vs SPIO; however, MEs can successfully be used as a contrast agent for phagocytic immune cells.
Collapse
Affiliation(s)
- Andrea Nuschke
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
| | - Caitrin Sobey-Skelton
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Bassel Dawod
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Brianna Kelly
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Marie-Laurence Tremblay
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Christa Davis
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
| | - James A Rioux
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada
- Department of Physics, Dalhousie University, Halifax, NS, Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, NS, Canada
| | - Kimberly Brewer
- Biomedical MRI Research Laboratory, IWK Health Centre, Halifax, NS, Canada.
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.
- Department of Physics, Dalhousie University, Halifax, NS, Canada.
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS, Canada.
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
3
|
Dechsupa N, Kosintarajit P, Kamkan K, Khanjina T, Sirikul C, Innuan P, Suwan A, Anukul N, Kantapan J. Iron(III)-Quercetin Complexes' Safety for MRI Cell Tracking in Cell Therapy Applications: Cytotoxic and Genotoxic Assessment. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2776. [PMID: 36014641 PMCID: PMC9414527 DOI: 10.3390/nano12162776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
The theranostic agent iron-quercetin complex (IronQ) provides a T1-positive magnetic resonance imaging (MRI) contrast agent. The magnetically IronQ-labeled cells can be used for cell tracking and have active biological applications in promoting cell and tissue regeneration. However, a detailed investigation of IronQ's cytotoxicity and genotoxicity is necessary. Thus, this study aimed to evaluate the possibility of IronQ inducing cytotoxicity and genotoxicity in peripheral blood mononuclear cells (PBMCs). We evaluated the vitality of cells, the production of reactive oxygen species (ROS), the level of antioxidant enzymes, and the stability of the genetic material in PBMCs treated with IronQ. The results show that IronQ had a negligible impact on toxicological parameters such as ROS production and lipid peroxidation, indicating that it is not harmful. IronQ-labeled PMBCs experienced an insignificant depletion of antioxidant enzyme levels at the highest concentration of IronQ. There is no evident genotoxicity in the magnetically IronQ-labeled PBMCs. The results show that IronQ does not potentiate the cytotoxicity and genotoxicity effects of the labeled PMBCs and might be safe for therapeutic and cell tracking purposes. These results could provide a reference guideline for the toxicological analysis of IronQ in in vivo studies.
Collapse
Affiliation(s)
- Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Radiation Research and Medical Imaging, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Panida Kosintarajit
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanyapak Kamkan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thanyalak Khanjina
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chonticha Sirikul
- Division of Transfusion Science, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phattarawadee Innuan
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Authaphinya Suwan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nampeung Anukul
- Division of Transfusion Science, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Radiation Research and Medical Imaging, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Makela AV, Schott MA, Madsen CS, Greeson EM, Contag CH. Magnetic Particle Imaging of Magnetotactic Bacteria as Living Contrast Agents Is Improved by Altering Magnetosome Arrangement. NANO LETTERS 2022; 22:4630-4639. [PMID: 35686930 DOI: 10.1021/acs.nanolett.1c05042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) can be used as imaging agents to differentiate between normal and diseased tissue or track cell movement. Magnetic particle imaging (MPI) detects the magnetic properties of SPIONs, providing quantitative and sensitive image data. MPI performance depends on the size, structure, and composition of nanoparticles. Magnetotactic bacteria produce magnetosomes with properties similar to those of synthetic nanoparticles, and these can be modified by mutating biosynthetic genes. The use of Magnetospirillum gryphiswaldense, MSR-1 with a mamJ deletion, containing clustered magnetosomes instead of typical linear chains, resulted in improved MPI signal and resolution. Bioluminescent MSR-1 with the mamJ deletion were administered into tumor-bearing and healthy mice. In vivo bioluminescence imaging revealed the viability of MSR-1, and MPI detected signals in livers and tumors. The development of living contrast agents offers opportunities for imaging and therapy with multimodality imaging guiding development of these agents by tracking the location, viability, and resulting biological effects.
Collapse
Affiliation(s)
- Ashley V Makela
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
| | - Melissa A Schott
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
| | - Cody S Madsen
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Emily M Greeson
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
5
|
Melo KP, Makela AV, Knier NN, Hamilton AM, Foster PJ. Magnetic microspheres can be used for magnetic particle imaging of cancer cells arrested in the mouse brain. Magn Reson Med 2021; 87:312-322. [PMID: 34453462 DOI: 10.1002/mrm.28987] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE Magnetic particle imaging (MPI) is a new imaging modality that sensitively and specifically detects superparamagnetic iron oxide nanoparticles (SPIOs). MRI cell tracking with SPIOs has very high sensitivity, but low specificity and quantification is difficult. MPI could overcome these limitations. There are no reports of micron-sized iron oxide particles (MPIO) for cell tracking by MPI. Therefore, the goal was to evaluate if MPIO can be used for in vivo detection and quantification of cancer cells distributed in the mouse brain by MPI. METHODS In the first experiment mice were injected with either 2.5 × 105 or 5.0 × 105 MPIO-labeled cancer cells and MPI was performed ex vivo. In a second experiment, mice received either 2.5 × 105 or 5.0 × 104 MPIO-labeled cells and MPI was performed in vivo. In a third experiment, mice were injected with 5.0 × 104 cells, labeled with either MPIO or ferucarbotran, and MPI was performed in vivo. RESULTS MPIO-labeled cells were visible in all MPI images of the mouse brain. The MPI signal and iron content measurements were greater for brains of mice that were injected with higher numbers of MPIO-labeled cells. Ferucarbotran-labeled cells were not detected in the brain by MPI. CONCLUSION This is the first example of the use of MPIO for cell tracking with MPI. With an intracardiac cell injection, ~15% of cells will arrest in the brain vasculature. For our lowest cell injection of 5.0 × 104 cells, this was ~10 000 cells, distributed throughout the brain.
Collapse
Affiliation(s)
- Kierstin P Melo
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Ashley V Makela
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan, USA
| | - Natasha N Knier
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Amanda M Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
6
|
Makela AV, Gaudet JM, Schott MA, Sehl OC, Contag CH, Foster PJ. Magnetic Particle Imaging of Macrophages Associated with Cancer: Filling the Voids Left by Iron-Based Magnetic Resonance Imaging. Mol Imaging Biol 2021; 22:958-968. [PMID: 31933022 DOI: 10.1007/s11307-020-01473-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Magnetic particle imaging (MPI) is an emerging molecular imaging technique that directly detects iron nanoparticles distributed in living subjects. Compared with imaging iron with magnetic resonance imaging (MRI), MPI signal can be measured to determine iron content in specific regions. In this paper, the detection of iron-labeled macrophages associated with cancer by MRI and MPI was compared. PROCEDURES Imaging was performed on 4T1 tumor-bearing mice 16-21 days post-cancer cell implantation, 24 h after intravenous injection of Ferucarbotran, a superparamagnetic iron oxide (SPIO) or Ferumoxytol, an ultra-small SPIO. Images of living mice were acquired on a 3T clinical MRI (General Electric, n = 6) or MPI (Magnetic Insight, n = 10) system. After imaging, tumors and lungs were removed, imaged by MPI and examined by histology. RESULTS MRI signal voids were observed within all tumors. In vivo, MPI signals were observed in the tumors of 4 of 5 mice after the administration of each contrast agent and in all excised tumors. Signal voids visualized by MRI were more apparent in tumors of mice injected with Ferumoxytol than those that received Ferucarbotran; this was consistent with iron content measured by MPI. Signal voids relating to macrophage uptake of iron were not detected in lungs by MRI, since air also appears hypointense. In vivo, MPI could not differentiate between iron in the lungs vs the high signal from iron in the liver. However, once the lungs were excised, MPI signal was detectable and quantifiable. Histologic examination confirmed iron within macrophages present in the tumors. CONCLUSIONS MPI provides quantitative information on in vivo iron labeling of macrophages that is not attainable with MRI. The optimal iron nanoparticle for MPI in general is still under investigation; however, for MPI imaging of macrophages labeled in vivo by intravenous administration, Ferumoxytol nanoparticles were superior to Ferucarbotran.
Collapse
Affiliation(s)
- Ashley V Makela
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA. .,Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Jeffrey M Gaudet
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA.,Magnetic Insight Inc, Alameda, CA, USA
| | - Melissa A Schott
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Olivia C Sehl
- Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Christopher H Contag
- The Institute for Quantitative Health Science & Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, MI, 48824, USA.,Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Paula J Foster
- Robarts Research Institute and Department of Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
7
|
Parkins KM, Melo KP, Chen Y, Ronald JA, Foster PJ. Visualizing tumour self-homing with magnetic particle imaging. NANOSCALE 2021; 13:6016-6023. [PMID: 33683241 DOI: 10.1039/d0nr07983a] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Due to their innate tumour homing capabilities, in recent years, circulating tumour cells (CTCs) have been engineered to express therapeutic genes for targeted treatment of primary and metastatic lesions. Additionally, previous studies have incorporated optical or PET imaging reporter genes to enable noninvasive monitoring of therapeutic CTCs in preclinical tumour models. An alternative method for tracking cells is to pre-label them with imaging probes prior to transplantation into the body. This is typically more sensitive to low numbers of cells since large amounts of probe can be concentrated in each cell. The objective of this work was to evaluate magnetic particle imaging (MPI) for the detection of iron-labeled experimental CTCs. CTCs were labeled with micro-sized iron oxide (MPIO) particles, administered via intra-cardiac injection in tumour bearing mice and were detected in the tumour region of the mammary fat pad. Iron content and tumour volumes were calculated. Ex vivo MPI of the tumours and immunohistochemistry were used to validate the imaging data. Here, we demonstrate for the first time the ability of MPI to sensitively detect systemically administered iron-labeled CTCs and to visualize tumour self-homing in a murine model of human breast cancer.
Collapse
Affiliation(s)
- Katie M Parkins
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | |
Collapse
|
8
|
Rizzo S, Padelli F, Rinaldi E, Gioeni D, Aquino D, Brizzola S, Acocella F, Spaggiari L, Baggi F, Bellomi M, Bruzzone MG, Petrella F. 7-T MRI tracking of mesenchymal stromal cells after lung injection in a rat model. Eur Radiol Exp 2020; 4:54. [PMID: 33029694 PMCID: PMC7541802 DOI: 10.1186/s41747-020-00183-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/04/2020] [Indexed: 01/01/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are able to migrate and engraft at sites of inflammation, injuries, and tumours, but little is known about their fate after local injection. The purpose of this study is to perform MSC tracking, combining in vivo 7-T magnetic resonance imaging (MRI) and histological assessment, following lung injection in a rat model. Methods Five lungs were injected with ferumoxide-labelled MSCs and five with perfluorocarbon-labelled MSCs and underwent 7-T MRI. MRI acquisitions were recorded immediately (T0), at 24 h (T24) and/or 48 h (T48) after injection. For each rat, labelled cells were assessed in the main organs by MRI. Target organs were harvested under sterile conditions from rats sacrificed 0, 24, or 48 h after injection and fixed for histological analysis via confocal and structured illumination microscopy. Results Ferumoxide-labelled MSCs were not detectable in the lungs, whereas they were not visible in the distant sites. Perfluorocarbon-labelled MSCs were seen in 5/5 injected lungs at T0, in 1/2 at T24, and in 1/3 at T48. The fluorine signal in the liver was seen in 3/5 at T0, in 1/2 at T24, and in 2/3 at T48. Post-mortem histology confirmed the presence of MSCs in the injected lung. Conclusions Ferumoxide-labelled cells were not seen at distant sites; a linear decay of injected perfluorocarbon-labelled MSCs was observed at T0, T24, and T48 in the lung. In more than half of the experiments, perfluorocarbon-labelled MSCs scattering to the liver was observed, with a similar decay over time as observed in the lung.
Collapse
Affiliation(s)
- Stefania Rizzo
- Imaging Institute of the Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900, Lugano, Switzerland. .,Facoltà di Scienze biomediche, Università della Svizzera italiana (USI), Via G. Buffi 13, 6904, Lugano, Switzerland. .,Clinica di Radiologia EOC, Istituto di Imaging della Svizzera Italiana (IIMSI), via Tesserete 46, 6900, Lugano, Switzerland.
| | - Francesco Padelli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Rinaldi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Gioeni
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milan, Italy
| | - Domenico Aquino
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefano Brizzola
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Fabio Acocella
- Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Spaggiari
- Department of Thoracic Surgery, IRCCS European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Fulvio Baggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Massimo Bellomi
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Department of Radiology, IRCCS European Institute of Oncology, Milan, Italy
| | - Maria Grazia Bruzzone
- Department of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Francesco Petrella
- Department of Thoracic Surgery, IRCCS European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,CRC StaMeTec Università degli studi di Milano, Milan, Italy
| |
Collapse
|
9
|
Reichardt W, von Elverfeldt D. Preclinical Applications of Magnetic Resonance Imaging in Oncology. Recent Results Cancer Res 2020; 216:405-437. [PMID: 32594394 DOI: 10.1007/978-3-030-42618-7_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The evolving possibilities of molecular imaging (MI) are fundamentally changing the way we look at cancer, with imaging paradigms now shifting away from basic morphological measures toward the longitudinal assessment of functional, metabolic, cellular, and molecular information in vivo. Recent developments of imaging methodology and probe molecules utilizing the vast number of novel animal models of human cancers have enhanced our ability to non-invasively characterize neoplastic tissue and follow anticancer treatments. While preclinical molecular imaging offers a whole palette of excellent methodology to choose from, we will focus on magnetic resonance imaging (MRI) techniques, since they provide excellent molecular imaging capabilities and bear high potential for clinical translation. Prerequisites and consequences of using animal models as surrogates of human cancers in preclinical molecular imaging are outlined. We present physical principles, values, and limitations of MRI as molecular imaging modality and comment on its high potential to non-invasively assess information on metabolism, hypoxia, angiogenesis, and cell trafficking in preclinical cancer research.
Collapse
Affiliation(s)
- Wilfried Reichardt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Dominik von Elverfeldt
- Medical Physics, Department of Radiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
Sehl OC, Makela AV, Hamilton AM, Foster PJ. Trimodal Cell Tracking In Vivo: Combining Iron- and Fluorine-Based Magnetic Resonance Imaging with Magnetic Particle Imaging to Monitor the Delivery of Mesenchymal Stem Cells and the Ensuing Inflammation. ACTA ACUST UNITED AC 2020; 5:367-376. [PMID: 31893235 PMCID: PMC6935990 DOI: 10.18383/j.tom.2019.00020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The therapeutic potential of mesenchymal stem cells (MSCs) is limited, as many cells undergo apoptosis following administration. In addition, the attraction of immune cells (predominately macrophages) to the site of implantation can lead to MSC rejection. We implemented a trimodal imaging technique to monitor the fate of transplanted MSCs and infiltrating macrophages in vivo. MSCs were labeled with an iron oxide nanoparticle (ferumoxytol) and then implanted within the hind limb muscle of 10 C57BI/6 mice. Controls received unlabeled MSCs (n = 5). A perfluorocarbon agent was administered intravenously for uptake by phagocytic macrophages in situ; 1 and 12 days later, the ferumoxytol-labeled MSCs were detected by proton (1H) magnetic resonance imaging (MRI) and magnetic particle imaging (MPI). Perfluorocarbon-labeled macrophages were detected by fluorine-19 (19F) MRI. 1H/19F MRI was acquired on a clinical scanner (3 T) using a dual-tuned surface coil and balanced steady-state free precession (bSSFP) sequence. The measured volume of signal loss and MPI signal declined over 12 days, which is consistent with the death and clearance of iron-labeled MSCs. 19F signal persisted over 12 days, suggesting the continuous infiltration of perfluorocarbon-labeled macrophages. Because MPI and 19F MRI signals are directly quantitative, we calculated estimates of the number of MSCs and macrophages present over time. The presence of MSCs and macrophages was validated with histology following the last imaging session. This is the first study to combine the use of iron- and fluorine-based MRI with MPI cell tracking.
Collapse
Affiliation(s)
- Olivia C Sehl
- Imaging Research Laboratories, Robarts Research Institute and.,Department of Medical Biophysics, University of Western Ontario, London, ON, Canada; and
| | - Ashley V Makela
- The Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI
| | | | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute and.,Department of Medical Biophysics, University of Western Ontario, London, ON, Canada; and
| |
Collapse
|
11
|
Nejadnik H, Jung KO, Theruvath AJ, Kiru L, Liu A, Wu W, Sulchek T, Pratx G, Daldrup-Link HE. Instant labeling of therapeutic cells for multimodality imaging. Theranostics 2020; 10:6024-6034. [PMID: 32483435 PMCID: PMC7255004 DOI: 10.7150/thno.39554] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/03/2020] [Indexed: 12/21/2022] Open
Abstract
Autologous therapeutic cells are typically harvested and transplanted in one single surgery. This makes it impossible to label them with imaging biomarkers through classical transfection techniques in a laboratory. To solve this problem, we developed a novel microfluidic device, which provides highly efficient labeling of therapeutic cells with imaging biomarkers through mechanoporation. Methods: Studies were performed with a new, custom-designed microfluidic device, which contains ridges, which compress adipose tissue-derived stem cells (ADSCs) during their device passage. Cell relaxation after compression leads to cell volume exchange for convective transfer of nanoparticles and nanoparticle uptake into the cell. ADSCs were passed through the microfluidic device doped with iron oxide nanoparticles and 18F-fluorodeoxyglucose (FDG). The cellular nanoparticle and radiotracer uptake was evaluated with DAB-Prussian blue, fluorescent microscopy, and inductively coupled plasma spectrometry (ICP). Labeled and unlabeled ADSCs were imaged in vitro as well as ex vivo in pig knee specimen with magnetic resonance imaging (MRI) and positron emission tomography (PET). T2 relaxation times and radiotracer signal were compared between labeled and unlabeled cell transplants using Student T-test with p<0.05. Results: We report significant labeling of ADSCs with iron oxide nanoparticles and 18F-FDG within 12+/-3 minutes. Mechanoporation of ADSCs with our microfluidic device led to significant nanoparticle (> 1 pg iron per cell) and 18F-FDG uptake (61 mBq/cell), with a labeling efficiency of 95%. The labeled ADSCs could be detected with MRI and PET imaging technologies: Nanoparticle labeled ADSC demonstrated significantly shorter T2 relaxation times (24.2±2.1 ms) compared to unlabeled cells (79.6±0.8 ms) on MRI (p<0.05) and 18F-FDG labeled ADSC showed significantly higher radiotracer uptake (614.3 ± 9.5 Bq / 1×104 cells) compared to controls (0.0 ± 0.0 Bq/ 1×104 cells) on gamma counting (p<0.05). After implantation of dual-labeled ADSCs into pig knee specimen, the labeled ADSCs revealed significantly shorter T2 relaxation times (41±0.6 ms) compared to unlabeled controls (90±1.8 ms) (p<0.05). Conclusion: The labeling of therapeutic cells with our new microfluidic device does not require any chemical intervention, therefore it is broadly and immediately clinically applicable. Cellular labeling using mechanoporation can improve our understanding of in vivo biodistributions of therapeutic cells and ultimately improve long-term outcomes of therapeutic cell transplants.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
| | - Kyung Oh Jung
- Department of Radiation Oncology, Stanford University, CA, 94305, USA
| | - Ashok J. Theruvath
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Louise Kiru
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
| | - Anna Liu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
| | - Todd Sulchek
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Guillem Pratx
- Department of Radiation Oncology, Stanford University, CA, 94305, USA
| | - Heike E. Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, CA, 94305, USA
- Department of Pediatrics, Stanford University, CA, 94305, USA
| |
Collapse
|
12
|
Bıyık E, Keskin K, Uh Dar S, Koç A, Çukur T. Factorized sensitivity estimation for artifact suppression in phase-cycled bSSFP MRI. NMR IN BIOMEDICINE 2020; 33:e4228. [PMID: 31985879 DOI: 10.1002/nbm.4228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Balanced steady-state free precession (bSSFP) imaging suffers from banding artifacts in the presence of magnetic field inhomogeneity. The purpose of this study is to identify an efficient strategy to reconstruct banding-free bSSFP images from multi-coil multi-acquisition datasets. METHOD Previous techniques either assume that a naïve coil-combination is performed a priori resulting in suboptimal artifact suppression, or that artifact suppression is performed for each coil separately at the expense of significant computational burden. Here we propose a tailored method that factorizes the estimation of coil and bSSFP sensitivity profiles for improved accuracy and/or speed. RESULTS In vivo experiments show that the proposed method outperforms naïve coil-combination and coil-by-coil processing in terms of both reconstruction quality and time. CONCLUSION The proposed method enables computationally efficient artifact suppression for phase-cycled bSSFP imaging with modern coil arrays. Rapid imaging applications can efficiently benefit from the improved robustness of bSSFP imaging against field inhomogeneity.
Collapse
Affiliation(s)
- Erdem Bıyık
- Department of Electrical Engineering, Stanford University, CA, USA
- Intelligent Data Analytics Research Program Department, Aselsan Research Center, Ankara, Turkey
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey
| | - Kübra Keskin
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Salman Uh Dar
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Aykut Koç
- Intelligent Data Analytics Research Program Department, Aselsan Research Center, Ankara, Turkey
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
| | - Tolga Çukur
- Department of Electrical and Electronics Engineering, Bilkent University, Ankara, Turkey
- National Magnetic Resonance Research Center (UMRAM), Bilkent University, Ankara, Turkey
- Neuroscience Program at Sabuncu Brain Research Center, Bilkent University, Ankara, Turkey
| |
Collapse
|
13
|
O'Brien-Moran Z, Bowen CV, Rioux JA, Brewer KD. Cell density quantification with TurboSPI: R 2* mapping with compensation for off-resonance fat modulation. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2019; 33:469-481. [PMID: 31872356 DOI: 10.1007/s10334-019-00817-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Tracking the migration of superparamagnetic iron oxide (SPIO)-labeled immune cells in vivo is valuable for understanding the immunogenic response to cancer and therapies. Quantitative cell tracking using TurboSPI-based R2* mapping is a promising development to improve accuracy in longitudinal studies on immune recruitment. However, off-resonance fat signal isochromats lead to modulations in the signal time-course that can be erroneously fit as R2* signal decay, overestimating the density of labeled cells, while excluding voxels with fat-typical modulations results in underestimation of cell density in voxels with mixed content. Approaches capable of accurate R2* estimation in the presence of fat are needed. METHODS We propose a dual-decay (separate R2f* and R2w* for fat and water) Dixon-based signal model that accounts for the presence of fat in a voxel to provide better estimates of SPIO-induced dephasing. This model was tested in silico, in phantoms with varying quantities of fat and SPIO-labeled cells, and in 5 mice injected with SPIO-labeled CD8+ T cells. RESULTS In silico single voxel simulations illustrate how the proposed dual-decay model provides stable R2w* estimates that are invariant to fat content. The proposed model outperforms previous methods when applied to in vitro samples of SPIO-labeled cells and oil prepared with oil content ≥ 15%. Preliminary in vivo results show that, compared to previous methods, the dual-decay model improves the balance of R2* mapping in fat-dense areas, which will yield more reliable analysis in future cell tracking studies. DISCUSSION The proposed model is a promising tool for quantitative TurboSPI R2* cell tracking, with further refinements offering the possibility of better specificity and sensitivity.
Collapse
Affiliation(s)
- Zoe O'Brien-Moran
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada.,Dalhousie University, Halifax, NS, Canada
| | - Chris Van Bowen
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada.,Dalhousie University, Halifax, NS, Canada
| | - James Allen Rioux
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada.,Dalhousie University, Halifax, NS, Canada
| | - Kimberly Dawn Brewer
- Biomedical Translational Imaging Centre, IWK Health Centre and Nova Scotia Health Authority, Halifax, NS, Canada. .,Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
14
|
Riberdy V, Litvack M, Stirrat E, Couch M, Post M, Santyr GE. Hyperpolarized 129 Xe imaging of embryonic stem cell-derived alveolar-like macrophages in rat lungs: proof-of-concept study using superparamagnetic iron oxide nanoparticles. Magn Reson Med 2019; 83:1356-1367. [PMID: 31556154 DOI: 10.1002/mrm.27999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/18/2019] [Accepted: 08/25/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE To measure regional changes in hyperpolarized 129 Xe MRI signal and apparent transverse relaxation ( T 2 ∗ ) because of instillation of SPION-labeled alveolar-like macrophages (ALMs) in the lungs of rats and compare to histology. METHODS MRI was performed in 6 healthy mechanically ventilated rats before instillation, as well as 5 min and 1 h after instillation of 4 million SPION-labeled ALMs into either the left or right lung. T 2 ∗ maps were calculated from 2D multi-echo data at each time point and changes in T 2 ∗ were measured and compared to control rats receiving 4 million unlabeled ALMs. Histology of the ex vivo lungs was used to compare the regional MRI findings with the locations of the SPION-labeled ALMs. RESULTS Regions of signal loss were observed immediately after instillation of unlabeled and SPION-labeled ALMs and persisted at least 1 h in the case of the SPION-labeled ALMs. This was reflected in the measurements of T 2 ∗ . One hour after the instillation of SPION-labeled ALMs, the T 2 ∗ decreased to 54.0 ± 7.0% of the baseline, compared to a full recovery to baseline after the instillation of unlabeled ALMs. Histology confirmed the co-localization of SPION-labeled ALMs with regions of signal loss and T 2 ∗ decreases for each rat. CONCLUSION Hyperpolarized 129 Xe MRI can detect the presence of SPION-labeled ALMs in the airways 1 h after instillation. This approach is promising for targeting and tracking of stem cells for the treatment of lung disease.
Collapse
Affiliation(s)
- Vlora Riberdy
- Translational Medicine Program, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael Litvack
- Translational Medicine Program, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Elaine Stirrat
- Translational Medicine Program, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, Canada
| | - Marcus Couch
- Translational Medicine Program, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Giles E Santyr
- Translational Medicine Program, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
15
|
MR and PET-CT monitoring of tissue-engineered vascular grafts in the ovine carotid artery. Biomaterials 2019; 216:119228. [DOI: 10.1016/j.biomaterials.2019.119228] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/16/2019] [Accepted: 05/25/2019] [Indexed: 12/19/2022]
|
16
|
Bär S, Oerther T, Weigel M, Müller A, Hucker P, Korvink JG, Ko CW, Wapler MC, Leupold J. On the application of balanced steady-state free precession to MR microscopy. MAGMA (NEW YORK, N.Y.) 2019; 32:437-447. [PMID: 30649708 DOI: 10.1007/s10334-019-00736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/30/2018] [Accepted: 01/03/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVE The applicability of the balanced steady-state free precession (bSSFP) sequence to the field of MR microscopy was investigated, since the potentially high SNR makes bSSFP attractive. However, particularly at ultra-high magnetic fields, a number of constraints emerge: the frequency sensitivity of the bSSFP signal, the duty cycle of the imaging gradients, and the intrinsic diffusion attenuation of the steady state due to the imaging gradients. MATERIALS AND METHODS Optimization of the bSSFP sequence was performed on three imaging systems (7 T and 9.4 T) suited for MR microscopy. Since biological samples are often imaged in the very proximity of materials from sample containers/holder or devices such as electrodes, several microscopy phantoms representing such circumstances were fabricated and examined with 3D bSSFP. RESULTS Artifact-free microscopic bSSFP images could be obtained with voxel sizes down to 16 µm × 16 µm × 78 µm and with an SNR gain of 25% over standard gradient echo images. CONCLUSION With appropriate choice of phantom materials, optimization of the flip angle to the diffusion-attenuated steady state and protocols considering duty-cycle limitations, bSSFP can be a valuable tool in MR microscopy.
Collapse
Affiliation(s)
- Sébastien Bär
- Department of Radiology, Medical Physics, Faculty of Medicine, Medical Center - University of Freiburg, Killianstrasse 5a, 79106, Freiburg, Germany.
- BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Freiburg, Germany.
- Department of Computer Science and Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Thomas Oerther
- Microimaging Applications, Bruker BioSpin GmbH, Rheinstetten, Germany
| | - Matthias Weigel
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, Basel, Switzerland
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Angelina Müller
- Department for Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Patrick Hucker
- Department of Radiology, Medical Physics, Faculty of Medicine, Medical Center - University of Freiburg, Killianstrasse 5a, 79106, Freiburg, Germany
| | - Jan G Korvink
- Institute of Microstructure Technology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Cheng-Wen Ko
- Department of Computer Science and Engineering, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Matthias C Wapler
- Department for Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Jochen Leupold
- Department of Radiology, Medical Physics, Faculty of Medicine, Medical Center - University of Freiburg, Killianstrasse 5a, 79106, Freiburg, Germany
- BrainLinks-BrainTools Cluster of Excellence, University of Freiburg, Freiburg, Germany
| |
Collapse
|
17
|
Cellular MRI Reveals Altered Brain Arrest of Genetically Engineered Metastatic Breast Cancer Cells. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:6501231. [PMID: 30733649 PMCID: PMC6348811 DOI: 10.1155/2019/6501231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/15/2018] [Accepted: 12/04/2018] [Indexed: 11/29/2022]
Abstract
Purpose The combined use of anatomical magnetic resonance imaging (MRI), cellular MRI, and bioluminescence imaging (BLI) allows for sensitive and improved monitoring of brain metastasis in preclinical cancer models. By using these complementary technologies, we can acquire measurements of viable single cell arrest in the brain after systemic administration, the clearance and/or retention of these cells thereafter, the growth into overt tumours, and quantification of tumour volume and relative cancer cell viability over time. While BLI is very useful in measuring cell viability, some considerations have been reported using cells engineered with luciferase such as increased tumour volume variation, changes in pattern of metastatic disease, and inhibition of in vivo tumour growth. Procedures Here, we apply cellular and anatomical MRI to evaluate in vivo growth differences between iron oxide labeled naïve (4T1BR5) and luciferase-expressing (4T1BR5-FLuc-GFP) murine brain-seeking breast cancer cells. Balb/C mice received an intracardiac injection of 20,000 cells and were imaged with MRI on days 0 and 14. Mice that received 4T1BR5-FLuc-GFP cells were also imaged with BLI on days 0 and 14. Results The number of signal voids in the brain (representing iron-labeled cancer cells) on day 0 was significantly higher in mice receiving 4T1BR5 cells compared to mice receiving 4T1BR5-FLuc-GFP cells (p < 0.0001). Mice that received 4T1BR5 cells also had significantly higher total brain tumour burden and number of brain metastases than mice that received 4T1BR5-FLuc-GFP cells (p < 0.0001). Conclusions By employing highly sensitive cellular MRI tools, we demonstrate that engineered cells did not form tumours as well as their naïve counterparts, which appear to primarily be due to a reduction in cell arrest. These results indicate that engineering cancer cells with reporter genes may alter their tropism towards particular organs and highlight another important consideration for research groups that use reporter gene imaging to track metastatic cancer cell fate in vivo.
Collapse
|
18
|
Rosenberg JT, Yuan X, Helsper SN, Bagdasarian FA, Ma T, Grant SC. Effects of labeling human mesenchymal stem cells with superparamagnetic iron oxides on cellular functions and magnetic resonance contrast in hypoxic environments and long-term monitoring. Brain Circ 2018; 4:133-138. [PMID: 30450421 PMCID: PMC6187941 DOI: 10.4103/bc.bc_18_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/27/2018] [Accepted: 09/10/2018] [Indexed: 01/25/2023] Open
Abstract
Ischemia, which involves decreased blood flow to a region and a corresponding deprivation of oxygen and nutrients, can be induced as a consequence of stroke or heart attack. A prevalent disease that affects many individuals worldwide, ischemic stroke results in functional and cognitive impairments, as neural cells in the brain receive inadequate nourishment and encounter inflammation and various other detrimental toxic factors that lead to their death. Given the scarce treatments for this disease in the clinic such as the administration of tissue plasminogen activator, which is only effective in a limited time window after the occurrence of stroke, it will be necessary to develop new strategies to ameliorate or prevent stroke-induced brain damage. Cell-based therapies appear to be a promising solution for treating ischemic stroke and many other ischemia-associated and neurodegenerative maladies. Particularly, human mesenchymal stem cells (hMSCs) are of interest for cell transplantation in stroke, given their multipotency, accessibility, and reparative abilities. To determine the fate and survival of hMSC, which will be imperative for successful transplantation therapies, these cells may be monitored using magnetic resonance imaging and transfected with superparamagnetic iron oxide (SPIO), a contrast agent that facilitates the detection of these hMSCs. This review encompasses pertinent research and findings to reveal the effects of SPIO on hMSC functions in the context of transplantation in ischemic environments and over extended time periods. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors' experiences.
Collapse
Affiliation(s)
- Jens T Rosenberg
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| | - Shannon N Helsper
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| | - F Andrew Bagdasarian
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| | - Samuel C Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
19
|
Zarghami N, Khrapitchev AA, Perez-Balderas F, Soto MS, Larkin JR, Bau L, Sibson NR. Optimization of molecularly targeted MRI in the brain: empirical comparison of sequences and particles. Int J Nanomedicine 2018; 13:4345-4359. [PMID: 30100719 PMCID: PMC6064157 DOI: 10.2147/ijn.s158071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Molecular MRI is an evolving field of research with strong translational potential. Selection of the appropriate MRI sequence, field strength and contrast agent depend largely on the application. The primary aims of the current study were to: 1) assess the sensitivity of different MRI sequences for detection of iron oxide particles in mouse brain; 2) determine the effect of magnetic field strength on detection of iron oxide particles in vivo; and 3) compare the sensitivity of targeted microparticles of iron oxide (MPIO) or ultra-small superparamagnetic iron oxide (USPIO) for detection of vascular cell adhesion molecule-1 (VCAM-1) in vivo. METHODS Mice were injected intrastriatally with interleukin 1β to induce VCAM-1 expression on the cerebral vasculature. Subsequently, animals were injected intravenously with either VCAM-MPIO or VCAM-USPIO and imaged 1 or 13 hours post-injection, respectively. MRI was performed at 4.7, 7.0, or 9.4 T, using three different T2*-weighted sequences: single gradient echo 3D (GE3D), multi-gradient echo 3D (MGE3D) and balanced steady-state free precession 3D (bSSFP3D). RESULTS MGE3D yielded the highest signal-to-noise ratio (SNR) and contrast-to-noise ratio (CNR) for the detection of iron oxide particles. All sequences showed a significant increase in SNR and CNR from 4.7 to 7.0 T, but no further improvement at 9.4 T. However, whilst targeted MPIO enabled sensitive detection of VCAM-1 expression on the cerebral vasculature, the long half-life (16.5 h vs 1.2 min) and lower relaxivity per particle (1.29×10-14 vs 1.18×10-9 Hz L/particle) of USPIO vs. MPIO rendered them impractical for molecular MRI. CONCLUSION These findings demonstrate clear advantages of MPIO compared to USPIO for molecularly-targeted MRI, and indicate that the MGE3D sequence is optimal for MPIO detection. Moreover, higher field strengths (7.0/9.4 T) showed enhanced sensitivity over lower field strengths (4.7 T). With the development of biodegradable MPIO, these agents hold promise for clinical translation.
Collapse
Affiliation(s)
- Niloufar Zarghami
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK,
| | - Alexandre A Khrapitchev
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK,
| | - Francisco Perez-Balderas
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK,
| | - Manuel Sarmiento Soto
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK,
| | - James R Larkin
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK,
| | - Luca Bau
- Institute of Biomedical Engineering, Department of Engineering Sciences, University of Oxford, Oxford, UK
| | - Nicola R Sibson
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK,
| |
Collapse
|
20
|
Shen Y, Yan L, Shao X, Zhao B, Bai J, Lu W, Wang DJ. Improved sensitivity of cellular MRI using phase-cycled balanced SSFP of ferumoxytol nanocomplex-labeled macrophages at ultrahigh field. Int J Nanomedicine 2018; 13:3839-3852. [PMID: 30013339 PMCID: PMC6039059 DOI: 10.2147/ijn.s169860] [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] [Indexed: 01/12/2023] Open
Abstract
Purpose The purpose of this study was to investigate the feasibility and sensitivity of cellular magnetic resonance imaging (MRI) with ferumoxytol nanocomplex-labeled macrophages at ultrahigh magnetic field of 7 T. Materials and methods THP-1-induced macrophages were labeled using self-assembling heparin + protamine + ferumoxytol nanocomplexes which were injected into a gelatin phantom visible on both microscope and MRI. Susceptibility-weighted imaging (SWI) and balanced steady-state free precession (bSSFP) pulse sequences were applied at 3 and 7 T. The average, maximum intensity projection, and root mean square combined images were generated for phase-cycled bSSFP images. The signal-to-noise ratio and contrast-to-noise ratio (CNR) efficiencies were calculated. Ex vivo experiments were then performed using a formalin-fixed pig brain injected witĥ100 and ~1,000 labeled cells, respectively, at both 3 and 7 T. Results A high cell labeling efficiency (.90%) was achieved with heparin + protamine + ferumoxytol nanocomplexes. Less than 100 cells were detectable in the gelatin phantom at both 3 and 7 T. The 7 T data showed more than double CNR efficiency compared to the corresponding sequences at 3 T. The CNR efficiencies of phase-cycled bSSFP images were higher compared to those of SWI, and the root mean square combined bSSFP showed the highest CNR efficiency with minimal banding. Following co-registration of microscope and MR images, more cells (51/63) were detected by bSSFP at 7 T than at 3 T (36/63). On pig brain, botĥ100 and ~1,000 cells were detected at 3 and 7 T. While the cell size appeared larger due to blooming effects on SWI, bSSFP allowed better contrast to precisely identify the location of the cells with higher signal-to-noise ratio efficiency. Conclusion The proposed cellular MRI with ferumoxytol nanocomplex-labeled macrophages at 7 T has a high sensitivity to detect, 100 cells. The proposed method has great translational potential and may have broad clinical applications that involve cell types with a primary phagocytic phenotype.
Collapse
Affiliation(s)
- Yelong Shen
- Laboratory of FMRI Technology (LOFT), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA, .,Shandong Medical Imaging Research Institute, School of Medicine, Shandong University, Jinan, Shangdong, China
| | - Lirong Yan
- Laboratory of FMRI Technology (LOFT), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA,
| | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA,
| | - Bin Zhao
- Shandong Medical Imaging Research Institute, School of Medicine, Shandong University, Jinan, Shangdong, China
| | - Jinlun Bai
- Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA
| | - Wange Lu
- Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA
| | - Danny Jj Wang
- Laboratory of FMRI Technology (LOFT), Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA, USA,
| |
Collapse
|
21
|
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
|
22
|
Parkins KM, Dubois VP, Hamilton AM, Makela AV, Ronald JA, Foster PJ. Multimodality cellular and molecular imaging of concomitant tumour enhancement in a syngeneic mouse model of breast cancer metastasis. Sci Rep 2018; 8:8930. [PMID: 29895974 PMCID: PMC5997674 DOI: 10.1038/s41598-018-27208-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/25/2018] [Indexed: 01/10/2023] Open
Abstract
The mechanisms that influence metastatic growth rates are poorly understood. One mechanism of interest known as concomitant tumour resistance (CTR) can be defined as the inhibition of metastasis by existing tumour mass. Conversely, the presence of a primary tumour has also been shown to increase metastatic outgrowth, termed concomitant tumour enhancement (CTE). The majority of studies evaluating CTR/CTE in preclinical models have relied on endpoint histological evaluation of tumour burden. The goal of this research was to use conventional magnetic resonance imaging (MRI), cellular MRI, and bioluminescence imaging to study the impact of a primary tumour on the development of brain metastases in a syngeneic mouse model. Here, we report that the presence of a 4T1 primary tumour significantly enhances total brain tumour burden in Balb/C mice. Using in vivo BLI/MRI we could determine this was not related to differences in initial arrest or clearance of viable cells in the brain, which suggests that the presence of a primary tumour can increase the proliferative growth of brain metastases in this model. The continued application of our longitudinal cellular and molecular imaging tools will yield a better understanding of the mechanism(s) by which this physiological inhibition (CTR) and/or enhancement (CTE) occurs.
Collapse
Affiliation(s)
- Katie M Parkins
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- The Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada
| | - Veronica P Dubois
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- The Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada
| | - Amanda M Hamilton
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
| | - Ashley V Makela
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- The Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada
| | - John A Ronald
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- The Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Paula J Foster
- Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada.
- The Department of Medical Biophysics, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
23
|
Zarghami N, Murrell DH, Jensen MD, Dick FA, Chambers AF, Foster PJ, Wong E. Half brain irradiation in a murine model of breast cancer brain metastasis: magnetic resonance imaging and histological assessments of dose-response. Radiat Oncol 2018; 13:104. [PMID: 29859114 PMCID: PMC5984731 DOI: 10.1186/s13014-018-1028-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/13/2018] [Indexed: 01/09/2023] Open
Abstract
Background Brain metastasis is becoming increasingly prevalent in breast cancer due to improved extra-cranial disease control. With emerging availability of modern image-guided radiation platforms, mouse models of brain metastases and small animal magnetic resonance imaging (MRI), we examined brain metastases’ responses from radiotherapy in the pre-clinical setting. In this study, we employed half brain irradiation to reduce inter-subject variability in metastases dose-response evaluations. Methods Half brain irradiation was performed on a micro-CT/RT system in a human breast cancer (MDA-MB-231-BR) brain metastasis mouse model. Radiation induced DNA double stranded breaks in tumors and normal mouse brain tissue were quantified using γ-H2AX immunohistochemistry at 30 min (acute) and 11 days (longitudinal) after half-brain treatment for doses of 8, 16 and 24 Gy. In addition, tumor responses were assessed volumetrically with in-vivo longitudinal MRI and histologically for tumor cell density and nuclear size. Results In the acute setting, γ-H2AX staining in tumors saturated at higher doses while normal mouse brain tissue continued to increase linearly in the phosphorylation of H2AX. While γ-H2AX fluorescence intensities returned to the background level in the brain 11 days after treatment, the residual γ-H2AX phosphorylation in the radiated tumors remained elevated compared to un-irradiated contralateral tumors. With radiation, MRI-derived relative tumor growth was significantly reduced compared to the un-irradiated side. While there was no difference in MRI tumor volume growth between 16 and 24 Gy, there was a significant reduction in tumor cell density from histology with increasing dose. In the longitudinal study, nuclear size in the residual tumor cells increased significantly as the radiation dose was increased. Conclusions Radiation damages to the DNAs in the normal brain parenchyma are resolved over time, but remain unrepaired in the treated tumors. Furthermore, there is a radiation dose response in nuclear size of surviving tumor cells. Increase in nuclear size together with unrepaired DNA damage indicated that the surviving tumor cells post radiation had continued to progress in the cell cycle with DNA replication, but failed cytokinesis. Half brain irradiation provides efficient evaluation of dose-response for cancer cell lines, a pre-requisite to perform experiments to understand radio-resistance in brain metastases.
Collapse
Affiliation(s)
- Niloufar Zarghami
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Donna H Murrell
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Michael D Jensen
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Frederick A Dick
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada.,London Regional Cancer Program, University of Western Ontario, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Ann F Chambers
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,London Regional Cancer Program, University of Western Ontario, London, Ontario, Canada.,Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - Paula J Foster
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Eugene Wong
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada. .,London Regional Cancer Program, University of Western Ontario, London, Ontario, Canada. .,Department of Oncology, University of Western Ontario, London, Ontario, Canada. .,Department of Physics and Astronomy, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
24
|
Effects of iron oxide contrast agent in combination with various transfection agents during mesenchymal stem cells labelling: An in vitro toxicological evaluation. Toxicol In Vitro 2018; 50:179-189. [PMID: 29577976 DOI: 10.1016/j.tiv.2018.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 02/02/2018] [Accepted: 03/19/2018] [Indexed: 12/18/2022]
Abstract
The use of iron oxide nanoparticles for different biomedical applications, hold immense promise to develop negative tissue contrast in magnetic resonance imaging (MRI). Previously, we have optimized the labelling of mesenchymal stem cells (MSCs) with iron oxide nanoparticles complexed to different transfection agents like poly-l-lysine (IO-PLL) and protamine sulfate (Fe-Pro) on the basis of relaxation behaviour and its biological expressions. However, there is a distinct need to investigate the biocompatibility and biosafety concerns coupled with its cytotoxicity and genotoxicity. This study was prepared to evaluate the viability of cells, generation of ROS, changes in actin cytoskeleton, investigation of cell death, level of GSH and TAC, activities of SOD and GPx, and stability of DNA in MSCs after labelling. Results demonstrated a marginal alteration in toxicological parameters like ROS generation, cell length, actin cytoskeleton, total apoptosis and DNA damage was detected after stem cell labelling. Insignificant depletion of GSH and SOD level, and increase in GPx and TAC level in MSCs were measured after labelling with IO-PLL and Fe-Pro complexes, which later on recovered and normalized to its baseline. This MSCs labelling could provide a reference guideline for toxicological analysis and relaxometry based in vivo MRI detection.
Collapse
|
25
|
Stem Cell Tracing Through MR Molecular Imaging. Tissue Eng Regen Med 2018; 15:249-261. [PMID: 30603551 DOI: 10.1007/s13770-017-0112-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/09/2017] [Accepted: 12/27/2017] [Indexed: 01/12/2023] Open
Abstract
Stem cell therapy opens a new window in medicine to overcome several diseases that remain incurable. It appears such diseases as cardiovascular disorders, brain injury, multiple sclerosis, urinary system diseases, cartilage lesions and diabetes are curable with stem cell transplantation. However, some questions related to stem cell therapy have remained unanswered. Stem cell imaging allows approval of appropriated strategies such as selection of the type and dose of stem cell, and also mode of cell delivery before being tested in clinical trials. MRI as a non-invasive imaging modality provides proper conditions for this aim. So far, different contrast agents such as superparamagnetic or paramagnetic nanoparticles, ultrasmall superparamagnetic nanoparticles, fluorine, gadolinium and some types of reporter genes have been used for imaging of stem cells. The core subject of these studies is to investigate the survival and differentiation of stem cells, contrast agent's toxicity and long term following of transplanted cells. The promising results of in vivo and some clinical trial studies may raise hope for clinical stem cells imaging with MRI.
Collapse
|
26
|
Makela AV, Foster PJ. Imaging macrophage distribution and density in mammary tumors and lung metastases using fluorine-19 MRI cell tracking. Magn Reson Med 2018; 80:1138-1147. [PMID: 29327789 DOI: 10.1002/mrm.27081] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/09/2017] [Accepted: 12/18/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Ashley V Makela
- Robarts Research Institute, London, Ontario, Canada.,The Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Paula J Foster
- Robarts Research Institute, London, Ontario, Canada.,The Department of Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
27
|
Reduction of T2 Relaxation Rates due to Large Volume Fractions of Magnetic Nanoparticles for All Motional Regimes. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8010101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
28
|
Shen F, Park JK. Toxicity Assessment of Iron Oxide Nanoparticles Based on Cellular Magnetic Loading Using Magnetophoretic Sorting in a Trapezoidal Microchannel. Anal Chem 2017; 90:920-927. [DOI: 10.1021/acs.analchem.7b03875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Fengshan Shen
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Je-Kyun Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro,Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
29
|
Tremblay ML, Davis C, Bowen CV, Stanley O, Parsons C, Weir G, Karkada M, Stanford MM, Brewer KD. Using MRI cell tracking to monitor immune cell recruitment in response to a peptide-based cancer vaccine. Magn Reson Med 2017; 80:304-316. [PMID: 29193231 DOI: 10.1002/mrm.27018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE MRI cell tracking can be used to monitor immune cells involved in the immunotherapy response, providing insight into the mechanism of action, temporal progression of tumor growth, and individual potency of therapies. To evaluate whether MRI could be used to track immune cell populations in response to immunotherapy, CD8+ cytotoxic T cells, CD4+ CD25+ FoxP3+ regulatory T cells, and myeloid-derived suppressor cells were labeled with superparamagnetic iron oxide particles. METHODS Superparamagnetic iron oxide-labeled cells were injected into mice (one cell type/mouse) implanted with a human papillomavirus-based cervical cancer model. Half of these mice were also vaccinated with DepoVaxTM (ImmunoVaccine, Inc., Halifax, Nova Scotia, Canada), a lipid-based vaccine platform that was developed to enhance the potency of peptide-based vaccines. RESULTS MRI visualization of CD8+ cytotoxic T cells, regulatory T cells, and myeloid-derived suppressor cells was apparent 24 h post-injection, with hypointensities due to iron-labeled cells clearing approximately 72 h post-injection. Vaccination resulted in increased recruitment of CD8+ cytotoxic T cells, and decreased recruitment of myeloid-derived suppressor cells and regulatory T cells to the tumor. We also found that myeloid-derived suppressor cell and regulatory T cell recruitment were positively correlated with final tumor volume. CONCLUSION This type of analysis can be used to noninvasively study changes in immune cell recruitment in individual mice over time, potentially allowing improved application and combination of immunotherapies. Magn Reson Med 80:304-316, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
| | - Christa Davis
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Chris V Bowen
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Olivia Stanley
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | - Cathryn Parsons
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada
| | | | - Mohan Karkada
- Wyss Institute at Harvard Medical School, Boston, Massachusetts, USA
| | - Marianne M Stanford
- Immunovaccine Inc., Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kimberly D Brewer
- Biomedical Translational Imaging Centre (BIOTIC), Halifax, Nova Scotia, Canada.,Department of Diagnostic Radiology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada.,School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
30
|
Le TA, Zhang X, Hoshiar AK, Yoon J. Real-Time Two-Dimensional Magnetic Particle Imaging for Electromagnetic Navigation in Targeted Drug Delivery. SENSORS 2017; 17:s17092050. [PMID: 28880220 PMCID: PMC5621117 DOI: 10.3390/s17092050] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/28/2017] [Accepted: 09/01/2017] [Indexed: 11/25/2022]
Abstract
Magnetic nanoparticles (MNPs) are effective drug carriers. By using electromagnetic actuated systems, MNPs can be controlled noninvasively in a vascular network for targeted drug delivery (TDD). Although drugs can reach their target location through capturing schemes of MNPs by permanent magnets, drugs delivered to non-target regions can affect healthy tissues and cause undesirable side effects. Real-time monitoring of MNPs can improve the targeting efficiency of TDD systems. In this paper, a two-dimensional (2D) real-time monitoring scheme has been developed for an MNP guidance system. Resovist particles 45 to 65 nm in diameter (5 nm core) can be monitored in real-time (update rate = 2 Hz) in 2D. The proposed 2D monitoring system allows dynamic tracking of MNPs during TDD and renders magnetic particle imaging-based navigation more feasible.
Collapse
Affiliation(s)
- Tuan-Anh Le
- School of Mechanical and Aerospace Engineering & ReCAPT, Gyeongsang National University, Jinju 660-701, Korea.
| | - Xingming Zhang
- School of Naval Architecture and Ocean Engineering, Harbin Institute of Technology at Weihai, Weihai 264209, China.
| | - Ali Kafash Hoshiar
- Faculty of Industrial and Mechanical Engineering, Islamic Azad University, Qazvin Branch, Qazvin 34199-15195, Iran.
| | - Jungwon Yoon
- School of Mechanical and Aerospace Engineering & ReCAPT, Gyeongsang National University, Jinju 660-701, Korea.
- School of Integrated Technology, Gwangju Institute of Science and Technology, 123 Cheomdan-gwagiro, Buk-gu, Gwangju 61005, Korea.
| |
Collapse
|
31
|
Rizzo S, Petrella F, Zucca I, Rinaldi E, Barbaglia A, Padelli F, Baggi F, Spaggiari L, Bellomi M, Bruzzone MG. In vitro labelling and detection of mesenchymal stromal cells: a comparison between magnetic resonance imaging of iron-labelled cells and magnetic resonance spectroscopy of fluorine-labelled cells. Eur Radiol Exp 2017; 1:6. [PMID: 29708157 PMCID: PMC5909334 DOI: 10.1186/s41747-017-0010-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/31/2017] [Indexed: 12/31/2022] Open
Abstract
Background Among the various stem cell populations used for cell therapy, adult mesenchymal stromal cells (MSCs) have emerged as a major new cell technology. These cells must be tracked after transplantation to monitor their migration within the body and quantify their accumulation at the target site. This study assessed whether rat bone marrow MSCs can be labelled with superparamagnetic iron oxide (SPIO) nanoparticles and perfluorocarbon (PFC) nanoemulsion formulations without altering cell viability and compared magnetic resonance imaging (MRI) and magnetic resonance spectroscopy (MRS) results from iron-labelled and fluorine-labelled MSCs, respectively. Methods Of MSCs, 2 × 106 were labelled with Molday ION Rhodamine-B (MIRB) and 2 × 106 were labelled with Cell Sense. Cell viability was evaluated by trypan blue exclusion method. Labelled MSCs were divided into four samples containing increasing cell numbers (0.125 × 106, 0.25 × 106, 0.5 × 106, 1 × 106) and scanned on a 7T MRI: for MIRB-labelled cells, phantoms and cells negative control, T1, T2 and T2* maps were acquired; for Cell Sense labelled cells, phantoms and unlabelled cells, a 19F non-localised single-pulse MRS sequence was acquired. Results In total, 86.8% and 83.6% of MIRB-labelled cells and Cell Sense-labelled cells were viable, respectively. MIRB-labelled cells were visible in all samples with different cell numbers; pellets containing 0.5 × 106 and 1 × 106 of Cell Sense-labelled cells showed a detectable 19F signal. Conclusions Our data support the use of both types of contrast material (SPIO and PFC) for MSCs labelling, although further efforts should be dedicated to improve the efficiency of PFC labelling.
Collapse
Affiliation(s)
- Stefania Rizzo
- 1Department of Radiology, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy
| | - Francesco Petrella
- 2Department of Thoracic Surgery, European Institute of Oncology, Milan, Italy.,5Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, via Festa del Perdono 7, 20142 Milan, Italy
| | - Ileana Zucca
- Scientific Department, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Elena Rinaldi
- Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Andrea Barbaglia
- Scientific Department, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Francesco Padelli
- Scientific Department, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Fulvio Baggi
- Neuroimmunology and Neuromuscular Diseases Unit, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| | - Lorenzo Spaggiari
- 2Department of Thoracic Surgery, European Institute of Oncology, Milan, Italy.,5Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, via Festa del Perdono 7, 20142 Milan, Italy
| | - Massimo Bellomi
- 1Department of Radiology, European Institute of Oncology, via Ripamonti 435, 20141 Milan, Italy.,5Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, via Festa del Perdono 7, 20142 Milan, Italy
| | - Maria Grazia Bruzzone
- Department of Neuroradiology, Neurological Institute IRCCS "Carlo Besta", Milan, Italy
| |
Collapse
|
32
|
Quantifying tumor associated macrophages in breast cancer: a comparison of iron and fluorine-based MRI cell tracking. Sci Rep 2017; 7:42109. [PMID: 28176853 PMCID: PMC5296729 DOI: 10.1038/srep42109] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/06/2017] [Indexed: 12/26/2022] Open
Abstract
Tumor associated macrophages (TAMs) are associated with tumor growth and metastasis. MRI can detect TAMs labeled with iron oxide (USPIO) or perfluorocarbon (PFC) agents. This study compared these two cell tracking approaches for imaging TAMs in vivo. 4T1 tumors were imaged with MRI at 4 days or 3 weeks post cell implantation after intravenous (i.v.) administration of either USPIO or PFC. Signal loss was detected within tumors at both time points post USPIO. Images acquired at 4 days demonstrated signal loss encompassing the entire tumor and around the periphery at 3 weeks. Number of black voxels suggested higher numbers of TAMs in the tumor at the later time point. After PFC administration, Fluorine-19 (19F) signal was detected in a similar spatial distribution as signal loss post USPIO. 19F signal quantification revealed that the number of 19F spins was not significantly different at the two time points, suggesting a similar number of TAMs were present in tumors but accumulated in different regions. 19F signal was higher centrally in tumors at 4 days and heterogenous around the periphery at 3 weeks. This study revealed that 19F-based cell tracking methods better represent TAM density and provides additional information not achievable with iron-based methods.
Collapse
|
33
|
Jiráková K, Šeneklová M, Jirák D, Turnovcová K, Vosmanská M, Babič M, Horák D, Veverka P, Jendelová P. The effect of magnetic nanoparticles on neuronal differentiation of induced pluripotent stem cell-derived neural precursors. Int J Nanomedicine 2016; 11:6267-6281. [PMID: 27920532 PMCID: PMC5125991 DOI: 10.2147/ijn.s116171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Introduction Magnetic resonance (MR) imaging is suitable for noninvasive long-term tracking. We labeled human induced pluripotent stem cell-derived neural precursors (iPSC-NPs) with two types of iron-based nanoparticles, silica-coated cobalt zinc ferrite nanoparticles (CZF) and poly-l-lysine-coated iron oxide superparamagnetic nanoparticles (PLL-coated γ-Fe2O3) and studied their effect on proliferation and neuronal differentiation. Materials and methods We investigated the effect of these two contrast agents on neural precursor cell proliferation and differentiation capability. We further defined the intracellular localization and labeling efficiency and analyzed labeled cells by MR. Results Cell proliferation was not affected by PLL-coated γ-Fe2O3 but was slowed down in cells labeled with CZF. Labeling efficiency, iron content and relaxation rates measured by MR were lower in cells labeled with CZF when compared to PLL-coated γ-Fe2O3. Cytoplasmic localization of both types of nanoparticles was confirmed by transmission electron microscopy. Flow cytometry and immunocytochemical analysis of specific markers expressed during neuronal differentiation did not show any significant differences between unlabeled cells or cells labeled with both magnetic nanoparticles. Conclusion Our results show that cells labeled with PLL-coated γ-Fe2O3 are suitable for MR detection, did not affect the differentiation potential of iPSC-NPs and are suitable for in vivo cell therapies in experimental models of central nervous system disorders.
Collapse
Affiliation(s)
- Klára Jiráková
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic
| | - Monika Šeneklová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Department of Neuroscience, Second Faculty of Medicine, Charles University
| | - Daniel Jirák
- MR-Unit, Radiodiagnostic and Interventional Radiology Department, Institute for Clinical and Experimental Medicine; Department of Biophysics, Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University
| | - Karolína Turnovcová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic
| | - Magda Vosmanská
- Department of Analytical Chemistry, University of Chemistry and Technology
| | - Michal Babič
- Department of Polymer Particles, Institute of Macromolecular Chemistry
| | - Daniel Horák
- Department of Polymer Particles, Institute of Macromolecular Chemistry
| | - Pavel Veverka
- Department of Magnetics and Superconductors, Institute of Physics, ASCR, Prague, Czech Republic
| | - Pavla Jendelová
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Department of Neuroscience, Second Faculty of Medicine, Charles University
| |
Collapse
|
34
|
Murrell DH, Zarghami N, Jensen MD, Dickson F, Chambers AF, Wong E, Foster PJ. MRI surveillance of cancer cell fate in a brain metastasis model after early radiotherapy. Magn Reson Med 2016; 78:1506-1512. [PMID: 27851873 DOI: 10.1002/mrm.26541] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/21/2016] [Accepted: 10/14/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Incidence of brain metastasis attributed to breast cancer is increasing and prognosis is poor. It is thought that disseminated dormant cancer cells persist in metastatic organs and may evade treatments, thereby facilitating a mechanism for recurrence. Radiotherapy is used to treat brain metastases clinically, but assessment has been limited to macroscopic tumor volumes detectable by clinical imaging. Here, we use cellular MRI to understand the concurrent responses of metastases and nonproliferative or slowly cycling cancer cells to radiotherapy. METHODS MRI cell tracking was used to investigate the impact of early cranial irradiation on the fate of individual iron-labeled cancer cells and outgrowth of breast cancer brain metastases in the human MDA-MB-231-BR-HER2 cell model. RESULTS Early whole-brain radiotherapy significantly reduced the outgrowth of metastases from individual disseminated cancer cells in treated animals compared to controls. However, the numbers of nonproliferative iron-retaining cancer cells in the brain were not significantly different. CONCLUSIONS Radiotherapy, when given early in cancer progression, is effective in preventing the outgrowth of solitary cancer cells to brain metastases. Future studies of the nonproliferative cancer cells' clonogenic potentials are warranted, given that their persistent presence suggests that they may have evaded treatment. Magn Reson Med 78:1506-1512, 2017. © 2016 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Donna H Murrell
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Niloufar Zarghami
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Michael D Jensen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Fiona Dickson
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Ann F Chambers
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Eugene Wong
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute, London, Ontario, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
35
|
Momeni A, Neelamegham S, Parashurama N. Current challenges for the targeted delivery and molecular imaging of stem cells in animal models. Bioengineered 2016; 8:316-324. [PMID: 27813700 PMCID: PMC5553333 DOI: 10.1080/21655979.2016.1233090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In contrast to conventional, molecular medicine that focuses on targeting specific pathways, stem cell therapy aims to perturb many related mechanisms in order to derive therapeutic benefit. This emerging modality is inherently complex due to the variety of cell types that can be used, delivery approaches that need to be optimized in order to target the cellular therapeutic to specific sites in vivo, and non-invasive imaging methods that are needed to monitor cell fate. This review highlights advancements in the field, with focus on recent publications that use preclinical animal models for cardiovascular stem cell therapy. It highlights studies where cell adhesion engineering (CAE) has been used to functionalize stem cells to home them to sites of therapy, much like peripheral blood neutrophils. It also describes the current state of molecular imaging approaches that aim to non-invasively track the spatio-temporal pattern of stem cell delivery in living subjects.
Collapse
Affiliation(s)
- Arezoo Momeni
- a Department of Chemical and Biological Engineering , University at Buffalo (State University of New York) , Furnas Hall, Buffalo , NY , USA.,b Clinical and Translation Research Center (CTRC) , University at Buffalo (State University of New York) , NY , USA
| | - Sriram Neelamegham
- a Department of Chemical and Biological Engineering , University at Buffalo (State University of New York) , Furnas Hall, Buffalo , NY , USA.,b Clinical and Translation Research Center (CTRC) , University at Buffalo (State University of New York) , NY , USA
| | - Natesh Parashurama
- a Department of Chemical and Biological Engineering , University at Buffalo (State University of New York) , Furnas Hall, Buffalo , NY , USA.,b Clinical and Translation Research Center (CTRC) , University at Buffalo (State University of New York) , NY , USA
| |
Collapse
|
36
|
Ilicak E, Senel LK, Biyik E, Çukur T. Profile-encoding reconstruction for multiple-acquisition balanced steady-state free precession imaging. Magn Reson Med 2016; 78:1316-1329. [DOI: 10.1002/mrm.26507] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 09/02/2016] [Accepted: 09/21/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Efe Ilicak
- Department of Electrical and Electronics Engineering; Bilkent University; Ankara Turkey
- National Magnetic Resonance Research Center (UMRAM); Bilkent University; Ankara Turkey
| | - Lutfi Kerem Senel
- Department of Electrical and Electronics Engineering; Bilkent University; Ankara Turkey
| | - Erdem Biyik
- Department of Electrical and Electronics Engineering; Bilkent University; Ankara Turkey
| | - Tolga Çukur
- Department of Electrical and Electronics Engineering; Bilkent University; Ankara Turkey
- National Magnetic Resonance Research Center (UMRAM); Bilkent University; Ankara Turkey
- Neuroscience Program, Graduate School of Engineering and Science; Bilkent University; Ankara Turkey
| |
Collapse
|
37
|
A multimodality imaging model to track viable breast cancer cells from single arrest to metastasis in the mouse brain. Sci Rep 2016; 6:35889. [PMID: 27767185 PMCID: PMC5073295 DOI: 10.1038/srep35889] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/06/2016] [Indexed: 01/08/2023] Open
Abstract
Cellular MRI involves sensitive visualization of iron-labeled cells in vivo but cannot differentiate between dead and viable cells. Bioluminescence imaging (BLI) measures cellular viability, and thus we explored combining these tools to provide a more holistic view of metastatic cancer cell fate in mice. Human breast carcinoma cells stably expressing Firefly luciferase were loaded with iron particles, injected into the left ventricle, and BLI and MRI were performed on days 0, 8, 21 and 28. The number of brain MR signal voids (i.e., iron-loaded cells) on day 0 significantly correlated with BLI signal. Both BLI and MRI signals decreased from day 0 to day 8, indicating a loss of viable cells rather than a loss of iron label. Total brain MR tumour volume on day 28 also correlated with BLI signal. Overall, BLI complemented our sensitive cellular MRI technologies well, allowing us for the first time to screen animals for successful injections, and, in addition to MR measures of cell arrest and tumor burden, provided longitudinal measures of cancer cell viability in individual animals. We predict this novel multimodality molecular imaging framework will be useful for evaluating the efficacy of emerging anti-cancer drugs at different stages of the metastatic cascade.
Collapse
|
38
|
Rosenberg JT, Yuan X, Grant S, Ma T. Tracking mesenchymal stem cells using magnetic resonance imaging. Brain Circ 2016; 2:108-113. [PMID: 30276283 PMCID: PMC6126273 DOI: 10.4103/2394-8108.192521] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/05/2016] [Accepted: 08/30/2016] [Indexed: 01/12/2023] Open
Abstract
Recent translational studies in the fields of tissue regeneration and cell therapy have characterized mesenchymal stem cells (MSCs) as a potentially effective and accessible measure for treating ischemic cerebral and neurodegenerative disorders such as stroke, Parkinson's disease, and amyotrophic lateral sclerosis. Developing more efficient cell tracking techniques bear the potential to optimize MSC transplantation therapies by providing a more accurate picture of the fate and area of effect of implanted cells. Currently, determining the location of transplanted MSCs involves a histological approach, but magnetic resonance imaging (MRI) presents a noninvasive paradigm that permits repeat evaluations. To visualize MSCs using MRI, the implanted cells must be treated with an intracellular contrast agent. These are commonly paramagnetic compounds, many of which are based on superparamagnetic iron oxide (SPIO) nanoparticles. Recent research has set out characterize the effects of SPIO-uptake on the cellular activity of in vitro human MSCs and the resultant influence that respective SPIO concentration has on MRI sensitivity. As these studies reveal, SPIO-uptake has no effect on the cellular processes of proliferation and differentiation while producing high contrast MRI signals. Moreover, transplantation of SPIO-labeled MSCs in animal models encouragingly showed no loss in MRI contrast, suggesting that SPIO labeling may be an appealing regime for lasting MRI detection. This study is a review article. Referred literature in this study has been listed in the reference part. The datasets supporting the conclusions of this article are available online by searching the PubMed. Some original points in this article come from the laboratory practice in our research centers and the authors’ experiences.
Collapse
Affiliation(s)
- Jens T Rosenberg
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Xuegang Yuan
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Samuel Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
39
|
Makela AV, Murrell DH, Parkins KM, Kara J, Gaudet JM, Foster PJ. Cellular Imaging With MRI. Top Magn Reson Imaging 2016; 25:177-186. [PMID: 27748707 DOI: 10.1097/rmr.0000000000000101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cellular magnetic resonance imaging (MRI) is an evolving field of imaging with strong translational and research potential. The ability to detect, track, and quantify cells in vivo and over time allows for studying cellular events related to disease processes and may be used as a biomarker for decisions about treatments and for monitoring responses to treatments. In this review, we discuss methods for labeling cells, various applications for cellular MRI, the existing limitations, strategies to address these shortcomings, and clinical cellular MRI.
Collapse
Affiliation(s)
- Ashley V Makela
- *Imaging Research Laboratories, Robarts Research Institute †Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
40
|
Gaudet JM, Hamilton AM, Chen Y, Fox MS, Foster PJ. Application of dual19F and iron cellular MRI agents to track the infiltration of immune cells to the site of a rejected stem cell transplant. Magn Reson Med 2016; 78:713-720. [DOI: 10.1002/mrm.26400] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/07/2016] [Accepted: 08/09/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Jeffrey M. Gaudet
- Imaging Research Laboratories, Robarts Research Institute; London Ontario Canada
- Department of Medical Biophysics; University of Western Ontario; London Ontario Canada
| | - Amanda M. Hamilton
- Imaging Research Laboratories, Robarts Research Institute; London Ontario Canada
| | - Yuanxin Chen
- Imaging Research Laboratories, Robarts Research Institute; London Ontario Canada
| | - Matthew S. Fox
- Imaging Research Laboratories, Robarts Research Institute; London Ontario Canada
- Department of Medical Biophysics; University of Western Ontario; London Ontario Canada
| | - Paula J. Foster
- Imaging Research Laboratories, Robarts Research Institute; London Ontario Canada
- Department of Medical Biophysics; University of Western Ontario; London Ontario Canada
| |
Collapse
|
41
|
Kokeny P, Cheng YCN, Liu S, Xie H, Jiang Q. Quantifications of in vivo labeled stem cells based on measurements of magnetic moments. Magn Reson Imaging 2016; 35:141-147. [PMID: 27594530 DOI: 10.1016/j.mri.2016.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/05/2016] [Accepted: 08/20/2016] [Indexed: 11/30/2022]
Abstract
Cells labeled by super paramagnetic iron-oxide (SPIO) nanoparticles are more easily seen in gradient echo MR images, but it has not been shown that the amount of nanoparticles or the number of cells can be directly quantified from MR images. This work utilizes a previously developed and improved Complex Image Summation around a Spherical or Cylindrical Object (CISSCO) method to quantify the magnetic moments of several clusters of SPIO nanoparticle labeled cells from archived rat brain images. With the knowledge of mass magnetization of the cell labeling agent and cell iron uptake, the number of cells in each nanoparticle cluster can be determined. Using a high pass filter with a reasonable size has little effect on each measured magnetic moment from the CISSCO method. These procedures and quantitative results may help improve the efficacy of cell-based treatments in vivo.
Collapse
Affiliation(s)
- Paul Kokeny
- Department of Radiology, Wayne State University, Detroit, MI 48201.
| | - Yu-Chung N Cheng
- Department of Radiology, Wayne State University, Detroit, MI 48201.
| | - Saifeng Liu
- The MRI Institute for Biomedical Research, 761 Lucerne Avenue, Waterloo, ON, Canada
| | - He Xie
- Department of Physics, Wayne State University, Detroit, MI 48201
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202
| |
Collapse
|
42
|
Kilian T, Fidler F, Kasten A, Nietzer S, Landgraf V, Weiß K, Walles H, Westphal F, Hackenberg S, Grüttner C, Steinke M. Stem cell labeling with iron oxide nanoparticles: impact of 3D culture on cell labeling maintenance. Nanomedicine (Lond) 2016; 11:1957-70. [PMID: 27456272 DOI: 10.2217/nnm-2016-0042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AIM We aimed to analyze the suitability of nanoparticles (M4E) for safe human mesenchymal stem cell (hMSC) labeling and determined cell labeling maintenance in 2D and 3D culture. MATERIALS & METHODS We investigated cell-particle interaction and the particles' impact on cell viability, growth and proliferation. We analyzed cell labeling maintenance in 2D and 3D culture invasively and noninvasively. RESULTS M4E do not affect cell viability, growth and proliferation and do not cause chromosomal aberrations. Cell labeling maintenance is up to five-times higher in 3D conditions compared with 2D culture. CONCLUSION M4E allow safe hMSC labeling and noninvasive identification. Our hMSC-loaded, 3D tissue-engineered construct could serve as a graft for regenerative therapies, in which M4E-labeled hMSCs can migrate to their target.
Collapse
Affiliation(s)
- Teresa Kilian
- Fraunhofer Institute for Interfacial Engineering & Biotechnology IGB, Translational Center "Regenerative Therapies for Oncology & Musculoskeletal Diseases" - Würzburg branch, Röntgenring 11, 97070 Würzburg, Germany
| | - Florian Fidler
- Research Center Magnetic-Resonance-Bavaria, Am Hubland, 97074 Würzburg, Germany
| | - Annika Kasten
- Department of Oral & Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Schillingallee 35, 18057 Rostock, Germany
| | - Sarah Nietzer
- Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Veronika Landgraf
- Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Katrin Weiß
- Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Heike Walles
- Fraunhofer Institute for Interfacial Engineering & Biotechnology IGB, Translational Center "Regenerative Therapies for Oncology & Musculoskeletal Diseases" - Würzburg branch, Röntgenring 11, 97070 Würzburg, Germany.,Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Fritz Westphal
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | - Stephan Hackenberg
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic & Reconstructive Head & Neck Surgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Cordula Grüttner
- Micromod Partikeltechnologie GmbH, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany
| | - Maria Steinke
- Fraunhofer Institute for Interfacial Engineering & Biotechnology IGB, Translational Center "Regenerative Therapies for Oncology & Musculoskeletal Diseases" - Würzburg branch, Röntgenring 11, 97070 Würzburg, Germany.,Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| |
Collapse
|
43
|
Voronina N, Lemcke H, Wiekhorst F, Kühn JP, Rimmbach C, Steinhoff G, David R. Non-viral magnetic engineering of endothelial cells with microRNA and plasmid-DNA-An optimized targeting approach. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:2353-2364. [PMID: 27389150 DOI: 10.1016/j.nano.2016.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/27/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022]
Abstract
Genetic modulation of angiogenesis is a powerful tool for the treatment of multiple disorders. Here, we describe a strategy to produce modified endothelial cells, which can be efficiently magnetically guided. First, we defined optimal transfection conditions with both plasmid and microRNA, using a polyethyleneimine/magnetic nanoparticle-based vector (PEI/MNP), previously designed in our group. Further, two approaches were assessed in vitro: direct vector guidance and magnetic targeting of transfected cells. Due to its higher efficiency, including simulated dynamic conditions, production of miR/PEI/MNP-modified magnetically responsive cells was selected for further detailed investigation. In particular, we have studied internalization of transfection complexes, functional capacities and intercellular communication of engineered cells and delivery of therapeutic miR. Moreover, we demonstrated that 104 miRNA/PEI/MNP-modified magnetically responsive cells loaded with 0.37pg iron/cell are detectable with MRI. Taken together, our in vitro findings show that PEI/MNP is highly promising as a multifunctional tool for magnetically guided angiogenesis regulation.
Collapse
Affiliation(s)
- Natalia Voronina
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | - Heiko Lemcke
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | | | - Jens-Peter Kühn
- Department of Radiology and Neuroradiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany;.
| | - Christian Rimmbach
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, University of Rostock, Rostock, Germany.
| |
Collapse
|
44
|
Parashurama N, Ahn BC, Ziv K, Ito K, Paulmurugan R, Willmann JK, Chung J, Ikeno F, Swanson JC, Merk DR, Lyons JK, Yerushalmi D, Teramoto T, Kosuge H, Dao CN, Ray P, Patel M, Chang YF, Mahmoudi M, Cohen JE, Goldstone AB, Habte F, Bhaumik S, Yaghoubi S, Robbins RC, Dash R, Yang PC, Brinton TJ, Yock PG, McConnell MV, Gambhir SS. Multimodality Molecular Imaging of Cardiac Cell Transplantation: Part II. In Vivo Imaging of Bone Marrow Stromal Cells in Swine with PET/CT and MR Imaging. Radiology 2016; 280:826-36. [PMID: 27332865 DOI: 10.1148/radiol.2016151150] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Purpose To quantitatively determine the limit of detection of marrow stromal cells (MSC) after cardiac cell therapy (CCT) in swine by using clinical positron emission tomography (PET) reporter gene imaging and magnetic resonance (MR) imaging with cell prelabeling. Materials and Methods Animal studies were approved by the institutional administrative panel on laboratory animal care. Seven swine received 23 intracardiac cell injections that contained control MSC and cell mixtures of MSC expressing a multimodality triple fusion (TF) reporter gene (MSC-TF) and bearing superparamagnetic iron oxide nanoparticles (NP) (MSC-TF-NP) or NP alone. Clinical MR imaging and PET reporter gene molecular imaging were performed after intravenous injection of the radiotracer fluorine 18-radiolabeled 9-[4-fluoro-3-(hydroxyl methyl) butyl] guanine ((18)F-FHBG). Linear regression analysis of both MR imaging and PET data and nonlinear regression analysis of PET data were performed, accounting for multiple injections per animal. Results MR imaging showed a positive correlation between MSC-TF-NP cell number and dephasing (dark) signal (R(2) = 0.72, P = .0001) and a lower detection limit of at least approximately 1.5 × 10(7) cells. PET reporter gene imaging demonstrated a significant positive correlation between MSC-TF and target-to-background ratio with the linear model (R(2) = 0.88, P = .0001, root mean square error = 0.523) and the nonlinear model (R(2) = 0.99, P = .0001, root mean square error = 0.273) and a lower detection limit of 2.5 × 10(8) cells. Conclusion The authors quantitatively determined the limit of detection of MSC after CCT in swine by using clinical PET reporter gene imaging and clinical MR imaging with cell prelabeling. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Natesh Parashurama
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Byeong-Cheol Ahn
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Keren Ziv
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Ken Ito
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Ramasamy Paulmurugan
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jürgen K Willmann
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jaehoon Chung
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Fumiaki Ikeno
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Julia C Swanson
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Denis R Merk
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jennifer K Lyons
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - David Yerushalmi
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Tomohiko Teramoto
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Hisanori Kosuge
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Catherine N Dao
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Pritha Ray
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Manishkumar Patel
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Ya-Fang Chang
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Morteza Mahmoudi
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Jeff Eric Cohen
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Andrew Brooks Goldstone
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Frezghi Habte
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Srabani Bhaumik
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Shahriar Yaghoubi
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Robert C Robbins
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Rajesh Dash
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Phillip C Yang
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Todd J Brinton
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Paul G Yock
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Michael V McConnell
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| | - Sanjiv S Gambhir
- From the Department of Radiology, James Clark Center, Molecular Imaging Program at Stanford, 318 Campus Drive West, Room E153, Stanford University, Stanford, CA 94305 (N.P., K.Z., K.I., R.P., J.K.W., D.Y., M.P., Y.F.C., F.H., S.Y., S.S.G.); Department of Cardiovascular Medicine (J.C., F.I., J.K.L., T.T., H.K., C.N.D., M.M., R.D., P.C.Y., T.J.B., P.G.Y., M.V.M.), Department of Cardiothoracic Surgery (J.C.S., D.R.M., J.E.C., A.B.G., R.C.R.), Department of Bioengineering (D.Y., P.G.Y., S.S.G.), Canary Center for Early Detection of Cancer (R.P., S.S.G.), and Department of Materials Science and Engineering (S.S.G.), Stanford University, Stanford, Calif; GE Global Research Center, General Electric, Niskayuna, NY (S.B.); Department of Nuclear Medicine, Kyungpook National University, Daegu, South Korea (B.C.A.); and Advanced Center for Treatment, Research, and Education ACTREC, Tata Memorial Centre, Navi Mumbai, India (P.R.)
| |
Collapse
|
45
|
Magnetic resonance imaging of pathogenic protozoan parasite Entamoeba histolytica labeled with superparamagnetic iron oxide nanoparticles. Invest Radiol 2016; 50:709-18. [PMID: 26135016 DOI: 10.1097/rli.0000000000000175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES The aim of this study was to establish a noninvasive tracking of the pathogenic parasite Entamoeba histolytica (Eh) after superparamagnetic iron oxide (SPIO) labeling by magnetic resonance imaging (MRI) on a single-cell level in vitro and in vivo in a mouse model for amebic liver abscess (ALA). MATERIALS AND METHODS Local institutional review committee on animal care approved all animal experiments. Entamoeba histolytica trophozoites were labeled with SPIO nanoparticles (SPIO-Eh). The uptake of SPIO by Eh was optimized using flow cytometry and visualized by bright field, fluorescence, and transmission electron microscopy. The viability of SPIO-Eh was assessed in vitro by determination of growth and ingestion rate of red blood cells. Migration of SPIO-Eh was proven by in vitro MRI in a preclinical 7 T MRI system using continually repeated MRI scans. In vivo distribution of SPIO-Eh within the mouse liver was assessed qualitatively and quantitatively by serial respiration-triggered T2*-weighted MRI, T2-weighted MRI, and R2* MR relaxometry up to 5 days after injection and correlated with immunohistology of the liver sections after removal. RESULTS Entamoeba histolytica can be efficiently labeled with SPIO without influence on parasite growth rate or phagocytic capacity. In vitro dynamic MRI allowed real-time migration monitoring and determination of velocity of single SPIO-Eh. In vivo SPIO-Eh showed signal decrease in T2*-weighted and increase of R2* in ALA formations. Motility of SPIO-Eh was necessary to induce ALA formations. CONCLUSIONS The present study demonstrates the feasibility of an efficient magnetic labeling and a noninvasive in vitro and in vivo MR tracking of the pathogenic protozoan Eh in a mouse model for ALA, thus representing in future a noninvasive imaging tool to study parasite, as well as on host-specific pathomechanisms.
Collapse
|
46
|
Ouyang Y, Kim TJ, Pratx G. Evaluation of a BGO-Based PET System for Single-Cell Tracking Performance by Simulation and Phantom Studies. Mol Imaging 2016; 15:15/0/1536012116646489. [PMID: 27175009 PMCID: PMC5293205 DOI: 10.1177/1536012116646489] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/03/2016] [Indexed: 12/27/2022] Open
Abstract
A recent method based on positron emission was reported for tracking moving point sources using the Inveon PET system. However, the effect of scanner background noise was not further explored. Here, we evaluate tracking with the Genisys4, a bismuth germanate-based PET system, which has no significant intrinsic background and may be better suited to tracking lower and/or faster activity sources. Position-dependent sensitivity of the Genisys4 was simulated in Geant4 Application for Tomographic Emission (GATE) using a static 18F point source. Trajectories of helically moving point sources with varying activity and rotation speed were reconstructed from list-mode data as described previously. Simulations showed that the Inveon’s ability to track sources within 2 mm of localization error is limited to objects with a velocity-to-activity ratio < 0.13 mm/decay, compared to < 0.29 mm/decay for the Genisys4. Tracking with the Genisys4 was then validated using a physical phantom of helically moving [18F] fluorodeoxyglucose-in-oil droplets (< 0.24 mm diameter, 139-296 Bq), yielding < 1 mm localization error under the tested conditions, with good agreement between simulated sensitivity and measured activity (Pearson correlation R = .64, P << .05 in a representative example). We have investigated the tracking performance with the Genisys4, and results suggest the feasibility of tracking low activity, point source-like objects with this system.
Collapse
Affiliation(s)
- Yu Ouyang
- Radiation Biophysics Laboratory, Department of Radiation Oncology, Stanford University, Palo Alto, CA, USA
| | - Tae Jin Kim
- Radiation Biophysics Laboratory, Department of Radiation Oncology, Stanford University, Palo Alto, CA, USA
| | - Guillem Pratx
- Radiation Biophysics Laboratory, Department of Radiation Oncology, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
47
|
Novotna B, Turnovcova K, Veverka P, Rössner P, Bagryantseva Y, Herynek V, Zvatora P, Vosmanska M, Klementova M, Sykova E, Jendelova P. The impact of silica encapsulated cobalt zinc ferrite nanoparticles on DNA, lipids and proteins of rat bone marrow mesenchymal stem cells. Nanotoxicology 2015; 10:662-70. [PMID: 26581309 DOI: 10.3109/17435390.2015.1107144] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Nanomaterials are currently the subject of intense research due to their wide variety of potential applications in the biomedical, optical and electronic fields. We prepared and tested cobalt zinc ferrite nanoparticles (Co0.5Zn0.5Fe2O4+γ [CZF-NPs]) encapsulated by amorphous silica in order to find a safe contrast agent and magnetic label for tracking transplanted cells within an organism using magnetic resonance imaging (MRI). Rat mesenchymal stem cells (rMSCs) were labeled for 48 h with a low, medium or high dose of CZF-NPs (0.05; 0.11 or 0.55 mM); silica NPs (Si-NPs; 0.11 mM) served as a positive control. The internalization of NPs into cells was verified by transmission electron microscopy. Biological effects were analyzed at the end of exposure and after an additional 72 h of cell growth without NPs. Compared to untreated cells, Annexin V/Propidium Iodide labeling revealed no significant cytotoxicity for any group of treated cells and only a high dose of CZF-NPs slowed down cell proliferation and induced DNA damage, manifested as a significant increase of DNA-strand breaks and oxidized DNA bases. This was accompanied by high concentrations of 15-F2t-isoprostane and carbonyl groups, demonstrating oxidative injury to lipids and proteins, respectively. No harmful effects were detected in cells exposed to the low dose of CZF-NPs. Nevertheless, the labeled cells still exhibited an adequate relaxation rate for MRI in repeated experiments and ICP-MS confirmed sufficient magnetic label concentrations inside the cells. The results suggest that the silica-coated CZF-NPs, when applied at a non-toxic dose, represent a promising contrast agent for cell labeling.
Collapse
Affiliation(s)
- Bozena Novotna
- a Department of Genetic Ecotoxicology , Institute of Experimental Medicine AS CR , v.v.i., Prague , Czech Republic
| | - Karolina Turnovcova
- a Department of Genetic Ecotoxicology , Institute of Experimental Medicine AS CR , v.v.i., Prague , Czech Republic
| | - Pavel Veverka
- b Institute of Physics, AS CR, v.v.i. , Prague , Czech Republic
| | - Pavel Rössner
- a Department of Genetic Ecotoxicology , Institute of Experimental Medicine AS CR , v.v.i., Prague , Czech Republic
| | - Yana Bagryantseva
- a Department of Genetic Ecotoxicology , Institute of Experimental Medicine AS CR , v.v.i., Prague , Czech Republic
| | - Vit Herynek
- c Institute for Clinical and Experimental Medicine , Prague , Czech Republic
| | | | - Magda Vosmanska
- e University of Chemistry and Technology , Prague , Czech Republic , and
| | - Mariana Klementova
- f New Technologies - Research Centre, University of West Bohemia , Pilsen , Czech Republic
| | - Eva Sykova
- a Department of Genetic Ecotoxicology , Institute of Experimental Medicine AS CR , v.v.i., Prague , Czech Republic
| | - Pavla Jendelova
- a Department of Genetic Ecotoxicology , Institute of Experimental Medicine AS CR , v.v.i., Prague , Czech Republic
| |
Collapse
|
48
|
Malosio ML, Esposito A, Brigatti C, Palmisano A, Piemonti L, Nano R, Maffi P, De Cobelli F, Del Maschio A, Secchi A. MR Imaging Monitoring of Iron-Labeled Pancreatic Islets in a Small Series of Patients: Islet Fate in Successful, Unsuccessful, and Autotransplantation. Cell Transplant 2015; 24:2285-96. [DOI: 10.3727/096368914x684060] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Islet transplantation is one of the most promising and effective therapies for restoring normoglycemia in type 1 diabetes (T1D) patients, but islet engraftment is one of the main obstacles hampering long-term success. Monitoring graft loss, caused either by immunological or nonimmunological events, occurring in the first phase after transplantation and at later stages of a patient's life is a very important issue. Among the imaging approaches previously applied, magnetic resonance imaging (MRI) monitoring of islet fate following labeling with superparamagnetic iron oxide agents yielded promising results. The aim of this study was to translate into patients the method of islet labeling and MRI monitoring developed in our preclinical setting and to compare imaging results with graft clinical outcome. Three T1D patients and one nondiabetic patient undergoing autotransplantation following subtotal pancreatectomy received Endorem®-labeled islets. Patients were monitored by MRI and metabolically (HbA1c, exogenous insulin requirement, and C-peptide, TEF) at 1, 3, and 7 days following transplantation and once a month up to 10 months. Labeled transplanted islets appeared as hypointense areas scattered within the liver parenchyma, whose absolute number at 24 h after transplantation reflected the labeling efficiency. In patients #1 and #3 with good midterm graft function, MRI follow-up showed an important early loss of hypointense spots followed by a slow and progressive disappearance at later timepoints. Graft loss of function in patient #2 4 weeks after transplantation was associated with the complete disappearance of all hypointense signals. The autotransplanted patient, stably insulin free, showed no significant signal reduction during the first 3 days, followed by loss of spots similar to a patient with good midterm graft function. These results suggest that MRI monitoring of islet transplantation at early time points could represent a meaningful readout for helping in predicting transplant failure or success, but its relevance for mid/long-term islet function assessment appears evanescent.
Collapse
Affiliation(s)
- Maria Luisa Malosio
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Antonio Esposito
- Radiology Department, San Raffaele Scientific Institute, Milan, Italy
- Center of Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Cristina Brigatti
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Palmisano
- Radiology Department, San Raffaele Scientific Institute, Milan, Italy
- Center of Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Human Islet Isolation and Transplantation Program, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Rita Nano
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Human Islet Isolation and Transplantation Program, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Paola Maffi
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Transplant Medicine Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Francesco De Cobelli
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Radiology Department, San Raffaele Scientific Institute, Milan, Italy
- Center of Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Del Maschio
- Radiology Department, San Raffaele Scientific Institute, Milan, Italy
- Center of Experimental Imaging, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Secchi
- Vita-Salute San Raffaele University, Milan, Italy
- Transplant Medicine Unit, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
49
|
Granot D, Nkansah MK, Bennewitz MF, Tang KS, Markakis EA, Shapiro EM. Clinically viable magnetic poly(lactide-co-glycolide) particles for MRI-based cell tracking. Magn Reson Med 2015; 71:1238-50. [PMID: 23568825 DOI: 10.1002/mrm.24741] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To design, fabricate, characterize, and in vivo assay clinically viable magnetic particles for MRI-based cell tracking. METHODS Poly(lactide-co-glycolide) (PLGA) encapsulated magnetic nano and microparticles were fabricated. Multiple biologically relevant experiments were performed to assess cell viability, cellular performance, and stem cell differentiation. In vivo MRI experiments were performed to separately test cell transplantation and cell migration paradigms, as well as in vivo biodegradation. RESULTS Highly magnetic nano (∼100 nm) and microparticles (∼1-2 µm) were fabricated. Magnetic cell labeling in culture occurred rapidly achieving 3-50 pg Fe/cell at 3 h for different particles types, and >100 pg Fe/cell after 10 h, without the requirement of a transfection agent, and with no effect on cell viability. The capability of magnetically labeled mesenchymal or neural stem cells to differentiate down multiple lineages, or for magnetically labeled immune cells to release cytokines following stimulation, was uncompromised. An in vivo biodegradation study revealed that NPs degraded ∼80% over the course of 12 weeks. MRI detected as few as 10 magnetically labeled cells, transplanted into the brains of rats. Also, these particles enabled the in vivo monitoring of endogenous neural progenitor cell migration in rat brains over 2 weeks. CONCLUSION The robust MRI properties and benign safety profile of these particles make them promising candidates for clinical translation for MRI-based cell tracking.
Collapse
Affiliation(s)
- Dorit Granot
- Molecular and Cellular MRI Laboratory, Magnetic Resonance Research Center, Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
50
|
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
|