1
|
McRae S, Martinez F, Foster P, Ronald J, Scholl T. Single-Frequency Birdcage Coils for Deep Tissue Perfluorocarbon Magnetic Resonance Imaging in Mice. NMR IN BIOMEDICINE 2025; 38:e5296. [PMID: 39648071 PMCID: PMC11625660 DOI: 10.1002/nbm.5296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 12/10/2024]
Abstract
Fluorine-19 (19F) MRI has become an established tool for in vivo cell tracking following ex vivo or in vivo labelling of various cell types with 19F perfluorocarbons (PFCs). Here, we developed and evaluated novel mouse-specific radiofrequency (RF) hardware for improved dual 1H anatomical imaging and deep tissue 19F MR detection of PFCs. Three linearly polarized birdcage RF coils were constructed-a dual-frequency 1H/19F coil, and a pair of single-frequency 1H and 19F coils, designed to be used sequentially. RF coil quality factors (Q values), signal homogeneity and sensitivity were benchmarked against a commercially constructed dual-frequency 1H/19F surface coil. RF homogeneity was assessed using a phantom designed to mimic PFC localization at depth in a mouse. The single-frequency birdcage coils (1H and 19F) displayed more uniform coverage and enhanced signal-to-noise ratios (SNRs) compared to both the birdcage and surface dual-frequency coils for 19F detection. Bilateral injection of a perfluoropolyether nanoemulsion into the footpads of female athymic nude mice, resulting in drainage to various lymph nodes and subsequent accumulation in lymph node macrophages, provided a platform to assess differences in SNRs and contrast-to-noise ratios (CNR) between both coil configurations as a function of depth and location. The single-frequency 1H coil provided significantly increased CNR in anatomical images (p < 0.001) with increased anatomical coverage compared to the dual-frequency surface coil. The single-frequency 19F birdcage coil offered increased PFC detectability with significantly higher SNR in renal, lumbar, sciatic and popliteal lymph nodes (p < 0.01) compared to the dual-frequency surface coil. Interestingly, the percentage difference between SNR measurements in lymph nodes between the single-frequency 19F coil and the 1H/19F surface coil had a linear relationship with increasing distance from the surface coil (R2 = 0.6352; p < 0.0001), indicating a potential disagreement for imaging experiments that rely on 19F spin quantification at increasing depth within the mouse using surface RF coils.
Collapse
Affiliation(s)
- Sean W. McRae
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
| | | | - Paula J. Foster
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
- Imaging Laboratories, Robarts Research InstituteUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's Health CareLondonOntarioCanada
| | - John A. Ronald
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
- Imaging Laboratories, Robarts Research InstituteUniversity of Western OntarioLondonOntarioCanada
- Lawson Health Research InstituteSt. Joseph's Health CareLondonOntarioCanada
| | - Timothy J. Scholl
- Department of Medical BiophysicsUniversity of Western OntarioLondonOntarioCanada
- Imaging Laboratories, Robarts Research InstituteUniversity of Western OntarioLondonOntarioCanada
- Department of Physics and AstronomyUniversity of Western OntarioLondonOntarioCanada
- Ontario Institute for Cancer ResearchTorontoOntarioCanada
| |
Collapse
|
2
|
Chen J, Pal P, Ahrens ET. Systems Engineering Approach Towards Sensitive Cellular Fluorine-19 MRI. NMR IN BIOMEDICINE 2025; 38:e5298. [PMID: 39648456 DOI: 10.1002/nbm.5298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/21/2024] [Accepted: 11/13/2024] [Indexed: 12/10/2024]
Abstract
In vivo fluorine-19 MRI using F-based tracer media has shown utility and versatility for a wide range of biomedical uses, particularly immune and stem cell detection, as well as biosensing. As with many advanced MRI acquisition techniques, the sensitivity and limit of detection (LOD) in vivo is a key consideration for a successful study outcome. In this review, we analyze the primary factors that limit cell LOD. The achievable sensitivity is strongly dependent on the specific composition of tracer, cell type of interest, cell activity, data acquisition and reconstruction methods, and MRI hardware design. Recent innovations in molecular 19F tracer design and image acquisition-reconstruction methods have achieved significant leaps in 19F MRI sensitivity, and integration of these new materials and methods into studies can result in > 10-fold improvement in LOD. These developments will help unlock the full potential of clinical 19F MRI for biomedical applications.
Collapse
Affiliation(s)
- Jiawen Chen
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, California, USA
| | - Piya Pal
- Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, California, USA
| | - Eric T Ahrens
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
3
|
Bulte JWM, Shakeri-Zadeh A. In Vivo MRI Tracking of Tumor Vaccination and Antigen Presentation by Dendritic Cells. Mol Imaging Biol 2022; 24:198-207. [PMID: 34581954 PMCID: PMC8477715 DOI: 10.1007/s11307-021-01647-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 01/24/2023]
Abstract
Cancer vaccination using tumor antigen-primed dendritic cells (DCs) was introduced in the clinic some 25 years ago, but the overall outcome has not lived up to initial expectations. In addition to the complexity of the immune response, there are many factors that determine the efficacy of DC therapy. These include accurate administration of DCs in the target tissue site without unwanted cell dispersion/backflow, sufficient numbers of tumor antigen-primed DCs homing to lymph nodes (LNs), and proper timing of immunoadjuvant administration. To address these uncertainties, proton (1H) and fluorine (19F) magnetic resonance imaging (MRI) tracking of ex vivo pre-labeled DCs can now be used to non-invasively determine the accuracy of therapeutic DC injection, initial DC dispersion, systemic DC distribution, and DC migration to and within LNs. Magnetovaccination is an alternative approach that tracks in vivo labeled DCs that simultaneously capture tumor antigen and MR contrast agent in situ, enabling an accurate quantification of antigen presentation to T cells in LNs. The ultimate clinical premise of MRI DC tracking would be to use changes in LN MRI signal as an early imaging biomarker to predict the efficacy of tumor vaccination and anti-tumor response long before treatment outcome becomes apparent, which may aid clinicians with interim treatment management.
Collapse
Affiliation(s)
- Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA.
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA.
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Ali Shakeri-Zadeh
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, MRB 659, 733 N. Broadway, MD, 21205, Baltimore, USA
| |
Collapse
|
4
|
Mali A, Kaijzel EL, Lamb HJ, Cruz LJ. 19F-nanoparticles: Platform for in vivo delivery of fluorinated biomaterials for 19F-MRI. J Control Release 2021; 338:870-889. [PMID: 34492234 DOI: 10.1016/j.jconrel.2021.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/19/2022]
Abstract
Fluorine-19 (19F) magnetic resonance imaging (MRI) features one of the most investigated and innovative techniques for quantitative and unambiguous cell tracking, providing information for both localization and number of cells. Because of the relative insensitivity of the MRI technique, a high number of magnetically equivalent fluorine atoms are required to gain detectable signals. However, an increased amount of 19F nuclei induces low solubility in aqueous solutions, making fluorine-based probes not suitable for in vivo imaging applications. In this context, nanoparticle-based platforms play a crucial role, since nanoparticles may carry a high payload of 19F-based contrast agents into the relevant cells or tissues, increase the imaging agents biocompatibility, and provide a highly versatile platform. In this review, we present an overview of the 19F-based nanoprobes for sensitive 19F-MRI, focusing on the main nanotechnologies employed to date, such as fluorine and theranostic nanovectors, including their design and applications.
Collapse
Affiliation(s)
- Alvja Mali
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Eric L Kaijzel
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
5
|
Helfer BM, Ponomarev V, Patrick PS, Blower PJ, Feitel A, Fruhwirth GO, Jackman S, Pereira Mouriès L, Park MVDZ, Srinivas M, Stuckey DJ, Thu MS, van den Hoorn T, Herberts CA, Shingleton WD. Options for imaging cellular therapeutics in vivo: a multi-stakeholder perspective. Cytotherapy 2021; 23:757-773. [PMID: 33832818 PMCID: PMC9344904 DOI: 10.1016/j.jcyt.2021.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/01/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Cell-based therapies have been making great advances toward clinical reality. Despite the increase in trial activity, few therapies have successfully navigated late-phase clinical trials and received market authorization. One possible explanation for this is that additional tools and technologies to enable their development have only recently become available. To support the safety evaluation of cell therapies, the Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee, a multisector collaborative committee, polled the attendees of the 2017 International Society for Cell & Gene Therapy conference in London, UK, to understand the gaps and needs that cell therapy developers have encountered regarding safety evaluations in vivo. The goal of the survey was to collect information to inform stakeholders of areas of interest that can help ensure the safe use of cellular therapeutics in the clinic. This review is a response to the cellular imaging interests of those respondents. The authors offer a brief overview of available technologies and then highlight the areas of interest from the survey by describing how imaging technologies can meet those needs. The areas of interest include imaging of cells over time, sensitivity of imaging modalities, ability to quantify cells, imaging cellular survival and differentiation and safety concerns around adding imaging agents to cellular therapy protocols. The Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee believes that the ability to understand therapeutic cell fate is vital for determining and understanding cell therapy efficacy and safety and offers this review to aid in those needs. An aim of this article is to share the available imaging technologies with the cell therapy community to demonstrate how these technologies can accomplish unmet needs throughout the translational process and strengthen the understanding of cellular therapeutics.
Collapse
Affiliation(s)
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - P Stephen Patrick
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Alexandra Feitel
- Formerly, Health and Environmental Sciences Institute, US Environmental Protection Agency, Washington, DC, USA
| | - Gilbert O Fruhwirth
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Shawna Jackman
- Charles River Laboratories, Shrewsbury, Massachusetts, USA
| | | | - Margriet V D Z Park
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, the Netherlands; Cenya Imaging BV, Amsterdam, the Netherlands
| | - Daniel J Stuckey
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Mya S Thu
- Visicell Medical Inc, La Jolla, California, USA
| | | | | | | |
Collapse
|
6
|
Waiczies S, Prinz C, Starke L, Millward JM, Delgado PR, Rosenberg J, Nazaré M, Waiczies H, Pohlmann A, Niendorf T. Functional Imaging Using Fluorine ( 19F) MR Methods: Basic Concepts. Methods Mol Biol 2021; 2216:279-299. [PMID: 33476007 PMCID: PMC9703275 DOI: 10.1007/978-1-0716-0978-1_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Kidney-associated pathologies would greatly benefit from noninvasive and robust methods that can objectively quantify changes in renal function. In the past years there has been a growing incentive to develop new applications for fluorine (19F) MRI in biomedical research to study functional changes during disease states. 19F MRI represents an instrumental tool for the quantification of exogenous 19F substances in vivo. One of the major benefits of 19F MRI is that fluorine in its organic form is absent in eukaryotic cells. Therefore, the introduction of exogenous 19F signals in vivo will yield background-free images, thus providing highly selective detection with absolute specificity in vivo. Here we introduce the concept of 19F MRI, describe existing challenges, especially those pertaining to signal sensitivity, and give an overview of preclinical applications to illustrate the utility and applicability of this technique for measuring renal function in animal models.This chapter is based upon work from the COST Action PARENCHIMA, a community-driven network funded by the European Cooperation in Science and Technology (COST) program of the European Union, which aims to improve the reproducibility and standardization of renal MRI biomarkers. This introduction chapter is complemented by two separate chapters describing the experimental procedure and data analysis.
Collapse
Affiliation(s)
- Sonia Waiczies
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.
| | - Christian Prinz
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Ludger Starke
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Jason M Millward
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Paula Ramos Delgado
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Jens Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- Siemens Healthcare, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.), Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
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: 0.2] [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
|
9
|
Constantinides C. Is There Preclinical and Clinical Value for 19F MRI in Stem Cell Cardiac Regeneration? Cell Transplant 2020; 29:963689720954434. [PMID: 33000632 PMCID: PMC7784514 DOI: 10.1177/0963689720954434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/05/2020] [Accepted: 08/12/2020] [Indexed: 11/24/2022] Open
Abstract
Cardiovascular regeneration aims to renew damaged or necrotic tissue and to enhance cardiac functional performance. Despite the hope arisen from the introduction and use of stem cells (SCs) as a novel cardiac regenerative approach, to-this-date, clinical trial findings are still ambivalent despite preclinical successes. Concurrently, noninvasive magnetic resonance imaging (MRI) advances have been based on nanotechnological breakthroughs that have (a) allowed fluorinated nanoparticles and ultrasmall iron oxide single-cell labeling, (b) explored imaging detection sensitivity limits (for preclinical/low-field clinical settings), and (c) accomplished cellular tracking in vivo. Nevertheless, outcomes have been far from ideal. Herein, the recently developed preclinical and clinical 1H and 19F MRI approaches for direct cardiac SC labeling techniques intended for cellular implantation and their potential for tracking these cells in health and infarcted states are summarized. To this extent, the potential preclinical and clinical values of 19F MRI and tracking of SCs for cardiac regeneration in myocardial infarction are questioned and challenged.
Collapse
|
10
|
Zare S, Mehrabani D, Jalli R, Saeedi Moghadam M, Manafi N, Mehrabani G, Jamhiri I, Ahadian S. MRI-Tracking of Dental Pulp Stem Cells In Vitro and In Vivo Using Dextran-Coated Superparamagnetic Iron Oxide Nanoparticles. J Clin Med 2019; 8:E1418. [PMID: 31505807 PMCID: PMC6780915 DOI: 10.3390/jcm8091418] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to track dental pulp stem cells (DPSCs) labeled with dextran-coated superparamagnetic iron oxide nanoparticles (SPIONs) using magnetic resonance imaging (MRI). Dental pulp was isolated from male Sprague Dawley rats and cultured in Dulbecco's modified Eagle's medium F12 (DMEM-F12) and 10% fetal bovine serum. Effects of SPIONs on morphology, viability, apoptosis, stemness, and osteogenic and adipogenic differentiation of DPSCs were assessed. Prussian blue staining and MRI were conducted to determine in vitro efficiency of SPIONs uptake by the cells. Both non-labeled and labeled DPSCs were adherent to culture plates and showed spindle-shape morphologies, respectively. They were positive for osteogenic and adipogenic induction and expression of cluster of differentiation (CD) 73 and CD90 biomarkers, but negative for expression of CD34 and CD45 biomarkers. The SPIONs were non-toxic and did not induce apoptosis in doses less than 25 mg/mL. Internalization of the SPIONs within the DPSCs was confirmed by Prussian blue staining and MRI. Our findings revealed that the MRI-based method could successfully monitor DPSCs labeled with dextran-coated SPIONs without any significant effect on osteogenic and adipogenic differentiation, viability, and stemness of DPSCs. We provided the in vitro evidence supporting the feasibility of an MRI-based method to monitor DPSCs labeled with SPIONs without any significant reduction in viability, proliferation, and differentiation properties of labeled cells, showing that internalization of SPIONs within DPSCs were not toxic at doses less than 25 mg/mL. In general, the SPION labeling does not seem to impair cell survival or differentiation. SPIONs are biocompatible, easily available, and cost effective, opening a new avenue in stem cell labeling in regenerative medicine.
Collapse
Affiliation(s)
- Shahrokh Zare
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
- Department of Biochemistry, School of Biotechnology and Agriculture, Shiraz Branch, Islamic Azad University, Shiraz, Fars 71987-74731, Iran.
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71987-74731, Iran.
- Comparative and Experimental Medicine Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| | - Reza Jalli
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
| | - Mahdi Saeedi Moghadam
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
| | - Navid Manafi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Tehran 14348-75451, Iran.
| | - Golshid Mehrabani
- Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118, USA.
| | - Iman Jamhiri
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran.
| | - Samad Ahadian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90049, USA.
| |
Collapse
|
11
|
Fluorine-19 Cellular MRI Detection of In Vivo Dendritic Cell Migration and Subsequent Induction of Tumor Antigen-Specific Immunotherapeutic Response. Mol Imaging Biol 2019; 22:549-561. [DOI: 10.1007/s11307-019-01393-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
12
|
Gálisová A, Herynek V, Swider E, Sticová E, Pátiková A, Kosinová L, Kříž J, Hájek M, Srinivas M, Jirák D. A Trimodal Imaging Platform for Tracking Viable Transplanted Pancreatic Islets In Vivo: F-19 MR, Fluorescence, and Bioluminescence Imaging. Mol Imaging Biol 2019; 21:454-464. [PMID: 30167995 PMCID: PMC6525139 DOI: 10.1007/s11307-018-1270-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Combining specific and quantitative F-19 magnetic resonance imaging (MRI) with sensitive and convenient optical imaging provides complementary information about the distribution and viability of transplanted pancreatic islet grafts. In this study, pancreatic islets (PIs) were labeled with positively charged multimodal nanoparticles based on poly(lactic-co-glycolic acid) (PLGA-NPs) with encapsulated perfluoro-15-crown-5-ether and the near-infrared fluorescent dye indocyanine green. PROCEDURES One thousand and three thousand bioluminescent PIs were transplanted into subcutaneous artificial scaffolds, which served as an alternative transplant site. The grafts were monitored using in vivo F-19 MR, fluorescence, and bioluminescence imaging in healthy rats for 2 weeks. RESULTS Transplanted PIs were unambiguously localized in the scaffolds by F-19 MRI throughout the whole experiment. Fluorescence was detected in the first 4 days after transplantation only. Importantly, in vivo bioluminescence correlated with the F-19 MRI signal. CONCLUSIONS We developed a trimodal imaging platform for in vivo examination of transplanted PIs. Fluorescence imaging revealed instability of the fluorescent dye and its limited applicability for longitudinal in vivo studies. A correlation between the bioluminescence signal and the F-19 MRI signal indicated the fast clearance of PLGA-NPs from the transplantation site after cell death, which addresses a major issue with intracellular imaging labels. Therefore, the proposed PLGA-NP platform is reliable for reflecting the status of transplanted PIs in vivo.
Collapse
Affiliation(s)
- A Gálisová
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - V Herynek
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - E Swider
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - E Sticová
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Pathology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - A Pátiková
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - L Kosinová
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - J Kříž
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - M Hájek
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - M Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, Netherlands
| | - D Jirák
- MR Unit, Department of Radiodiagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| |
Collapse
|
13
|
Rothe M, Jahn A, Weiss K, Hwang JH, Szendroedi J, Kelm M, Schrader J, Roden M, Flögel U, Bönner F. In vivo 19F MR inflammation imaging after myocardial infarction in a large animal model at 3 T. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2018; 32:5-13. [DOI: 10.1007/s10334-018-0714-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/04/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022]
|
14
|
Abstract
The development of novel nanoparticles consisting of both diagnostic and therapeutic components has increased over the past decade. These "theranostic" nanoparticles have been tailored toward one or more types of imaging modalities and have been developed for optical imaging, magnetic resonance imaging, ultrasound, computed tomography, and nuclear imaging comprising both single-photon computed tomography and positron emission tomography. In this review, we focus on state-of-the-art theranostic nanoparticles that are capable of both delivering therapy and self-reporting/tracking disease through imaging. We discuss challenges and the opportunity to rapidly adjust treatment for individualized medicine.
Collapse
Affiliation(s)
- Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Dean Ho
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
- Weintraub Center for Reconstructive Biotechnology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
15
|
Fink C, Gaudet JM, Fox MS, Bhatt S, Viswanathan S, Smith M, Chin J, Foster PJ, Dekaban GA. 19F-perfluorocarbon-labeled human peripheral blood mononuclear cells can be detected in vivo using clinical MRI parameters in a therapeutic cell setting. Sci Rep 2018; 8:590. [PMID: 29330541 PMCID: PMC5766492 DOI: 10.1038/s41598-017-19031-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022] Open
Abstract
A 19Fluorine (19F) perfluorocarbon cell labeling agent, when employed with an appropriate cellular MRI protocol, allows for in vivo cell tracking. 19F cellular MRI can be used to non-invasively assess the location and persistence of cell-based cancer vaccines and other cell-based therapies. This study was designed to determine the feasibility of labeling and tracking peripheral blood mononuclear cells (PBMC), a heterogeneous cell population. Under GMP-compliant conditions human PBMC were labeled with a 19F-based MRI cell-labeling agent in a manner safe for autologous re-injection. Greater than 99% of PBMC labeled with the 19F cell-labeling agent without affecting functionality or affecting viability. The 19F-labeled PBMC were detected in vivo in a mouse model at the injection site and in a draining lymph node. A clinical cellular MR protocol was optimized for the detection of PBMC injected both at the surface of a porcine shank and at a depth of 1.2 cm, equivalent to depth of a human lymph node, using a dual 1H/19F dual switchable surface radio frequency coil. This study demonstrates it is feasible to label and track 19F-labeled PBMC using clinical MRI protocols. Thus, 19F cellular MRI represents a non-invasive imaging technique suitable to assess the effectiveness of cell-based cancer vaccines.
Collapse
Affiliation(s)
- Corby Fink
- Molecular Medicine Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, 1151 Richmond Street North, London, Ontario, N6A 5B7, Canada
| | - Jeffrey M Gaudet
- Imaging Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, 1151 Richmond Street North, London, Ontario, N6A 5B7, Canada
| | - Matthew S Fox
- Imaging Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, 1151 Richmond Street North, London, Ontario, N6A 5B7, Canada
| | - Shashank Bhatt
- 200 Elizabeth Street, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Sowmya Viswanathan
- IBBME, University of Toronto, University Health Network, 200 Elizabeth Street, Toronto, Ontario, M5G 2C4, Canada
| | - Michael Smith
- Molecular Medicine Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, 1151 Richmond Street North, London, Ontario, N6A 5B7, Canada
| | - Joseph Chin
- Division Of Surgery, Division of Surgical Oncology, London Health Sciences Centre, 800 Commissioners Rd E, London, Ontario, N6A 5W9, Canada
| | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, 1151 Richmond Street North, London, Ontario, N6A 5B7, Canada
| | - Gregory A Dekaban
- Molecular Medicine Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, 1151 Richmond Street North, London, Ontario, N6A 5B7, Canada.
| |
Collapse
|
16
|
Kadyk LC, DeWitt ND, Gomperts B. Proceedings: Regenerative Medicine for Lung Diseases: A CIRM Workshop Report. Stem Cells Transl Med 2017; 6:1823-1828. [PMID: 28791807 PMCID: PMC6430051 DOI: 10.1002/sctm.17-0047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/22/2017] [Indexed: 12/20/2022] Open
Abstract
The mission of the California Institute of Regenerative Medicine (CIRM) is to accelerate treatments to patients with unmet medical needs. In September 2016, CIRM sponsored a workshop held at the University of California, Los Angeles, to discuss regenerative medicine approaches for treatment of lung diseases and to identify the challenges remaining for advancing such treatments to the clinic and market approval. Workshop participants discussed current preclinical and clinical approaches to regenerative medicine in the lung, as well as the biology of lung stem cells and the role of stem cells in the etiology of various lung diseases. The outcome of this effort was the recognition that whereas transient cell delivery approaches are leading the way in the clinic, recent advances in the understanding of lung stem cell biology, in vitro and in vivo disease modeling, gene editing and replacement methods, and cell engraftment approaches raise the prospect of developing cures for some lung diseases in the foreseeable future. In addition, advances in in vitro modeling using lung organoids and "lung on a chip" technology are setting the stage for high quality small molecule drug screening to develop treatments for lung diseases with complex biology. Stem Cells Translational Medicine 2017;6:1823-1828.
Collapse
Affiliation(s)
- Lisa C. Kadyk
- California Institute for Regenerative MedicineOaklandCaliforniaUSA
| | | | - Brigitte Gomperts
- Children's Discovery and Innovation Institute, Broad Stem Cell Research Center, Jonsson Comprehensive Cancer Center, Departments of Pediatrics and Pulmonary Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
17
|
Differential of live and dead cells by magnetic resonance imaging. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1899-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
18
|
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: 0.8] [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
|
19
|
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: 2.1] [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
|
20
|
Kenny GD, Shaw KP, Sivachelvam S, White AJ, Botnar RM, T.M. de Rosales R. A bisphosphonate for 19F-magnetic resonance imaging. J Fluor Chem 2016; 184:58-64. [PMID: 27110036 PMCID: PMC4834630 DOI: 10.1016/j.jfluchem.2016.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 12/15/2022]
Abstract
19F-magnetic resonance imaging (MRI) is a promising technique that may allow us to measure the concentration of exogenous fluorinated imaging probes quantitatively in vivo. Here, we describe the synthesis and characterisation of a novel geminal bisphosphonate (19F-BP) that contains chemically-equivalent fluorine atoms that show a single and narrow 19F resonance and a bisphosphonate group that may be used for labelling inorganic materials based in calcium phosphates and metal oxides. The potential of 19F-BP to provide contrast was analysed in vitro and in vivo using 19F-MRI. In vitro studies demonstrated the potential of 19F-BP as an MRI contrast agent in the millimolar concentration range with signal-to-noise ratios (SNR) comparable to previously reported fluorinated probes. The preliminary in vivo MRI study reported here allowed us to visualise the biodistribution of 19F-BP, showing uptake in the liver and in the bladder/urinary system areas. However, bone uptake was not observed. In addition, 19F-BP showed undesirable toxicity effects in mice that prevent further studies with this compound at the required concentrations for MRI contrast. This study highlights the importance of developing 19F MRI probes with the highest signal intensity achievable.
Collapse
Affiliation(s)
- Gavin D. Kenny
- Division of Imaging Sciences & Biomedical Engineering, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK
| | - Karen P. Shaw
- Division of Imaging Sciences & Biomedical Engineering, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK
| | - Saranja Sivachelvam
- Division of Imaging Sciences & Biomedical Engineering, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK
| | - Andrew J.P. White
- Department of Chemistry, Imperial College London, Exhibition Road, South Kensington, London SW7 2AZ, UK
| | - Rene M. Botnar
- Division of Imaging Sciences & Biomedical Engineering, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK
| | - Rafael T.M. de Rosales
- Division of Imaging Sciences & Biomedical Engineering, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK
| |
Collapse
|
21
|
Bouchlaka MN, Ludwig KD, Gordon JW, Kutz MP, Bednarz BP, Fain SB, Capitini CM. (19)F-MRI for monitoring human NK cells in vivo. Oncoimmunology 2016; 5:e1143996. [PMID: 27467963 DOI: 10.1080/2162402x.2016.1143996] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 01/13/2023] Open
Abstract
The availability of clinical-grade cytokines and artificial antigen-presenting cells has accelerated interest in using natural killer (NK) cells as adoptive cellular therapy (ACT) for cancer. One of the technological shortcomings of translating therapies from animal models to clinical application is the inability to effectively and non-invasively track these cells after infusion in patients. We have optimized the nonradioactive isotope fluorine-19 ((19)F) as a means to label and track NK cells in preclinical models using magnetic resonance imaging (MRI). Human NK cells were expanded with interleukin (IL)-2 and labeled in vitro with increasing concentrations of (19)F. Doses as low as 2 mg/mL (19)F were detected by MRI. NK cell viability was only decreased at 8 mg/mL (19)F. No effects on NK cell cytotoxicity against K562 leukemia cells were observed with 2, 4 or 8 mg/mL (19)F. Higher doses of (19)F, 4 mg/mL and 8 mg/mL, led to an improved (19)F signal by MRI with 3 × 10(11) (19)F atoms per NK cell. The 4 mg/mL (19)F labeling had no effect on NK cell function via secretion of granzyme B or interferon gamma (IFNγ), compared to NK cells exposed to vehicle alone. (19)F-labeled NK cells were detectable immediately by MRI after intratumoral injection in NSG mice and up to day 8. When (19)F-labeled NK cells were injected subcutaneously, we observed a loss of signal through time at the site of injection suggesting NK cell migration to distant organs. The (19)F perfluorocarbon is a safe and effective reagent for monitoring the persistence and trafficking of NK cell infusions in vivo, and may have potential for developing novel imaging techniques to monitor ACT for cancer.
Collapse
Affiliation(s)
- Myriam N Bouchlaka
- Department of Pediatrics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, WI, USA
| | - Kai D Ludwig
- Department of Medical Physics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, WI, USA
| | - Jeremy W Gordon
- Department of Medical Physics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, WI, USA
| | - Matthew P Kutz
- Department of Pediatrics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, WI, USA
| | - Bryan P Bednarz
- Department of Medical Physics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, WI, USA
| | - Sean B Fain
- Department of Medical Physics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Biomedical Engineering, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Christian M Capitini
- Department of Pediatrics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health , Madison, WI, USA
| |
Collapse
|
22
|
Schmieder AH, Caruthers SD, Keupp J, Wickline SA, Lanza GM. Recent Advances in 19Fluorine Magnetic Resonance Imaging with Perfluorocarbon Emulsions. ENGINEERING (BEIJING, CHINA) 2015; 1:475-489. [PMID: 27110430 PMCID: PMC4841681 DOI: 10.15302/j-eng-2015103] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The research roots of 19fluorine (19F) magnetic resonance imaging (MRI) date back over 35 years. Over that time span, 1H imaging flourished and was adopted worldwide with an endless array of applications and imaging approaches, making magnetic resonance an indispensable pillar of biomedical diagnostic imaging. For many years during this timeframe, 19F imaging research continued at a slow pace as the various attributes of the technique were explored. However, over the last decade and particularly the last several years, the pace and clinical relevance of 19F imaging has exploded. In part, this is due to advances in MRI instrumentation, 19F/1H coil designs, and ultrafast pulse sequence development for both preclinical and clinical scanners. These achievements, coupled with interest in the molecular imaging of anatomy and physiology, and combined with a cadre of innovative agents, have brought the concept of 19F into early clinical evaluation. In this review, we attempt to provide a slice of this rich history of research and development, with a particular focus on liquid perfluorocarbon compound-based agents.
Collapse
Affiliation(s)
- Anne H. Schmieder
- Division of Cardiology, Washington University School of Medical, St. Louis, MO 63110, USA
| | - Shelton D. Caruthers
- Toshiba Medical Research Institute USA, Inc., Cleveland, OH 44143, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Jochen Keupp
- Philips Research Hamburg, Hamburg 22335, Germany
| | - Samuel A. Wickline
- Division of Cardiology, Washington University School of Medical, St. Louis, MO 63110, USA
| | - Gregory M. Lanza
- Division of Cardiology, Washington University School of Medical, St. Louis, MO 63110, USA
- Correspondence author.
| |
Collapse
|
23
|
Siloxane Nanoprobes for Labeling and Dual Modality Functional Imaging of Neural Stem Cells. Ann Biomed Eng 2015; 44:816-27. [PMID: 26597417 DOI: 10.1007/s10439-015-1514-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/13/2015] [Indexed: 12/15/2022]
Abstract
Cell therapy represents a promising therapeutic for a myriad of medical conditions, including cancer, traumatic brain injury, and cardiovascular disease among others. A thorough understanding of the efficacy and cellular dynamics of these therapies necessitates the ability to non-invasively track cells in vivo. Magnetic resonance imaging (MRI) provides a platform to track cells as a non-invasive modality with superior resolution and soft tissue contrast. We recently reported a new nanoprobe platform for cell labeling and imaging using fluorophore doped siloxane core nanoemulsions as dual modality ((1)H MRI/Fluorescence), dual-functional (oximetry/detection) nanoprobes. Here, we successfully demonstrate the labeling, dual-modality imaging, and oximetry of neural progenitor/stem cells (NPSCs) in vitro using this platform. Labeling at a concentration of 10 μL/10(4) cells with a 40%v/v polydimethylsiloxane core nanoemulsion, doped with rhodamine, had minimal effect on viability, no effect on migration, proliferation and differentiation of NPSCs and allowed for unambiguous visualization of labeled NPSCs by (1)H MR and fluorescence and local pO2 reporting by labeled NPSCs. This new approach for cell labeling with a positive contrast (1)H MR probe has the potential to improve mechanistic knowledge of current therapies, and guide the design of future cell therapies due to its clinical translatability.
Collapse
|
24
|
Davies GL, Brown A, Blackburn O, Tropiano M, Faulkner S, Beer PD, Davis JJ. Ligation driven (19)F relaxation enhancement in self-assembled Ln(III) complexes. Chem Commun (Camb) 2015; 51:2918-20. [PMID: 25586055 DOI: 10.1039/c4cc09952d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Strong bidentate ligation between a fluorinated isophthalate and binuclear lanthanide-DO3A species yields a new class of (19)F NMR agent with very high nuclear relaxation rates at physiologically-relevant pH.
Collapse
Affiliation(s)
- Gemma-Louise Davies
- Department of Chemistry, University of Oxford, South Parks Road, Oxford, OX1 3QR, UK.
| | | | | | | | | | | | | |
Collapse
|
25
|
Bernsen MR, Guenoun J, van Tiel ST, Krestin GP. Nanoparticles and clinically applicable cell tracking. Br J Radiol 2015; 88:20150375. [PMID: 26248872 DOI: 10.1259/bjr.20150375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In vivo cell tracking has emerged as a much sought after tool for design and monitoring of cell-based treatment strategies. Various techniques are available for pre-clinical animal studies, from which much has been learned and still can be learned. However, there is also a need for clinically translatable techniques. Central to in vivo cell imaging is labelling of cells with agents that can give rise to signals in vivo, that can be detected and measured non-invasively. The current imaging technology of choice for clinical translation is MRI in combination with labelling of cells with magnetic agents. The main challenge encountered during the cell labelling procedure is to efficiently incorporate the label into the cell, such that the labelled cells can be imaged at high sensitivity for prolonged periods of time, without the labelling process affecting the functionality of the cells. In this respect, nanoparticles offer attractive features since their structure and chemical properties can be modified to facilitate cellular incorporation and because they can carry a high payload of the relevant label into cells. While these technologies have already been applied in clinical trials and have increased the understanding of cell-based therapy mechanism, many challenges are still faced.
Collapse
Affiliation(s)
- Monique R Bernsen
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands.,2 Department of Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Jamal Guenoun
- 1 Department of Radiology, Erasmus MC, Rotterdam, Netherlands
| | | | | |
Collapse
|
26
|
Srinivas M, Tel J, Schreibelt G, Bonetto F, Cruz LJ, Amiri H, Heerschap A, Figdor CG, de Vries IJM. PLGA-encapsulated perfluorocarbon nanoparticles for simultaneous visualization of distinct cell populations by 19F MRI. Nanomedicine (Lond) 2015; 10:2339-48. [DOI: 10.2217/nnm.15.76] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: In vivo imaging using 19F MRI is advantageous, due to its ability to quantify cell numbers, but is limited for a lack of suitable labels. Here, we formulate two stable and clinically applicable labels for tracking two populations of primary human dendritic cells (DCs) simultaneously. Materials & methods: Plasmacytoid and myeloid DCs are able to take up sufficient nanoparticles (200 nm) for imaging (1012 19F's per cell), despite being relatively nonphagocytic. Results: Clinically relevant numbers of labeled DCs could be imaged in about 10 min, even on a clinical scanner. Conclusion: We demonstrate the use of perfluorocarbon nanoparticles for simultaneous 19F MRI of distinct cell populations in a clinical setting, without spectroscopic imaging.
Collapse
Affiliation(s)
- Mangala Srinivas
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jurjen Tel
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Fernando Bonetto
- Instituto de Física del Litoral (CONICET – UNL), Santa Fe, Argentina
| | - Luis-Javier Cruz
- Molecular Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Houshang Amiri
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Arend Heerschap
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, & Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
27
|
Ji Y, Waiczies H, Winter L, Neumanova P, Hofmann D, Rieger J, Mekle R, Waiczies S, Niendorf T. Eight-channel transceiver RF coil array tailored for ¹H/¹⁹F MR of the human knee and fluorinated drugs at 7.0 T. NMR IN BIOMEDICINE 2015; 28:726-737. [PMID: 25916199 DOI: 10.1002/nbm.3300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 06/04/2023]
Abstract
The purpose of this study was to evaluate the feasibility of an eight-channel dual-tuned transceiver surface RF coil array for combined (1)H/(19)F MR of the human knee at 7.0 T following application of (19)F-containing drugs. The (1)H/(19)F RF coil array includes a posterior module with two (1)H loop elements and two anterior modules, each consisting of one (1)H and two (19)F elements. The decoupling of neighbor elements is achieved by a shared capacitor. Electromagnetic field simulations were performed to afford uniform transmission fields and to be in accordance with RF safety guidelines. Localized (19)F MRS was conducted with 47 and 101 mmol/L of flufenamic acid (FA) – a (19)F-containing non-steroidal anti-inflammatory drug – to determine T1 and T2 and to study the (19)F signal-to-dose relationship. The suitability of the proposed approach for (1)H/(19)F MR was examined in healthy subjects. Reflection coefficients of each channel were less than -17 dB and coupling between channels was less than -11 dB. Q(L)/Q(U) was less than 0.5 for all elements. MRS results demonstrated signal stability with 1% variation. T1 and T2 relaxation times changed with concentration of FA: T1 /T2 = 673/31 ms at 101 mmol/L and T1 /T2 = 616/26 ms at 47 mmol/L. A uniform signal and contrast across the patella could be observed in proton imaging. The sensitivity of the RF coil enabled localization of FA ointment administrated to the knee with an in-plane spatial resolution of (1.5 × 1.5) mm(2) achieved in a total scan time of approximately three minutes, which is well suited for translational human studies. This study shows the feasibility of combined (1)H/(19)F MRI of the knee at 7.0 T and proposes T1 and T2 mapping methods for quantifying fluorinated drugs in vivo. Further technological developments are necessary to promote real-time bioavailability studies and quantification of (19)F-containing medicinal compounds in vivo.
Collapse
Affiliation(s)
- Yiyi Ji
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Institute of Biophysics and Biomedical Engineering, Faculty of Sciences of the University of Lisbon, Lisbon, Portugal
| | - Helmar Waiczies
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
- MRI.TOOLS GmbH, Berlin, Germany
| | - Lukas Winter
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Pavla Neumanova
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniela Hofmann
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Ralf Mekle
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Sonia Waiczies
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility (BUFF), Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
28
|
Gaudet JM, Ribot EJ, Chen Y, Gilbert KM, Foster PJ. Tracking the fate of stem cell implants with fluorine-19 MRI. PLoS One 2015; 10:e0118544. [PMID: 25767871 PMCID: PMC4358825 DOI: 10.1371/journal.pone.0118544] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/20/2015] [Indexed: 01/13/2023] Open
Abstract
Background In this study we used cellular magnetic resonance imaging (MRI) to detect mesenchymal stem cells (MSC) labeled with a Fluorine-19 (19F) agent. 19F-MRI offers unambiguous detection and in vivo quantification of labeled cells. Methods We investigated two common stem cell transplant mouse models: an immune competent, syngeneic transplant model and an immune compromised, xenograft transplant model. 19F labelled stem cells were implanted intramuscularly into the hindlimb of healthy mice. The transplant was then monitored for up to 17 days using 19F-MRI, after which the tissue was excised for fluorescence microscopy and immunohistochemisty. Results Immediately following transplantation, 19F-MRI quantification correlated very well with the expected cell number in both models. The 19F signal decreased over time in both models, with a more rapid decrease in the syngeneic model. By endpoint, only 2/7 syngeneic mice had any detectable 19F signal. In the xenograft model, all mice had detectable signal at endpoint. Fluorescence microscopy and immunohistochemistry were used to show that the 19F signal was related to the presence of bystander labeled macrophages, and not original MSC. Conclusions Our results show that 19F-MRI is an excellent tool for verifying the delivery of therapeutic cells early after transplantation. However, in certain circumstances the transfer of cellular label to other bystander cells may confuse interpretation of the long-term fate of the transplanted cells.
Collapse
Affiliation(s)
- Jeffrey M. Gaudet
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
- * E-mail:
| | - Emeline J. Ribot
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
| | - Yuhua Chen
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
| | - Kyle M. Gilbert
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
| | - Paula J. Foster
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
29
|
Cell tracking using 19F magnetic resonance imaging: Technical aspects and challenges towards clinical applications. Eur Radiol 2014; 25:726-35. [DOI: 10.1007/s00330-014-3474-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 10/03/2014] [Accepted: 10/16/2014] [Indexed: 01/03/2023]
|
30
|
Tirotta I, Dichiarante V, Pigliacelli C, Cavallo G, Terraneo G, Bombelli FB, Metrangolo P, Resnati G. (19)F magnetic resonance imaging (MRI): from design of materials to clinical applications. Chem Rev 2014; 115:1106-29. [PMID: 25329814 DOI: 10.1021/cr500286d] [Citation(s) in RCA: 340] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ilaria Tirotta
- Laboratory of Nanostructured Fluorinated Materials (NFMLab), Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta" and ‡Fondazione Centro Europeo Nanomedicina, Politecnico di Milano , Milan 20131, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Naumova AV, Modo M, Moore A, Murry CE, Frank JA. Clinical imaging in regenerative medicine. Nat Biotechnol 2014; 32:804-18. [PMID: 25093889 PMCID: PMC4164232 DOI: 10.1038/nbt.2993] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/15/2014] [Indexed: 01/09/2023]
Abstract
In regenerative medicine, clinical imaging is indispensable for characterizing damaged tissue and for measuring the safety and efficacy of therapy. However, the ability to track the fate and function of transplanted cells with current technologies is limited. Exogenous contrast labels such as nanoparticles give a strong signal in the short term but are unreliable long term. Genetically encoded labels are good both short- and long-term in animals, but in the human setting they raise regulatory issues related to the safety of genomic integration and potential immunogenicity of reporter proteins. Imaging studies in brain, heart and islets share a common set of challenges, including developing novel labeling approaches to improve detection thresholds and early delineation of toxicity and function. Key areas for future research include addressing safety concerns associated with genetic labels and developing methods to follow cell survival, differentiation and integration with host tissue. Imaging may bridge the gap between cell therapies and health outcomes by elucidating mechanisms of action through longitudinal monitoring.
Collapse
Affiliation(s)
- Anna V Naumova
- 1] Department of Radiology, University of Washington, Seattle, Washington, USA. [2] Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA. [3] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Michel Modo
- 1] McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [2] Centre for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [3] Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. [4] Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna Moore
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Charles E Murry
- 1] Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA. [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA. [3] Department of Pathology, University of Washington, Seattle, Washington, USA. [4] Department of Bioengineering, University of Washington, Seattle, Washington, USA. [5] Department of Medicine/Cardiology, University of Washington, Seattle, Washington, USA
| | - Joseph A Frank
- 1] Radiology and Imaging Sciences, Clinical, National Institutes of Health, Bethesda, Maryland, USA. [2] National Institutes of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
32
|
Tirotta I, Mastropietro A, Cordiglieri C, Gazzera L, Baggi F, Baselli G, Bruzzone MG, Zucca I, Cavallo G, Terraneo G, Baldelli Bombelli F, Metrangolo P, Resnati G. A Superfluorinated Molecular Probe for Highly Sensitive in Vivo19F-MRI. J Am Chem Soc 2014; 136:8524-7. [DOI: 10.1021/ja503270n] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
33
|
Ribot EJ, Gaudet JM, Chen Y, Gilbert KM, Foster PJ. In vivo MR detection of fluorine-labeled human MSC using the bSSFP sequence. Int J Nanomedicine 2014; 9:1731-9. [PMID: 24748787 PMCID: PMC3986292 DOI: 10.2147/ijn.s59127] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mesenchymal stem cells (MSC) are used to restore deteriorated cell environments. There is a need to specifically track these cells following transplantation in order to evaluate different methods of implantation, to follow their migration within the body, and to quantify their accumulation at the target. Cellular magnetic resonance imaging (MRI) using fluorine-based nanoemulsions is a great means to detect these transplanted cells in vivo because of the high specificity for fluorine detection and the capability for precise quantification. This technique, however, has low sensitivity, necessitating improvement in MR sequences. To counteract this issue, the balanced steady-state free precession (bSSFP) imaging sequence can be of great interest due to the high signal-to-noise ratio (SNR). Furthermore, it can be applied to obtain 3D images within short acquisition times. In this paper, bSSFP provided accurate quantification of samples of the perfluorocarbon Cell Sense-labeled cells in vitro. Cell Sense was internalized by human MSC (hMSC) without adverse alterations in cell viability or differentiation into adipocytes/osteocytes. The bSSFP sequence was applied in vivo to track and quantify the signals from both Cell Sense-labeled and iron-labeled hMSC after intramuscular implantation. The fluorine signal was observed to decrease faster and more significantly than the volume of iron-associated voids, which points to the advantage of quantifying the fluorine signal and the complexity of quantifying signal loss due to iron.
Collapse
Affiliation(s)
- Emeline J Ribot
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
| | - Jeffrey M Gaudet
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Yuhua Chen
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
| | - Kyle M Gilbert
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
| | - Paula J Foster
- Imaging Research Laboratories, Robarts Research Institute, London, ON, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| |
Collapse
|
34
|
Hamilton AM, Mallett C, Foster PJ. High-resolution MRI and nanoparticles: the future of brain imaging. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT: Cellular MRI uses superparamagnetic iron oxide nanoparticles to label cells (in vitro or in vivo) for detection in magnetic resonance images. The infiltration of inflammatory macrophages can be visualized in brain diseases, such as multiple sclerosis, stroke and Alzheimer‘s disease, and correlates with disease severity and responses to treatments. Mesenchymal stromal cells, neural stem cells and immune cells used as cell therapies in CNS diseases can be tracked in vivo over time to determine their migration and dispersion. Tracking labeled cancer cells provides information about metastasis and proliferative status in preclinical tumor models. Ongoing technical improvements come from the development of new particles, the use of fluorine-based contrast agents and the refinement of high-field MRI for cell tracking.
Collapse
Affiliation(s)
- Amanda M Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, ON, N6A 5K8, Canada
| | - Christiane Mallett
- Imaging Research Laboratories, Robarts Research Institute, London, ON, N6A 5K8, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, ON, Canada
| |
Collapse
|
35
|
Kooreman NG, Ransohoff JD, Wu JC. Tracking gene and cell fate for therapeutic gain. NATURE MATERIALS 2014; 13:106-9. [PMID: 24452344 PMCID: PMC4892936 DOI: 10.1038/nmat3868] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Parallel advances in molecular imaging modalities and in gene- and cell-based therapeutics have significantly advanced their respective fields. Here we discuss how the collaborative, preclinical intersection of these technologies will facilitate more informed and effective clinical translation of relevant therapies.
Collapse
Affiliation(s)
- Nigel G. Kooreman
- Stanford Cardiovascular Institute
- Departments of Medicine and Radiology (Molecular Imaging Program)
| | - Julia D. Ransohoff
- Stanford Cardiovascular Institute
- Departments of Medicine and Radiology (Molecular Imaging Program)
| | - Joseph C. Wu
- Stanford Cardiovascular Institute
- Departments of Medicine and Radiology (Molecular Imaging Program)
- Institute of Stem Cell Biology and Regenerative Medicine
| |
Collapse
|
36
|
Rolfe BE, Blakey I, Squires O, Peng H, Boase NRB, Alexander C, Parsons PG, Boyle GM, Whittaker AK, Thurecht KJ. Multimodal Polymer Nanoparticles with Combined 19F Magnetic Resonance and Optical Detection for Tunable, Targeted, Multimodal Imaging in Vivo. J Am Chem Soc 2014; 136:2413-9. [DOI: 10.1021/ja410351h] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | | | | | | | - Cameron Alexander
- School
of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Peter G. Parsons
- Queensland
Institute for Medical Research, The Royal Brisbane Hospital, Herston, Queensland 4006, Australia
| | - Glen M. Boyle
- Queensland
Institute for Medical Research, The Royal Brisbane Hospital, Herston, Queensland 4006, Australia
| | | | | |
Collapse
|
37
|
Patrick MJ, Janjic JM, Teng H, O’Hear MR, Brown CW, Stokum JA, Schmidt BF, Ahrens ET, Waggoner AS. Intracellular pH measurements using perfluorocarbon nanoemulsions. J Am Chem Soc 2013; 135:18445-57. [PMID: 24266634 PMCID: PMC4208472 DOI: 10.1021/ja407573m] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We report the synthesis and formulation of unique perfluorocarbon (PFC) nanoemulsions enabling intracellular pH measurements in living cells via fluorescent microscopy and flow cytometry. These nanoemulsions are formulated to readily enter cells upon coincubation and contain two cyanine-based fluorescent reporters covalently bound to the PFC molecules, specifically Cy3-PFC and CypHer5-PFC conjugates. The spectral and pH-sensing properties of the nanoemulsions were characterized in vitro and showed the unaltered spectral behavior of dyes after formulation. In rat 9L glioma cells loaded with nanoemulsion, the local pH of nanoemulsions was longitudinally quantified using optical microscopy and flow cytometry and displayed a steady decrease in pH to a level of 5.5 over 3 h, indicating rapid uptake of nanoemulsion to acidic compartments. Overall, these reagents enable real-time optical detection of intracellular pH in living cells in response to pharmacological manipulations. Moreover, recent approaches for in vivo cell tracking using magnetic resonance imaging (MRI) employ intracellular PFC nanoemulsion probes to track cells using (19)F MRI. However, the intracellular fate of these imaging probes is poorly understood. The pH-sensing nanoemulsions allow the study of the fate of the PFC tracer inside the labeled cell, which is important for understanding the PFC cell loading dynamics, nanoemulsion stability and cell viability over time.
Collapse
Affiliation(s)
- Michael J. Patrick
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA
| | - Jelena M. Janjic
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA
| | - Haibing Teng
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA
| | - Meredith R. O’Hear
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA
| | - Cortlyn W. Brown
- Department of Biological Sciences, University of Chicago, Chicago, IL
| | - Jesse A. Stokum
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA
| | - Brigitte F. Schmidt
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA
| | - Eric T. Ahrens
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, PA
| | - Alan S. Waggoner
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA
| |
Collapse
|
38
|
Aspord C, Laurin D, Janier MF, Mandon CA, Thivolet C, Villiers C, Mowat P, Madec AM, Tillement O, Perriat P, Louis C, Bérard F, Marche PN, Plumas J, Billotey C. Paramagnetic nanoparticles to track and quantify in vivo immune human therapeutic cells. NANOSCALE 2013; 5:11409-11415. [PMID: 23838997 DOI: 10.1039/c3nr34240a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
This study aims to investigate gadolinium-based nanoparticles (Gd-HNP) for in vitro labeling of human plasmacytoid dendritic cells (HuPDC) to allow for in vivo tracking and HuPDC quantifying using magnetic resonance imaging (MRI) following parenteral injection. Human plasmacytoid DC were labeled (LabHuPDC) with fluorescent Gd-HNP (Gd-FITC-HNP) and injected via intraperitoneal and intravenous routes in 4-5 NOD-SCID β2m(-/-)mice (treated mice = TM). Control mice (CM) were similarly injected with unlabeled HuPDC. In vivo 7 T MRI was performed 24 h later and all spleens were removed in order to measure Gd and fluorescence contents and identify HuPDC. Gd-FITC-HNP efficiently labeled HuPDC (0.05 to 0.1 pg per cell), without altering viability and activation properties. The magnetic resonance (MR) signal was exclusively due to HuPDC. The normalized MR splenic intensity for TM was significantly higher than for CM (p < 0.024), and highly correlated with the spleen Gd content (r = 0.97), and the number of HuPDC found in the spleen (r = 0.94). Gd-FITC-HNP allowed for in vivo tracking and HuPDC quantifying by means of MRI following parenteral injection, with very high sensitivity (<3000 cells per mm(3)). The safety of these new nanoparticle types must be confirmed via extensive toxicology tests including in vivo stability and biodistribution studies.
Collapse
Affiliation(s)
- Caroline Aspord
- R&D Laboratory, Etablissement Français du Sang Rhône Alpes, La Tronche, F-38701 France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chen J, Pan H, Lanza GM, Wickline SA. Perfluorocarbon nanoparticles for physiological and molecular imaging and therapy. Adv Chronic Kidney Dis 2013; 20:466-78. [PMID: 24206599 DOI: 10.1053/j.ackd.2013.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/20/2013] [Accepted: 08/20/2013] [Indexed: 11/11/2022]
Abstract
Herein, we review the use of non-nephrotoxic perfluorocarbon nanoparticles (PFC NPs) for noninvasive detection and therapy of kidney diseases, and we provide a synopsis of other related literature pertinent to their anticipated clinical application. Recent reports indicate that PFC NPs allow for quantitative mapping of kidney perfusion and oxygenation after ischemia-reperfusion injury with the use of a novel multinuclear (1)H/(19)F magnetic resonance imaging approach. Furthermore, when conjugated with targeting ligands, the functionalized PFC NPs offer unique and quantitative capabilities for imaging inflammation in the kidney of atherosclerotic ApoE-null mice. In addition, PFC NPs can facilitate drug delivery for treatment of inflammation, thrombosis, and angiogenesis in selected conditions that are comorbidities for kidney failure. The excellent safety profile of PFC NPs with respect to kidney injury positions these nanomedicine approaches as promising diagnostic and therapeutic candidates for treating and following acute and chronic kidney diseases.
Collapse
|
40
|
Dewitte H, Geers B, Liang S, Himmelreich U, Demeester J, De Smedt SC, Lentacker I. Design and evaluation of theranostic perfluorocarbon particles for simultaneous antigen-loading and 19F-MRI tracking of dendritic cells. J Control Release 2013; 169:141-9. [DOI: 10.1016/j.jconrel.2013.03.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/12/2013] [Accepted: 03/14/2013] [Indexed: 12/30/2022]
|
41
|
Ahrens ET, Zhong J. In vivo MRI cell tracking using perfluorocarbon probes and fluorine-19 detection. NMR IN BIOMEDICINE 2013; 26:860-71. [PMID: 23606473 PMCID: PMC3893103 DOI: 10.1002/nbm.2948] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/29/2013] [Accepted: 02/21/2013] [Indexed: 05/08/2023]
Abstract
This article presents a brief review of preclinical in vivo cell-tracking methods and applications using perfluorocarbon (PFC) probes and fluorine-19 ((19) F) MRI detection. Detection of the (19) F signal offers high cell specificity and quantification ability in spin density-weighted MR images. We discuss the compositions of matter, methods and applications of PFC-based cell tracking using ex vivo and in situ PFC labeling in preclinical studies of inflammation and cellular therapeutics. We also address the potential applicability of (19) F cell tracking to clinical trials.
Collapse
Affiliation(s)
- Eric T Ahrens
- Department of Biological Sciences and Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
42
|
Cassidy MC, Chan HR, Ross BD, Bhattacharya PK, Marcus CM. In vivo magnetic resonance imaging of hyperpolarized silicon particles. NATURE NANOTECHNOLOGY 2013; 8:363-368. [PMID: 23644571 DOI: 10.1038/nnano.2013.65] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 03/19/2013] [Indexed: 06/02/2023]
Abstract
Silicon-based micro- and nanoparticles have gained popularity in a wide range of biomedical applications due to their biocompatibility and biodegradability in vivo, as well as their flexible surface chemistry, which allows drug loading, functionalization and targeting. Here, we report direct in vivo imaging of hyperpolarized (29)Si nuclei in silicon particles by magnetic resonance imaging. Natural physical properties of silicon provide surface electronic states for dynamic nuclear polarization, extremely long depolarization times, insensitivity to the in vivo environment or particle tumbling, and surfaces favourable for functionalization. Potential applications to gastrointestinal, intravascular and tumour perfusion imaging at subpicomolar concentrations are presented. These results demonstrate a new background-free imaging modality applicable to a range of inexpensive, readily available and biocompatible silicon particles.
Collapse
Affiliation(s)
- M C Cassidy
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| | | | | | | | | |
Collapse
|
43
|
Dekaban GA, Hamilton AM, Fink CA, Au B, de Chickera SN, Ribot EJ, Foster PJ. Tracking and evaluation of dendritic cell migration by cellular magnetic resonance imaging. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 5:469-83. [PMID: 23633389 DOI: 10.1002/wnan.1227] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/28/2013] [Accepted: 03/19/2013] [Indexed: 01/15/2023]
Abstract
Cellular magnetic resonance imaging (MRI) is a means by which cells labeled ex vivo with a contrast agent can be detected and tracked over time in vivo. This technology provides a noninvasive method with which to assess cell-based therapies in vivo. Dendritic cell (DC)-based vaccines are a promising cancer immunotherapy, but its success is highly dependent on the injected DC migrating to a secondary lymphoid organ such as a nearby lymph node. There the DC can interact with T cells to elicit a tumor-specific immune response. It is important to verify DC migration in vivo using a noninvasive imaging modality, such as cellular MRI, so that important information regarding the anatomical location and persistence of the injected DC in a targeted lymph node can be provided. An understanding of DC biology is critical in ascertaining how to label DC with sufficient contrast agent to render them detectable by MRI. While iron oxide nanoparticles provide the best sensitivity for detection of DC in vivo, a clinical grade iron oxide agent is not currently available. A clinical grade (19) Fluorine-based perfluorcarbon nanoemulsion is available but is less sensitive, and its utility to detect DC migration in humans remains to be demonstrated using clinical scanners presently available. The ability to quantitatively track DC migration in vivo can provide important information as to whether different DC maturation and activation protocols result in improved DC migration efficiency which will determine the vaccine's immunogenicity and ultimately the tumor immunotherapy's outcome in humans.
Collapse
Affiliation(s)
- Gregory A Dekaban
- BioTherapeutics Research Laboratories, Robarts Research Institute and Department of Microbiology & Immunology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
44
|
van Heeswijk RB, De Blois J, Kania G, Gonzales C, Blyszczuk P, Stuber M, Eriksson U, Schwitter J. Selective in vivo visualization of immune-cell infiltration in a mouse model of autoimmune myocarditis by fluorine-19 cardiac magnetic resonance. Circ Cardiovasc Imaging 2013; 6:277-84. [PMID: 23343515 DOI: 10.1161/circimaging.112.000125] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The goal of this study was to characterize the performance of fluorine-19 ((19)F) cardiac magnetic resonance (CMR) for the specific detection of inflammatory cells in a mouse model of myocarditis. Intravenously administered perfluorocarbons are taken up by infiltrating inflammatory cells and can be detected by (19)F-CMR. (19)F-labeled cells should, therefore, generate an exclusive signal at the inflamed regions within the myocardium. METHODS AND RESULTS Experimental autoimmune myocarditis was induced in BALB/c mice. After intravenous injection of 2×200 µL of a perfluorocarbon on day 19 and 20 (n=9) after immunization, in vivo (19)F-CMR was performed at the peak of myocardial inflammation (day 21). In 5 additional animals, perfluorocarbon combined with FITC (fluorescein isothiocyanate) was administered for postmortem immunofluorescence and flow-cytometry analyses. Control experiments were performed in 9 animals. In vivo (19)F-CMR detected myocardial inflammation in all experimental autoimmune myocarditis-positive animals. Its resolution was sufficient to identify even small inflammatory foci, that is, at the surface of the right ventricle. Postmortem immunohistochemistry and flow cytometry confirmed the presence of perfluorocarbon in macrophages, dendritic cells, and granulocytes, but not in lymphocytes. The myocardial volume of elevated (19)F signal (rs=0.96; P<0.001), the (19)F signal-to-noise ratio (rs=0.92; P<0.001), and the (19)F signal integral (rs=0.96; P<0.001) at day 21 correlated with the histological myocarditis severity score. CONCLUSIONS In vivo (19)F-CMR was successfully used to visualize the inflammation specifically and robustly in experimental autoimmune myocarditis, and thus allowed for an unprecedented insight into the involvement of inflammatory cells in the disease process.
Collapse
|
45
|
Interaction of tumor cells with the immune system: implications for dendritic cell therapy and cancer progression. Drug Discov Today 2013; 18:35-42. [DOI: 10.1016/j.drudis.2012.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/30/2012] [Accepted: 07/18/2012] [Indexed: 01/21/2023]
|
46
|
Srinivas M, Boehm-Sturm P, Figdor CG, de Vries IJ, Hoehn M. Labeling cells for in vivo tracking using 19F MRI. Biomaterials 2012; 33:8830-40. [DOI: 10.1016/j.biomaterials.2012.08.048] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 08/22/2012] [Indexed: 12/11/2022]
|
47
|
Diou O, Tsapis N, Fattal E. Targeted nanotheranostics for personalized cancer therapy. Expert Opin Drug Deliv 2012; 9:1475-87. [DOI: 10.1517/17425247.2012.736486] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
48
|
Aarntzen EHJG, Srinivas M, Radu CG, Punt CJA, Boerman OC, Figdor CG, Oyen WJG, de Vries IJM. In vivo imaging of therapy-induced anti-cancer immune responses in humans. Cell Mol Life Sci 2012; 70:2237-57. [PMID: 23052208 PMCID: PMC3676735 DOI: 10.1007/s00018-012-1159-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/27/2012] [Accepted: 09/03/2012] [Indexed: 12/16/2022]
Abstract
Immunotherapy aims to re-engage and revitalize the immune system in the fight against cancer. Research over the past decades has shown that the relationship between the immune system and human cancer is complex, highly dynamic, and variable between individuals. Considering the complexity, enormous effort and costs involved in optimizing immunotherapeutic approaches, clinically applicable tools to monitor therapy-induced immune responses in vivo are most warranted. However, the development of such tools is complicated by the fact that a developing immune response encompasses several body compartments, e.g., peripheral tissues, lymph nodes, lymphatic and vascular systems, as well as the tumor site itself. Moreover, the cells that comprise the immune system are not static but constantly circulate through the vascular and lymphatic system. Molecular imaging is considered the favorite candidate to fulfill this task. The progress in imaging technologies and modalities has provided a versatile toolbox to address these issues. This review focuses on the detection of therapy-induced anticancer immune responses in vivo and provides a comprehensive overview of clinically available imaging techniques as well as perspectives on future developments. In the discussion, we will focus on issues that specifically relate to imaging of the immune system and we will discuss the strengths and limitations of the current clinical imaging techniques. The last section provides future directions that we envision to be crucial for further development.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Helfer BM, Balducci A, Sadeghi Z, O'Hanlon C, Hijaz A, Flask CA, Wesa A. ¹⁹F MRI tracer preserves in vitro and in vivo properties of hematopoietic stem cells. Cell Transplant 2012; 22:87-97. [PMID: 22862925 DOI: 10.3727/096368912x653174] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have numerous therapeutic applications including immune reconstitution, enzyme replacement, regenerative medicine, and immunomodulation. The trafficking and persistence of these cells after administration is a fundamental question for future therapeutic applications of HSCs. Here, we describe the safe and efficacious labeling of human CD34(+) HSCs with a novel, self-delivering perfluorocarbon ¹⁹F magnetic resonance imaging (MRI) tracer, which has recently been authorized for use in a clinical trial to track therapeutic cells. While various imaging contrast agents have been used to track cellular therapeutics, the impact of this MRI tracer on HSC function has not previously been studied. Both human CD34(+) and murine bone marrow (BM) HSCs were effectively labeled with the MRI tracer, with only a slight reduction in viability, relative to mock-labeled cells. In a pilot study, ¹⁹F MRI enabled the rapid evaluation of HSC delivery/retention following administration into a rat thigh muscle, revealing the dispersal of HSCs after injection, but not after surgical implantation. To investigate effects on cell functionality, labeled and unlabeled human HSCs were tested in in vitro colony forming unit (CFU) assays, which resulted in equal numbers of total CFU as well as individual CFU types, indicating that labeling did not alter multipotency. Cobblestone assay forming cell precursor frequency was also unaffected, providing additional evidence that stem cell function was preserved after labeling. In vivo tests of multipotency and reconstitution studies in mice with murine BM containing labeled HSCs resulted in normal development of CFU in the spleen, compared to unlabeled cells, and reconstitution of both lymphoid and myeloid compartments. The lack of interference in these complex biological processes provides strong evidence that the function and therapeutic potential of the HSCs are likely maintained after labeling. These data support the safety and efficacy of the MRI tracer for clinical tracking of human stem cells.
Collapse
Affiliation(s)
- Brooke M Helfer
- Celsense, Inc., Department of Research and Development, Pittsburgh, PA, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Andralojc K, Srinivas M, Brom M, Joosten L, de Vries IJM, Eizirik DL, Boerman OC, Meda P, Gotthardt M. Obstacles on the way to the clinical visualisation of beta cells: looking for the Aeneas of molecular imaging to navigate between Scylla and Charybdis. Diabetologia 2012; 55:1247-57. [PMID: 22358499 PMCID: PMC3328679 DOI: 10.1007/s00125-012-2491-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 01/09/2012] [Indexed: 12/25/2022]
Abstract
For more than a decade, researchers have been trying to develop non-invasive imaging techniques for the in vivo measurement of viable pancreatic beta cells. However, in spite of intense research efforts, only one tracer for positron emission tomography (PET) imaging is currently under clinical evaluation. To many diabetologists it may remain unclear why the imaging world struggles to develop an effective method for non-invasive beta cell imaging (BCI), which could be useful for both research and clinical purposes. Here, we provide a concise overview of the obstacles and challenges encountered on the way to such BCI, in both native and transplanted islets. We discuss the major difficulties posed by the anatomical and cell biological features of pancreatic islets, as well as the chemical and physical limits of the main imaging modalities, with special focus on PET, SPECT and MRI. We conclude by indicating new avenues for future research in the field, based on several remarkable recent results.
Collapse
Affiliation(s)
- K. Andralojc
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - M. Srinivas
- Department of Tumour Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - M. Brom
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - L. Joosten
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - I. J. M. de Vries
- Department of Tumour Immunology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - D. L. Eizirik
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - O. C. Boerman
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| | - P. Meda
- Deparment of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - M. Gotthardt
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, the Netherlands
| |
Collapse
|