1
|
Pruller J, Pham TT, Blower JE, Charoenphun P, Volpe A, Sunassee K, Mullen GED, Blower PJ, Smith RAG, Ma MT. An indium-111-labelled membrane-targeted peptide for cell tracking with radionuclide imaging. RSC Chem Biol 2023; 4:65-73. [PMID: 36685254 PMCID: PMC9811519 DOI: 10.1039/d2cb00164k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
Cell labelling agents that enable longitudinal in vivo tracking of administered cells will support the clinical development of cell-based therapies. Radionuclide imaging with gamma and positron-emitting radioisotopes can provide quantitative and longitudinal mapping of cells in vivo. To make this widely accessible and adaptable to a range of cell types, new, versatile and simple methods for directly radiolabelling cells are required. We have developed [111In]In-DTPA-CTP, the first example of a radiolabelled peptide that binds to the extracellular membrane of cells, for tracking cell distribution in vivo using Single Photon Emission Computed Tomography (SPECT). [111In]In-DTPA-CTP consists of (i) myristoyl groups for insertion into the phospholipid bilayer, (ii) positively charged lysine residues for electrostatic association with negatively charged phospholipid groups at the cell surface and (iii) a diethylenetriamine pentaacetate derivative that coordinates the γ-emitting radiometal, [111In]In3+. [111In]In-DTPA-CTP binds to 5T33 murine myeloma cells, enabling qualitative SPECT tracking of myeloma cells' accumulation in lungs immediately after intravenous administration. This is the first report of a radiolabelled cell-membrane binding peptide for use in cell tracking.
Collapse
Affiliation(s)
- Johanna Pruller
- Randall Division of Cell and Molecular Biophysics, King's College London UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Truc Thuy Pham
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Julia E Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Putthiporn Charoenphun
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
- Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University Bangkok Thailand
| | - Alessia Volpe
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Kavitha Sunassee
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Gregory E D Mullen
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Richard A G Smith
- MRC Centre for Transplantation, King's College London, Guy's Hospital London UK
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| |
Collapse
|
2
|
Friberger I, Gontu V, Harris RA, Tran TA, Lundberg J, Holmin S. Phenotyping of Macrophages After Radiolabeling and Safety of Intra-arterial Transplantation Assessed by SPECT/CT and MRI. Cell Transplant 2023; 32:9636897231212780. [PMID: 38009543 PMCID: PMC10683405 DOI: 10.1177/09636897231212780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 11/29/2023] Open
Abstract
Cell therapy is an integral modality of regenerative medicine. Macrophages are known for their sensitivity to activation stimuli and capability to recruit other immune cells to the sites of injury and healing. In addition, the route of administration can impact engraftment and efficacy of cell therapy, and modern neuro-interventional techniques provide the possibility for selective intra-arterial (IA) delivery to the central nervous system (CNS) with very low risk. The effects of radiolabelling and catheter transport on differentially activated macrophages were evaluated. Furthermore, the safety of selective IA administration of these macrophages to the rabbit brain was assessed by single-photon emission computed tomography/computed tomography (SPECT/CT) and ultra-high-field (9.4 T) magnetic resonance imaging (MRI). Cells were successfully labeled with (111In)In-(oxinate)3 and passed through a microcatheter with preserved phenotype. No cells were retained in the healthy rabbit brain after IA administration, and no adverse events could be observed either 1 h (n = 6) or 24 h (n = 2) after cell administration. The procedure affected both lipopolysaccharide/gamma interferon (LPS/IFNγ) activated cells and interleukin 4 (IL4), interleukin 10 (IL10)/transforming growth factor beta 1 (TGFβ1) activated cells to some degree. The LPS/IFNγ activated cells had a significant increase in their phagocytotic function. Overall, the major impact on the cell phenotypes was due to the radiolabeling and not passage through the catheter. Unstimulated cells were substantially affected by both radiolabeling and catheter administration and are hence not suited for this procedure, while both activated macrophages retained their initial phenotypes. In conclusion, activated macrophages are suitable candidates for targeted IA administration without adverse effects on normal, healthy brain parenchyma.
Collapse
Affiliation(s)
- Ida Friberger
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Vamsi Gontu
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Thuy A Tran
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiopharmacy, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
4
|
Sato N, Choyke PL. Whole-Body Imaging to Assess Cell-Based Immunotherapy: Preclinical Studies with an Update on Clinical Translation. Mol Imaging Biol 2022; 24:235-248. [PMID: 34816284 PMCID: PMC8983636 DOI: 10.1007/s11307-021-01669-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/28/2022]
Abstract
In the past decades, immunotherapies against cancers made impressive progress. Immunotherapy includes a broad range of interventions that can be separated into two major groups: cell-based immunotherapies, such as adoptive T cell therapies and stem cell therapies, and immunomodulatory molecular therapies such as checkpoint inhibitors and cytokine therapies. Genetic engineering techniques that transduce T cells with a cancer-antigen-specific T cell receptor or chimeric antigen receptor have expanded to other cell types, and further modulation of the cells to enhance cancer targeting properties has been explored. Because cell-based immunotherapies rely on cells migrating to target organs or tissues, there is a growing interest in imaging technologies that non-invasively monitor transferred cells in vivo. Here, we review whole-body imaging methods to assess cell-based immunotherapy using a variety of examples. Following a review of preclinically used cell tracking technologies, we consider the status of their clinical translation.
Collapse
Affiliation(s)
- Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10/Rm. B3B406, 10 Center Dr, Bethesda, MD, 20892, USA.
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10/Rm. B3B69F, 10 Center Dr, Bethesda, MD, 20892, USA
| |
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
|
Xu Y, Zheng H, Schumacher D, Liehn EA, Slabu I, Rusu M. Recent Advancements of Specific Functionalized Surfaces of Magnetic Nano- and Microparticles as a Theranostics Source in Biomedicine. ACS Biomater Sci Eng 2021; 7:1914-1932. [PMID: 33856199 DOI: 10.1021/acsbiomaterials.0c01393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Magnetic nano- and microparticles (MNMPs) belong to a highly versatile class of colloids with actuator and sensor properties that have been broadly studied for their application in theranostics such as molecular imaging and drug delivery. The use of advanced biocompatible, biodegradable polymers and polyelectrolytes as MNMP coating materials is essential to ensure the stability of MNMPs and enable efficient drug release while at the same time preventing cytotoxic effects. In the past years, huge progress has been made in terms of the design of MNMPs. Especially, the understanding of coating formation with respect to control of drug loading and release kinetics on the molecular level has significantly advanced. In this review, recent advancements in the field of MNMP surface engineering and the applicability of MNMPs in research fields of medical imaging, diagnosis, and nanotherapeutics are presented and discussed. Furthermore, in this review the main emphasis is put on the manipulation of biological specimens and cell trafficking, for which MNMPs represent a favorable tool enabling transport processes of drugs through cell membranes. Finally, challenges and future perspectives for applications of MNMPs as theranostic nanomaterials are discussed.
Collapse
Affiliation(s)
- Yichen Xu
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Pauwelstr. 30, 52074 Aachen, Germany
| | - Huabo Zheng
- Department of Cardiology, Pulmonology, Angiology, and Intensive Care, University Hospital, RWTH Aachen, Pauwelstr. 30, 52074 Aachen, Germany
| | - David Schumacher
- Department of Anesthesiology, University Hospital, RWTH Aachen, 52074 Aachen, Germany
| | - Elisa Anamaria Liehn
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Pauwelstr. 30, 52074 Aachen, Germany.,Department of Cardiology, Pulmonology, Angiology, and Intensive Care, University Hospital, RWTH Aachen, Pauwelstr. 30, 52074 Aachen, Germany.,Department of Pathology, Institute of Pathology "Victor Babes", Splaiul Independentei nr. 99-101, Sector 5, 050096 Bucharest, Romania
| | - Ioana Slabu
- Institute of Applied Medical Engineering, Helmholtz Institute, Medical Faculty, RWTH Aachen, Pauwelstr. 20, 52074 Aachen, Germany
| | - Mihaela Rusu
- Department of Pathology, Institute of Pathology "Victor Babes", Splaiul Independentei nr. 99-101, Sector 5, 050096 Bucharest, Romania.,Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen, Pauwelstr. 30, 52074 Aachen, Germany
| |
Collapse
|
7
|
Kurebayashi Y, Choyke PL, Sato N. Imaging of cell-based therapy using 89Zr-oxine ex vivo cell labeling for positron emission tomography. Nanotheranostics 2021; 5:27-35. [PMID: 33391973 PMCID: PMC7738941 DOI: 10.7150/ntno.51391] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
With the rapid development of anti-cancer cell-based therapies, such as adoptive T cell therapies using tumor-infiltrating T cells, T cell receptor transduced T cells, and chimeric antigen receptor T cells, there has been a growing interest in imaging technologies to non-invasively track transferred cells in vivo. Cell tracking using ex vivo cell labeling with positron emitting radioisotopes for positron emission tomography (PET) imaging has potential advantages over single-photon emitting radioisotopes. These advantages include intrinsically higher resolution, higher sensitivity, and higher signal-to-background ratios. Here, we review the current status of recently developed Zirconium-89 (89Zr)-oxine ex vivo cell labeling with PET imaging focusing on its applications and future perspectives. Labeling of cells with 89Zr-oxine is completed in a series of relatively simple steps, and its low radioactivity doses required for imaging does not interfere with the proliferation or function of the labeled immune cells. Preclinical studies have revealed that 89Zr-oxine PET allows high-resolution in vivo tracking of labeled cells for 1-2 weeks after cell transfer both in mice and non-human primates. These results provide a strong rationale for the clinical translation of 89Zr-oxine PET-based imaging of cell-based therapy.
Collapse
Affiliation(s)
| | | | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
In vivo stem cell tracking using scintigraphy in a canine model of DMD. Sci Rep 2020; 10:10681. [PMID: 32606364 PMCID: PMC7327062 DOI: 10.1038/s41598-020-66388-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 05/07/2020] [Indexed: 12/28/2022] Open
Abstract
One of the main challenges in cell therapy for muscle diseases is to efficiently target the muscle. To address this issue and achieve better understanding of in vivo cell fate, we evaluated the relevance of a non-invasive cell tracking method in the Golden Retriever Muscular Dystrophy (GRMD) model, a well-recognised model of Duchenne Muscular Dystrophy (DMD). Mesoangioblasts were directly labelled with 111In-oxine, and injected through one of the femoral arteries. The scintigraphy images obtained provided the first quantitative mapping of the immediate biodistribution of mesoangioblasts in a large animal model of DMD. The results revealed that cells were trapped by the first capillary filters: the injected limb and the lung. During the days following injection, radioactivity was redistributed to the liver. In vitro studies, performed with the same cells prepared for injecting the animal, revealed prominent cell death and 111In release. In vivo, cell death resulted in 111In release into the vasculature that was taken up by the liver, resulting in a non-specific and non-cell-bound radioactive signal. Indirect labelling methods would be an attractive alternative to track cells on the mid- and long-term.
Collapse
|
9
|
Galli F, Aguilera JV, Palermo B, Markovic SN, Nisticò P, Signore A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res 2020; 39:89. [PMID: 32423420 PMCID: PMC7236372 DOI: 10.1186/s13046-020-01586-y] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-infiltrating immune cells play a key role against cancer. However, malignant cells are able to evade the immune response and establish a very complex balance in which different immune subtypes may drive tumor progression, metastatization and resistance to therapy. New immunotherapeutic approaches aim at restoring the natural balance and increase immune response against cancer by different mechanisms. The complexity of these interactions and the heterogeneity of immune cell subpopulations are a real challenge when trying to develop new immunotherapeutics and evaluate or predict their efficacy in vivo. To this purpose, molecular imaging can offer non-invasive diagnostic tools like radiopharmaceuticals, contrast agents or fluorescent dyes. These agents can be useful for preclinical and clinical purposes and can overcome [18F]FDG limitations in discriminating between true-progression and pseudo-progression. This review provides a comprehensive overview of immune cells involved in microenvironment, available immunotherapies and imaging agents to highlight the importance of new therapeutic biomarkers and their in vivo evaluation to improve the management of cancer patients.
Collapse
Affiliation(s)
- Filippo Galli
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, Roma, Italy.
| | - Jesus Vera Aguilera
- Department of oncology and Department of Immunology, Mayo Clinic, (MN), Rochester, USA
| | - Belinda Palermo
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Svetomir N Markovic
- Department of oncology and Department of Immunology, Mayo Clinic, (MN), Rochester, USA
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, Roma, Italy
| |
Collapse
|
10
|
Masterson CH, Curley GF, Laffey JG. Modulating the distribution and fate of exogenously delivered MSCs to enhance therapeutic potential: knowns and unknowns. Intensive Care Med Exp 2019; 7:41. [PMID: 31346794 PMCID: PMC6658643 DOI: 10.1186/s40635-019-0235-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/07/2019] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are undergoing intensive translational research for several debilitating conditions, including critical illnesses such as ARDS and sepsis. MSCs exert diverse biologic effects via their interaction with host tissues, via mechanisms that require the MSC to be in close proximity to the area of injury. Fully harnessing the therapeutic potential of advanced medicinal therapeutic products such as MSCs and their successful translation to clinical use requires a detailed understanding of MSC distribution and persistence in the injured tissues. Key aspects include understanding MSC distribution within the body, the response of the host to MSC administration, and the ultimate fate of exogenously administered MSCs within the host. Factors affecting this interaction include the MSC tissue source, the in vitro MSC culture conditions, the route of MSC administration and the specific issues relating to the target disease state, each of which remains to be fully characterised. Understanding these factors may generate strategies to modify MSC distribution and fate that may enhance their therapeutic effect. This review will examine our understanding of the mechanisms of action of MSCs, the early and late phase distribution kinetics of MSCs following in vivo administration, the ultimate fate of MSCs following administration and the potential importance of these MSC properties to their therapeutic effects. We will critique current cellular imaging and tracking methodologies used to track exogenous MSCs and their suitability for use in patients, discuss the insights they provide into the distribution and fate of MSCs after administration, and suggest strategies by which MSC biodistribution and fate may be modulated for therapeutic effect and clinical use. In conclusion, a better understanding of patterns of biodistribution and of the fate of MSCs will add important additional safety data regarding MSCs, address regulatory requirements, and may uncover strategies to increase the distribution and/or persistence of MSC at the sites of injury, potentially increasing their therapeutic potential for multiple disorders.
Collapse
Affiliation(s)
- Claire H Masterson
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Gerard F Curley
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland Education and Research Centre Smurfit Building, Beaumont Hospital, Dublin, 9, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland. .,School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland. .,Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Galway, Ireland.
| |
Collapse
|
11
|
Li X, Hacker M. Molecular imaging in stem cell-based therapies of cardiac diseases. Adv Drug Deliv Rev 2017; 120:71-88. [PMID: 28734900 DOI: 10.1016/j.addr.2017.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/06/2017] [Accepted: 07/16/2017] [Indexed: 12/26/2022]
Abstract
In the past 15years, despite that regenerative medicine has shown great potential for cardiovascular diseases, the outcome and safety of stem cell transplantation has shown controversial results in the published literature. Medical imaging might be useful for monitoring and quantifying transplanted cells within the heart and to serially characterize the effects of stem cell therapy of the myocardium. From the multiple available noninvasive imaging techniques, magnetic resonance imaging and nuclear imaging by positron (PET) or single photon emission computer tomography (SPECT) are the most used clinical approaches to follow the fate of transplanted stem cells in vivo. In this article, we provide a review on the role of different noninvasive imaging modalities and discuss their advantages and disadvantages. We focus on the different in-vivo labeling and reporter gene imaging strategies for stem cell tracking as well as the concept and reliability to use imaging parameters as noninvasive surrogate endpoints for the evaluation of the post-therapeutic outcome.
Collapse
Affiliation(s)
- Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria.
| |
Collapse
|
12
|
Potential Therapeutic Mechanisms and Tracking of Transplanted Stem Cells: Implications for Stroke Treatment. Stem Cells Int 2017; 2017:2707082. [PMID: 28904531 PMCID: PMC5585684 DOI: 10.1155/2017/2707082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/08/2017] [Accepted: 07/30/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy is a promising potential therapeutic strategy to treat cerebral ischemia in preclinical and clinical trials. Currently proposed treatments for stroke employing stem cells include the replacement of lost neurons and integration into the existing host circuitry, the release of growth factors to support and promote endogenous repair processes, and the secretion of extracellular vesicles containing proteins, noncoding RNA, or DNA to regulate gene expression in recipient cells and achieve immunomodulation. Progress has been made to elucidate the precise mechanisms underlying stem cell therapy and the homing, migration, distribution, and differentiation of transplanted stem cells in vivo using various imaging modalities. Noninvasive and safe tracer agents with high sensitivity and image resolution must be combined with long-term monitoring using imaging technology to determine the optimal therapy for stroke in terms of administration route, dosage, and timing. This review discusses potential therapeutic mechanisms of stem cell transplantation for the treatment of stroke and the limitations of current therapies. Methods to label transplanted cells and existing imaging systems for stem cell labeling and in vivo tracking will also be discussed.
Collapse
|
13
|
Gervois P, Wolfs E, Ratajczak J, Dillen Y, Vangansewinkel T, Hilkens P, Bronckaers A, Lambrichts I, Struys T. Stem Cell-Based Therapies for Ischemic Stroke: Preclinical Results and the Potential of Imaging-Assisted Evaluation of Donor Cell Fate and Mechanisms of Brain Regeneration. Med Res Rev 2016; 36:1080-1126. [DOI: 10.1002/med.21400] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Pascal Gervois
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Esther Wolfs
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Jessica Ratajczak
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Yörg Dillen
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tim Vangansewinkel
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Petra Hilkens
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Annelies Bronckaers
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Ivo Lambrichts
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tom Struys
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| |
Collapse
|
14
|
Lezaic L, Socan A, Peitl PK, Poglajen G, Sever M, Cukjati M, Cernelc P, Vrtovec B. Imaging and 1-day kinetics of intracoronary stem cell transplantation in patients with idiopathic dilated cardiomyopathy. Nucl Med Biol 2016; 43:410-4. [PMID: 27179747 DOI: 10.1016/j.nucmedbio.2015.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/18/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Stem cell transplantation is an emerging method of treatment for patients with cardiovascular disease. There are few studies completed or ongoing on stem cell therapy in patients with idiopathic dilated cardiomyopathy (IDCM). Information on stem cell homing and distribution in the myocardium after transplantation might provide important insight into effectiveness of transplantation procedure. AIM To assess early engraftment, retention and migration of intracoronarily transplanted stem cells in the myocardium of patients with advanced dilated cardiomyopathy of non-ischaemic origin using stem cell labeling with (99m)Tc-exametazime (HMPAO). MATERIALS, METHODS Thirty-five patients with IDCM and advanced heart failure were included in the study. Autologous hematopoietic (CD34+) stem cells were harvested by peripheral blood apheresis after bone marrow stimulation, labeled with (99m)Tc-HMPAO, tested for viability and injected into coronary vessel supplying areas of myocardium selected by myocardial perfusion scintigraphy as dysfunctional yet viable. Imaging was performed 1h and 18h after transplantation. RESULTS Myocardial stem cell retention ranged from 0 to 1.44% on early and 0-0.97% on delayed imaging. Significant efflux of stem cells occurred from site of delivery in this time period (p<0.001). Stem cell viability was not affected by labeling. CONCLUSION Stem cell labeling with (99m)Tc-HMPAO is a feasible method for stem cell tracking after transplantation in patients with IDCM.
Collapse
Affiliation(s)
- Luka Lezaic
- Department for Nuclear Medicine, UMC Ljubljana, Slovenia.
| | - Aljaz Socan
- Department for Nuclear Medicine, UMC Ljubljana, Slovenia
| | | | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia
| | - Matjaz Sever
- Department of Hematology, UMC Ljubljana, Slovenia
| | - Marko Cukjati
- National Blood Transfusion Institute, Ljubljana, Slovenia
| | | | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia; Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Comparison of three remote radiolabelling methods for long-circulating liposomes. J Control Release 2015; 220:239-244. [DOI: 10.1016/j.jconrel.2015.10.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/23/2015] [Accepted: 10/24/2015] [Indexed: 01/03/2023]
|
16
|
Lost signature: progress and failures in in vivo tracking of implanted stem cells. Appl Microbiol Biotechnol 2015; 99:9907-22. [DOI: 10.1007/s00253-015-6965-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/25/2015] [Accepted: 08/27/2015] [Indexed: 01/01/2023]
|
17
|
Keith MCL, Tokita Y, Tang XL, Ghafghazi S, Moore JB, Hong KU, Elmore JB, Amraotkar AR, Guo H, Ganzel BL, Grubb KJ, Flaherty MP, Vajravelu BN, Wysoczynski M, Bolli R. Effect of the stop-flow technique on cardiac retention of c-kit positive human cardiac stem cells after intracoronary infusion in a porcine model of chronic ischemic cardiomyopathy. Basic Res Cardiol 2015; 110:503. [PMID: 26150250 DOI: 10.1007/s00395-015-0503-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 01/03/2023]
Abstract
It is commonly thought that the optimal method for intracoronary administration of cells is to stop coronary flow during cell infusion, in order to prolong cell/vascular wall contact, enhance adhesion, and promote extravasation of cells into the interstitial space. However, occlusion of a coronary artery with a balloon involves serious risks of vascular damage and/or dissection, particularly in non-stented segments such as those commonly found in patients with heart failure. It remains unknown whether the use of the stop-flow technique results in improved donor cell retention. Acute myocardial infarction was produced in 14 pigs. One to two months later, pigs received 10 million indium-111 oxyquinoline (oxine)-labeled c-kit(pos) human cardiac stem cells (hCSCs) via intracoronary infusion with (n = 7) or without (n = 7) balloon inflation. Pigs received cyclosporine to prevent acute graft rejection. Animals were euthanized 24 h later and hearts harvested for radioactivity measurements. With the stop-flow technique, the retention of hCSCs at 24 h was 5.41 ± 0.80 % of the injected dose (n = 7), compared with 4.87 ± 0.62 % without coronary occlusion (n = 7), (P = 0.60). When cells are delivered intracoronarily in a clinically relevant porcine model of chronic ischemic cardiomyopathy, the use of the stop-flow technique does not result in greater myocardial cell retention at 24 h compared with non-occlusive infusion. These results have practical implications for the design of cell therapy trials. Our observations suggest that the increased risk of complications secondary to coronary manipulation and occlusion is not warranted.
Collapse
Affiliation(s)
- Matthew C L Keith
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gavins FNE, Smith HK. Cell tracking technologies for acute ischemic brain injury. J Cereb Blood Flow Metab 2015; 35:1090-9. [PMID: 25966948 PMCID: PMC4640284 DOI: 10.1038/jcbfm.2015.93] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/07/2015] [Accepted: 04/09/2015] [Indexed: 02/06/2023]
Abstract
Stem cell therapy has showed considerable potential in the treatment of stroke over the last decade. In order that these therapies may be optimized, the relative benefits of growth factor release, immunomodulation, and direct tissue replacement by therapeutic stem cells are widely under investigation. Fundamental to the progress of this research are effective imaging techniques that enable cell tracking in vivo. Direct analysis of the benefit of cell therapy includes the study of cell migration, localization, division and/or differentiation, and survival. This review explores the various imaging tools currently used in clinics and laboratories, addressing image resolution, long-term cell monitoring, imaging agents/isotopes, as well as safety and costs associated with each technique. Finally, burgeoning tracking techniques are discussed, with emphasis on multimodal imaging.
Collapse
Affiliation(s)
- Felicity NE Gavins
- Molecular and Cellular Physiology Department, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Helen K Smith
- Molecular and Cellular Physiology Department, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| |
Collapse
|
19
|
Badar A, Kiru L, Kalber TL, Jathoul A, Straathof K, Årstad E, Lythgoe MF, Pule M. Fluorescence-guided development of a tricistronic vector encoding bimodal optical and nuclear genetic reporters for in vivo cellular imaging. EJNMMI Res 2015; 5:18. [PMID: 25853023 PMCID: PMC4385325 DOI: 10.1186/s13550-015-0097-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/10/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In vivo imaging using genetic reporters is a central supporting tool in the development of cell and gene therapies affording us the ability to selectively track the therapeutic indefinitely. Previous studies have demonstrated the utility of the human norepinephrine transporter (hNET) as a positron emission tomography/single photon emission computed tomography (PET/SPECT) genetic reporter for in vivo cellular imaging. Here, our aim was to extend on this work and construct a tricistronic vector with dual optical (firefly luciferase) and nuclear (hNET) in vivo imaging and ex vivo histochemical capabilities. Guiding this development, we describe how a fluorescent substrate for hNET, 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP(+)), can be used to optimise vector design and serve as an in vitro functional screen. METHODS Vectors were designed to co-express a bright red-shifted firefly luciferase (FLuc), hNET and a small marker gene RQR8. Genes were co-expressed using 2A peptide linkage, and vectors were transduced into a T cell line, SupT1. Two vectors were constructed with different gene orientations; FLuc.2A.RQR8.2A.hNET and hNET.2A.FLuc.2A.RQR8. hNET function was assessed using ASP(+)-guided flow cytometry. In vivo cellular conspicuity was confirmed using sequential bioluminescence imaging (BLI) and SPECT imaging of transduced SupT1 cells injected into the flanks of mice. RESULTS SupT1/FLuc.2A.RQR8.2A.hNET cells resulted in >4-fold higher ASP(+) uptake compared to SupT1/hNET.2A.FLuc.2A.RQR8, suggesting that 2A orientation effected hNET function. SupT1/FLuc.2A.RQR8.2A.hNET cells were readily visualised with both BLI and SPECT, demonstrating high signal to noise at 24 h post (123)I-meta-iodobenzylguanidine (MIBG) administration. CONCLUSIONS In this study, a pre-clinical tricistronic vector with flow cytometry, BLI, SPECT and histochemical capabilities was constructed, which can be widely applied in cell tracking studies supporting the development of cell therapies. The study further demonstrates that hNET function in engineered cells can be assessed using ASP(+)-guided flow cytometry in place of costly radiosubstrate methodologies. This fluorogenic approach is unique to the hNET PET/SPECT reporter and may prove valuable when screening large numbers of cell lines or vector/mutant constructs.
Collapse
Affiliation(s)
- Adam Badar
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Louise Kiru
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Tammy L Kalber
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Amit Jathoul
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Karin Straathof
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Erik Årstad
- />Department of Chemistry and Institute of Nuclear Medicine, University College London, 235 Euston Road (T-5), London, NW1 2BU UK
| | - Mark F Lythgoe
- />Division of Medicine, Centre for Advanced Biomedical Imaging (CABI), University College London, 72 Huntley Street, London, WC1E 6DD UK
| | - Martin Pule
- />UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD UK
| |
Collapse
|
20
|
Cussó L, Mirones I, Peña-Zalbidea S, García-Vázquez V, García-Castro J, Desco M. Combination of single-photon emission computed tomography and magnetic resonance imaging to track 111in-oxine-labeled human mesenchymal stem cells in neuroblastoma-bearing mice. Mol Imaging 2015; 13. [PMID: 25248941 DOI: 10.2310/7290.2014.00033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Homing is an inherent, complex, multistep process performed by cells such as human bone marrow mesenchymal stem cells (hMSCs) to travel from a distant location to inflamed or damaged tissue and tumors. This ability of hMSCs has been exploited as a tumor-targeting strategy in cell-based cancer therapy. The purpose of this study was to investigate the applicability of 111In-oxine for tracking hMSCs in vivo by combining single-photon emission computed tomography (SPECT) and magnetic resonance imaging (MRI). 111In-labeled hMSCs (106 cells) were infused intraperitoneally in neuroblastoma-bearing mice, whereas a control group received a dose of free 111In-oxine. SPECT and MRI studies were performed 24 and 48 hours afterwards. Initially, the images showed similar activity in the abdomen in both controls and hMSC-injected animals. In general, abdominal activity decreases at 48 hours. hMSC-injected animals showed increased uptake in the tumor area at 48 hours, whereas the control group showed a low level of activity at 24 hours, which decreased at 48 hours. In conclusion, tracking 111In-labeled hMSCs combining SPECT and MRI is feasible and may be transferable to clinical research. The multimodal combination is essential to ensure appropriate interpretation of the images.
Collapse
|
21
|
|
22
|
Lezaic L, Haddad F, Vrtovec B, Wu JC. Imaging cardiac stem cell transplantation using radionuclide labeling techniques: clinical applications and future directions. Methodist Debakey Cardiovasc J 2014; 9:218-22. [PMID: 24298314 DOI: 10.14797/mdcj-9-4-218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stem cell therapy is emerging as a potential new therapy for patients with advanced heart failure. In recent years, advances in molecular imaging have allowed monitoring of stem cell homing and survival. In this review article, we will discuss the clinical application and future directions of stem cell imaging in advanced heart failure.
Collapse
Affiliation(s)
- Luka Lezaic
- University Medical Centre Ljubljana, Slovenia
| | | | | | | |
Collapse
|
23
|
Moudgil R, Dick AJ. Regenerative Cell Imaging in Cardiac Repair. Can J Cardiol 2014; 30:1323-34. [DOI: 10.1016/j.cjca.2014.08.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/29/2014] [Accepted: 08/29/2014] [Indexed: 01/03/2023] Open
|
24
|
In vivo bio-distribution and homing of endothelial outgrowth cells in a tumour model. Nucl Med Biol 2014; 41:848-55. [DOI: 10.1016/j.nucmedbio.2014.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 05/28/2014] [Accepted: 07/14/2014] [Indexed: 01/16/2023]
|
25
|
Jadczyk T, Faulkner A, Madeddu P. Stem cell therapy for cardiovascular disease: the demise of alchemy and rise of pharmacology. Br J Pharmacol 2014; 169:247-68. [PMID: 22712727 DOI: 10.1111/j.1476-5381.2012.01965.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Regenerative medicine holds great promise as a way of addressing the limitations of current treatments of ischaemic disease. In preclinical models, transplantation of different types of stem cells or progenitor cells results in improved recovery from ischaemia. Furthermore, experimental studies indicate that cell therapy influences a spectrum of processes, including neovascularization and cardiomyogenesis as well as inflammation, apoptosis and interstitial fibrosis. Thus, distinct strategies might be required for specific regenerative needs. Nonetheless, clinical studies have so far investigated a relatively small number of options, focusing mainly on the use of bone marrow-derived cells. Rapid clinical translation resulted in a number of small clinical trials that do not have sufficient power to address the therapeutic potential of the new approach. Moreover, full exploitation has been hindered so far by the absence of a solid theoretical framework and inadequate development plans. This article reviews the current knowledge on cell therapy and proposes a model theory for interpretation of experimental and clinical outcomes from a pharmacological perspective. Eventually, with an increased association between cell therapy and traditional pharmacotherapy, we will soon need to adopt a unified theory for understanding how the two practices additively interact for a patient's benefit.
Collapse
Affiliation(s)
- T Jadczyk
- Third Division of Cardiology, Medical University of Silesia, Katovice, Poland
| | | | | |
Collapse
|
26
|
Radiopharmaceutical stem cell tracking for neurological diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:417091. [PMID: 24982880 PMCID: PMC4055613 DOI: 10.1155/2014/417091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 05/03/2014] [Indexed: 01/27/2023]
Abstract
Although neurological ailments continue to be some of the main causes of disease burden in the world, current therapies such as pharmacological agents have limited potential in the restoration of neural functions. Cell therapies, firstly applied to treat different hematological diseases, are now being investigated in preclinical and clinical studies for neurological illnesses. However, the potential applications and mechanisms for such treatments are still poorly comprehended and are the focus of permanent research. In this setting, noninvasive in vivo imaging allows better understanding of several aspects of stem cell therapies. Amongst the various methods available, radioisotope cell labeling has become one of the most promising since it permits tracking of cells after injection by different routes to investigate their biodistribution. A significant increase in the number of studies utilizing this method has occurred in the last years. Here, we review the different radiopharmaceuticals, imaging techniques, and findings of the preclinical and clinical reports published up to now. Moreover, we discuss the limitations and future applications of radioisotope cell labeling in the field of cell transplantation for neurological diseases.
Collapse
|
27
|
Katsikis A, Koutelou M. Cardiac Stem Cell Imaging by SPECT and PET. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9265-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Napp LC, Templin C. What You See is What You Get? Imaging of Cell Therapy for Cardiac Regeneration. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-013-9243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
29
|
Srinivas M, Melero I, Kaempgen E, Figdor CG, de Vries IJM. Cell tracking using multimodal imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2013; 8:432-8. [DOI: 10.1002/cmmi.1561] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 07/04/2013] [Accepted: 07/07/2013] [Indexed: 12/24/2022]
Affiliation(s)
- Mangala Srinivas
- Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre; Department of Tumor Immunology; Nijmegen The Netherlands
| | - Ignacio Melero
- University of Navarra; CIMA, CUV and Medica School; Pamplona Spain
| | - Eckhart Kaempgen
- University Hospital Erlangen; Dept. of Dermatology; Erlangen Germany
| | - Carl G. Figdor
- Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre; Department of Tumor Immunology; Nijmegen The Netherlands
| | - I. Jolanda M. de Vries
- Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre; Department of Tumor Immunology; Nijmegen The Netherlands
| |
Collapse
|
30
|
Mathiasen AB, Kastrup J. Non-invasive in-vivo imaging of stem cells after transplantation in cardiovascular tissue. Am J Cancer Res 2013; 3:561-72. [PMID: 23946822 PMCID: PMC3741605 DOI: 10.7150/thno.5787] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 03/03/2013] [Indexed: 12/11/2022] Open
Abstract
Stem cell therapy for degenerative diseases, including ischemic heart disease is now a clinical reality. In the search for the optimal cell type for each patient category, many different stem cell subpopulations have been used. In addition, different cell processing procedures and delivery methods have been utilized. Moreover, choices of endpoints have varied between studies. Diverging results have been reported from clinical experiences, with some studies demonstrating promising results with improved cardiac function and reduced mortality and clinical symptoms, and others have seen no improvements. To better understand the underlying mechanisms of these results, a reverse translation from bedside to bench has been opened. Non-invasive cell tracking after implantation has a pivotal role in this translation. Imaging based methods can help elucidate important issues such as retention, migration and efficacy of the transplanted cells. Great effort is being made in finding new and better imaging techniques for different imaging modalities, and much have already been learned. But there are still many unanswered questions. In this review, we give an overview of the imaging modalities used for cell tracking and summarize the latest advances within the field.
Collapse
|
31
|
Hauzeur JP, Bernard C, Egrise D, Kurth W, Van Cauwenberge H, Lechanteur C, Gillet P, Beguin Y, Malaise M, Hustinx R. (111)Indium-oxine labelling for evaluating the homing process of autologous osteoblasts implanted percutaneously in atrophic nonunion fractures. INTERNATIONAL ORTHOPAEDICS 2012. [PMID: 23180104 DOI: 10.1007/s00264-012-1719-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE The aim of the study was to control the in vivo localisation of implanted cells in cell-based therapies. Labelling cells with (111)indium-oxine is one of the most interesting methods proposed. We evaluated this method in the setting of autologous osteoblast implantation in nonunion fractures. METHODS An in vitro study of osteoblasts was conducted after (111)indium-oxine labelling. Radioactivity retention and viability, proliferation and the ability to produce alkaline phosphatase were evaluated in a seven-day culture. In vivo labelling of implanted osteoblastic cells was conducted during a therapeutic trial of atrophic nonunion fractures, with the leakage outside the nonunion site and local uptake evolution at four, 24 and 48 hour being studied. RESULTS The mean labelling efficiency for osteoprogenitors was 78.8 ± 4.6 %. The intracellular retention was 89.4 ± 2.1 % at three hours and 67.3 ± 4.7 % at 18 hours. The viability assessed at three hours was 93.7 ± 0.6 %. After seven days of culture, morphology and alkaline phosphatase staining were similar for both labelled and unlabelled control cells, although the proliferation rate was decreased in the labelled cells. Some local intraosseous leakage was observed in four of 17 cases. All patients showed uptake at the injection site, with four having no other uptake. Four patients showed additional uptake in the bladder, liver and spleen, while 11 patients had additional uptake in the lungs in addition to the bladder, liver and spleen. The activity ratios (injection site/body) were 48 ± 28 % at four hours, 40 ± 25 % at 24 hours and 35 ± 25 % at 48 hours. After correcting for decay, the activity within the injection site was 82 ± 15 % at 24 hours and 69 ± 11 % at 48 hours compared with the activity measured at four hours. No relationship was found between uptake and radiological bone repair. CONCLUSIONS The (111)indium-oxine labelling appears to be a good method for monitoring the behaviour of the osteoblastic cells after their implantation in atrophic nonunion fractures.
Collapse
|
32
|
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
|
33
|
Imaging of Cells and Nanoparticles: Implications for Drug Delivery to the Brain. Pharm Res 2012; 29:3213-34. [DOI: 10.1007/s11095-012-0826-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/05/2012] [Indexed: 01/03/2023]
|
34
|
Umbilical cord blood mononuclear cell transplantation for neonatal hypoxic-ischemic encephalopathy. Pediatr Res 2012; 71:464-73. [PMID: 22430382 DOI: 10.1038/pr.2011.59] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite recent advances in the treatment of neonatal hypoxic-ischemic encephalopathy (HIE) using therapeutic hypothermia, at least 30% of the cooled infants will die or have moderate/severe neurological disability. Umbilical cord blood cells (UCBCs), which are readily available at birth, have been shown to reduce sensorimotor and/or cognitive impairments in several models of brain damage, representing a promising option for the treatment of neurological diseases. In this review, we discuss recent preclinical studies that assessed the effects of UCBC transplantation in the Rice-Vannucci animal model of HIE. We also review the possible cell types and mechanisms involved in the therapeutic effect of UCBC transplantation, including neuroprotection, immunomodulation, and stimulation of neural plasticity and regeneration. In addition, we discuss how neuroimaging methods, such as bioluminescence imaging, nuclear-medicine imaging, or magnetic resonance imaging, could be used to evaluate the biodistribution of UCBCs in both preclinical and clinical studies.
Collapse
|
35
|
Gildehaus FJ, Haasters F, Drosse I, Wagner E, Zach C, Mutschler W, Cumming P, Bartenstein P, Schieker M. Impact of indium-111 oxine labelling on viability of human mesenchymal stem cells in vitro, and 3D cell-tracking using SPECT/CT in vivo. Mol Imaging Biol 2012; 13:1204-14. [PMID: 21080231 DOI: 10.1007/s11307-010-0439-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE This study investigates the effects of (111)In-oxine incorporation on human mesenchymal stem cells' (hMSC) biology and viability, and the applicability of (111)In-oxine for single-photon emission computed tomography/X-ray computed tomography (SPECT/CT) monitoring of hMSC in vivo. PROCEDURES HMSC were labelled with 10 Bq/cell. Cellular retention of radioactivity, cell survival, and migration were evaluated over 48 h. Metabolic activity was assessed over 14 days and the hMSC's stem cell character was evaluated. Serial SPECT/CT was performed after intra-osseous injection to athymic rats over 48 h. RESULTS Labelling efficiency was 25%, with 61% of incorporated (111)In remaining in the hMSC at 48 h. The radiolabelling was without effect on cell viability, stem cell character, and plasticity, whereas metabolic activity and migration were significantly reduced. Grafted cells could be imaged in situ with SPECT/CT. CONCLUSIONS (111)In-oxine labelling moderately impaired hMSC's functional integrity while preserving their stem cell character. Combined SPECT/CT imaging of (111)In-oxine-labelled hMSC opens the possibility for non-invasive sequential monitoring of therapeutic stem cells.
Collapse
|
36
|
IL-2 stimulated but not unstimulated NK cells induce selective disappearance of peripheral blood cells: concomitant results to a phase I/II study. PLoS One 2011; 6:e27351. [PMID: 22096557 PMCID: PMC3212563 DOI: 10.1371/journal.pone.0027351] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 10/14/2011] [Indexed: 01/11/2023] Open
Abstract
In an ongoing clinical phase I/II study, 16 pediatric patients suffering from high risk leukemia/tumors received highly purified donor natural killer (NK) cell immunotherapy (NK-DLI) at day (+3) +40 and +100 post haploidentical stem cell transplantation. However, literature about the influence of NK-DLI on recipient's immune system is scarce. Here we present concomitant results of a noninvasive in vivo monitoring approach of recipient's peripheral blood (PB) cells after transfer of either unstimulated (NK-DLI(unstim)) or IL-2 (1000 U/ml, 9-14 days) activated NK cells (NK-DLI(IL-2 stim)) along with their ex vivo secreted cytokine/chemokines. We performed phenotypical and functional characterizations of the NK-DLIs, detailed flow cytometric analyses of various PB cells and comprehensive cytokine/chemokine arrays before and after NK-DLI. Patients of both groups were comparable with regard to remission status, immune reconstitution, donor chimerism, KIR mismatching, stem cell and NK-DLI dose. Only after NK-DLI(IL-2 stim) was a rapid, almost complete loss of CD56(bright)CD16(dim/-) immune regulatory and CD56(dim)CD16(+) cytotoxic NK cells, monocytes, dendritic cells and eosinophils from PB circulation seen 10 min after infusion, while neutrophils significantly increased. The reduction of NK cells was due to both, a decrease in patients' own CD69(-) NCR(low)CD62L(+) NK cells as well as to a diminishing of the transferred cells from the NK-DLI(IL-2 stim) with the CD56(bright)CD16(+/-)CD69(+)NCR(high)CD62L(-) phenotype. All cell counts recovered within the next 24 h. Transfer of NK-DLI(IL-2 stim) translated into significantly increased levels of various cytokines/chemokines (i.e. IFN-γ, IL-6, MIP-1β) in patients' PB. Those remained stable for at least 1 h, presumably leading to endothelial activation, leukocyte adhesion and/or extravasation. In contrast, NK-DLI(unstim) did not cause any of the observed effects. In conclusion, we assume that the adoptive transfer of NK-DLI(IL-2 stim) under the influence of ex vivo and in vivo secreted cytokines/chemokines may promote NK cell trafficking and therefore might enhance efficacy of immunotherapy.
Collapse
|
37
|
In vivo tracking of transplanted mononuclear cells using manganese-enhanced magnetic resonance imaging (MEMRI). PLoS One 2011; 6:e25487. [PMID: 22003393 PMCID: PMC3189206 DOI: 10.1371/journal.pone.0025487] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 09/06/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Transplantation of mononuclear cells (MNCs) has previously been tested as a method to induce therapeutic angiogenesis to treat limb ischemia in clinical trials. Non-invasive high resolution imaging is required to track the cells and evaluate clinical relevance after cell transplantation. The hypothesis that MRI can provide in vivo detection and long-term observation of MNCs labeled with manganese contrast-agent was investigated in ischemic rat legs. METHODS AND FINDINGS The Mn-labeled MNCs were evaluated using 7-tesla high-field magnetic resonance imaging (MRI). Intramuscular transplanted Mn-labeled MNCs were visualized with MRI for at least 7 and up to 21 days after transplantation in the ischemic leg. The distribution of Mn-labeled MNCs was similar to that of ¹¹¹In-labeled MNCs measured with single-photon emission computed tomography (SPECT) and DiI-dyed MNCs with fluorescence microscopy. In addition, at 1-2 days after transplantation the volume of the site injected with intact Mn-labeled MNCs was significantly larger than that injected with dead MNCs, although the dead Mn-labeled MNCs were also found for approximately 2 weeks in the ischemic legs. The area covered by CD31-positive cells (as a marker of capillary endothelial cells) in the intact Mn-MNCs implanted site at 43 days was significantly larger than that at a site implanted with dead Mn-MNCs. CONCLUSIONS The present Mn-enhanced MRI method enabled visualization of the transplanted area with a 150-175 µm in-plane spatial resolution and allowed the migration of labeled-MNCs to be observed for long periods in the same subject. After further optimization, MRI-based Mn-enhanced cell-tracking could be a useful technique for evaluation of cell therapy both in research and clinical applications.
Collapse
|
38
|
Psaltis PJ, Simari RD, Rodriguez-Porcel M. Emerging roles for integrated imaging modalities in cardiovascular cell-based therapeutics: a clinical perspective. Eur J Nucl Med Mol Imaging 2011; 39:165-81. [PMID: 21901381 DOI: 10.1007/s00259-011-1925-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/18/2011] [Indexed: 12/20/2022]
Abstract
Despite preclinical promise, the progress of cell-based therapy to clinical cardiovascular practice has been slowed by several challenges and uncertainties that have been highlighted by the conflicting results of human trials. Most telling has been the revelation that current strategies fall short of achieving sufficient retention and engraftment of cells to meet the ambitious objective of myocardial regeneration. This has sparked novel research into the refinement of cell biology and delivery to overcome these shortcomings. Within this context, molecular imaging has emerged as a valuable tool for providing noninvasive surveillance of cell fate in vivo. Direct and indirect labelling of cells can be coupled with clinically relevant imaging modalities, such as radionuclide single photon emission computed tomography and positron emission tomography, and magnetic resonance imaging, to assess their short- and long-term distributions, along with their viability, proliferation and functional interaction with the host myocardium. This review details the strengths and limitations of the different cell labelling and imaging techniques and their potential application to the clinical realm. We also consider the broader, multifaceted utility of imaging throughout the cell therapy process, providing a discussion of its considerable value during cell delivery and its importance during the evaluation of cardiac outcomes in clinical studies.
Collapse
Affiliation(s)
- Peter J Psaltis
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
39
|
Cell tracking in cardiac repair: what to image and how to image. Eur Radiol 2011; 22:189-204. [PMID: 21735069 PMCID: PMC3229694 DOI: 10.1007/s00330-011-2190-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 04/21/2011] [Accepted: 05/09/2011] [Indexed: 01/01/2023]
Abstract
Stem cell therapies hold the great promise and interest for cardiac regeneration among scientists, clinicians and patients. However, advancement and distillation of a standard treatment regimen are not yet finalised. Into this breach step recent developments in the imaging biosciences. Thus far, these technical and protocol refinements have played a critical role not only in the evaluation of the recovery of cardiac function but also in providing important insights into the mechanism of action of stem cells. Molecular imaging, in its many forms, has rapidly become a necessary tool for the validation and optimisation of stem cell engrafting strategies in preclinical studies. These include a suite of radionuclide, magnetic resonance and optical imaging strategies to evaluate non-invasively the fate of transplanted cells. In this review, we highlight the state-of-the-art of the various imaging techniques for cardiac stem cell presenting the strengths and limitations of each approach, with a particular focus on clinical applicability.
Collapse
|
40
|
Welling MM, Duijvestein M, Signore A, van der Weerd L. In vivo biodistribution of stem cells using molecular nuclear medicine imaging. J Cell Physiol 2011; 226:1444-52. [PMID: 21413018 DOI: 10.1002/jcp.22539] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studies on stem cell are rapidly developing since these cells have great therapeutic potential for numerous diseases and has generated much promise as well as confusion due to contradictory results. Major questions in this research field have been raised as to how and in which numbers stem cells home to target tissues after administration, whether the cells engraft and differentiate, and what their long-term fate is. To answer these questions, reliable in vivo tracking techniques are essential. In vivo molecular imaging techniques using magnetic resonance imaging, bioluminescence, and scintigraphy have been applied for this purpose in experimental studies. The aim of this review is to discuss various radiolabeling techniques for early stem cell tracking, the need for validation of viability and performance of the cells after labeling, and the routes of administration in experimental animal models. In addition, we evaluate current problems and directions related to stem cell tracking using radiolabels, including a possible role for their clinical implementation.
Collapse
Affiliation(s)
- Mick M Welling
- Department of Radiology, Molecular Imaging Laboratories Leiden, Section Nuclear Medicine, Leiden University Medical Center, Leiden, Netherlands.
| | | | | | | |
Collapse
|
41
|
Musialek P, Tekieli L, Kostkiewicz M, Majka M, Szot W, Walter Z, Zebzda A, Pieniazek P, Kadzielski A, Banys RP, Olszowska M, Pasowicz M, Zmudka K, Tracz W. Randomized transcoronary delivery of CD34(+) cells with perfusion versus stop-flow method in patients with recent myocardial infarction: Early cardiac retention of ⁹⁹(m)Tc-labeled cells activity. J Nucl Cardiol 2011; 18:104-16. [PMID: 21161463 PMCID: PMC3032199 DOI: 10.1007/s12350-010-9326-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 09/20/2010] [Indexed: 12/21/2022]
Abstract
BACKGROUND For transcoronary progenitor cells' administration, injections under flow arrest (over-the-wire balloon technique, OTW) are used universally despite lack of evidence for being required for cell delivery or being effective in stimulating myocardial engraftment. Flow-mediated endothelial rolling is mandatory for subsequent cell adhesion and extravasation. METHODS To optimize cell directing toward the coronary endothelium under maintained flow, the authors developed a cell-delivery side-holed perfusion catheter (PC). Thirty-four patients (36-69 years, 30 men) with primary stent-assisted angioplasty-treated anterior MI (peak TnI 151 [53-356]ng/dL, mean[range]) were randomly assigned to OTW or PC autologous ⁹⁹Tc-extametazime-labeled bone marrow CD34(+) cells (4.34 [0.92-7.54] × 10⁶) administration at 6-14 days after pPCI (LVEF 37.1 [24-44]%). Myocardial perfusion (⁹⁹(m)Tc-MIBI) and labeled cells' activity were evaluated (SPECT) at, respectively, 36-48 h prior to and 60 min after delivery. RESULTS In contrast to OTW coronary occlusions, no intolerance or ventricular arrhythmia occurred with PC cells' administration (P < .001). One hour after delivery, 4.86 [1.7-7.6]% and 5.05 [2.2-9.9]% activity was detected in the myocardium (OTW and PC, respectively, P = .84). Labeled cell activity was clearly limited to the (viable) peri-infarct zone in 88% patients, indicating that the infarct core zone may be largely inaccessible to transcoronary-administered cells. CONCLUSIONS Irrespective of the transcoronary delivery method, only ≈ 5% of native (i.e., non-engineered) CD34(+) cells spontaneously home to the injured myocardium, and cell retention occurs preferentially in the viable peri-infarct zone. Although the efficacy of cell delivery is not increased with the perfusion method, by avoiding provoking ischemic episodes PC offers a rational alternative to the OTW delivery.
Collapse
Affiliation(s)
- Piotr Musialek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Lukasz Tekieli
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Magdalena Kostkiewicz
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Marcin Majka
- Department of Transplantation, Jagiellonian University, Krakow, Poland
| | | | - Zbigniew Walter
- Department of Hematology, Jagiellonian University, Krakow, Poland
| | - Anna Zebzda
- Department of Transplantation, Jagiellonian University, Krakow, Poland
| | - Piotr Pieniazek
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | | | | | - Maria Olszowska
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | | | - Krzysztof Zmudka
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| | - Wieslawa Tracz
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Institute of Cardiology, Jagiellonian University, ul. Pradnicka 80, 31-202 Krakow, Poland
- John Paul II Hospital, Krakow, Poland
| |
Collapse
|
42
|
Battistella V, de Freitas GR, da Fonseca LMB, Mercante D, Gutfilen B, Goldenberg RCS, Vieira Dias J, Kasai-Brunswick TH, Wajnberg E, Rosado-de-Castro PH, Alves-Leon SV, Mendez-Otero R, Andre C. Safety of autologous bone marrow mononuclear cell transplantation in patients with nonacute ischemic stroke. Regen Med 2011; 6:45-52. [DOI: 10.2217/rme.10.97] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
43
|
Non-surgical stem cell delivery strategies and in vivo cell tracking to injured myocardium. Int J Cardiovasc Imaging 2010; 27:367-83. [PMID: 20577813 PMCID: PMC3092059 DOI: 10.1007/s10554-010-9658-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 06/11/2010] [Indexed: 01/10/2023]
Abstract
Heart failure is a major economic and public health problem. Despite the recent advances in drug therapy and coronary revascularization, the lost cardiomyocytes due to necrosis and apoptosis are not replaced by new myocardial tissue. Cell therapy is an interesting therapeutic option as it potentially improves contractility and restores regional ventricular function. Early clinical data demonstrated that cell transplantation, mainly delivered through non-surgical methods, is safe and feasible. However, several important issues need to be elucidated. This includes, next to determining the best cell type, the optimal delivery strategy, the biodistribution and the survival of implanted stem cells after transplantation. In this view, pre-clinical animal experiments are indispensable. Reporter genes, magnetic or radioactive labeling of stem cells have been developed to observe the fate and the distribution of transplanted cells using non-invasive imaging techniques. Several studies have demonstrated that these direct and non-direct labeling techniques may become an important tool in cell therapy. Integration of cell delivery and cell tracking will probably be a key for the success of cell therapy in patients. This review will provide a comprehensive overview on the various cell tracking and non-surgical cell delivery techniques, which are highly important in view of experimental and clinical studies.
Collapse
|
44
|
Gholamrezanezhad A, Mirpour S, Ardekani JM, Bagheri M, Alimoghadam K, Yarmand S, Malekzadeh R. Cytotoxicity of 111In-oxine on mesenchymal stem cells: a time-dependent adverse effect. Nucl Med Commun 2009; 30:210-6. [PMID: 19262283 DOI: 10.1097/mnm.0b013e328318b328] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Radioactive labeling with indium (In) tracers has been among the most widely used methods for tracking stem cells. As the first experiment on human stem cells, we designed a study to continuously follow the influence of In labeling on stem cell viability during the 2-week period of postlabeling. METHODS After culturing mesenchymal stem cells (MSCs), we divided the cells into six samples, each of which contained 1x10 MSCs. The first sample was considered as the control. The remaining five samples (samples 2-6) were labeled with the following doses of In-oxine, respectively: 0.76, 1.64, 3.48, 5.33, and 7.16 MBq/10 MSCs. To evaluate the effects of In-oxine labeling on cellular viability and count, all samples were examined immediately after labeling (2 h) as well as 24, 48 h, and 5, 7, and 14 days postlabeling. RESULTS No statistically significant relationship was found between labeling efficiency and administered dose. Associations between the specific activity and radiotracer dosage was significant (P=0.001, r=0.9). In addition, a negative correlation was noted between radiotracer dosage and viability during the 2-week period of follow-up. CONCLUSION Cytotoxic effects of In on human stem cells is a time-dependent phenomenon and hence, assessment of the stem cell viability immediately after labeling (which is frequently made in clinical trials) is unable to detect adverse effects of this radiopharmaceutical on the integrity of stem cells. Even low doses of In-oxine are accompanied by significant cell loss in a 2-week period. Although it has been confirmed that nuclear medicine techniques are the most sensitive methods for stem cell tracking, we recommend that the application of this tracking technique should be treated with great reserve, and if necessary, as little of In-oxine as possible should be added to the cells (or only a limited portion of the cells should be labeled) to minimize cell death.
Collapse
|
45
|
Abstract
Hematopoietic, stromal and organ-specific stem cells are under evaluation for therapeutic efficacy in cell-based therapies of cardiac, neurological and other disorders. It is critically important to track the location of directly transplanted or infused cells that can serve as gene carrier/delivery vehicles for the treatment of disease processes and be able to noninvasively monitor the temporal and spatial homing of these cells to target tissues. Moreover, it is also necessary to determine their engraftment efficiency and functional capability following transplantation. There are various in vivo imaging modalities used to track the movement and incorporation of administered cells. Tagging stem cells with different contrast agents can make these cells probes for different imaging modalities. Recent reports have shown that stem cells labeled with iron oxides can be used as cellular MRI probes demonstrating the cell trafficking to target tissues. In this review, we will discuss the status and future prospect of stem cell tracking by cellular MRI for cell-based therapy.
Collapse
Affiliation(s)
- Ali S Arbab
- Henry Ford Hospital, Cellular & Molecular Imaging Laboratory,Department of Radiology, 1 Ford Place, 2F Detroit, MI 48202, USA.
| | | |
Collapse
|
46
|
Bonios MJ, Terrovitis JV, Abraham MR. Stem cell: "KANT" you see it? J Cardiovasc Transl Res 2008; 1:103-5. [PMID: 20559903 DOI: 10.1007/s12265-008-9023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Accepted: 03/26/2008] [Indexed: 10/22/2022]
Affiliation(s)
- Michael J Bonios
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
47
|
Gautam UK, Fang X, Bando Y, Zhan J, Golberg D. Synthesis, structure, and multiply enhanced field-emission properties of branched ZnS nanotube-in nanowire core-shell heterostructures. ACS NANO 2008; 2:1015-1021. [PMID: 19206499 DOI: 10.1021/nn800013b] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
We report on the synthesis of a novel core-shell metal-semiconductor heterostructure where In forms the core nanowire and wurtzite ZnS forms the shell nanotube. In addition, controlled reaction conditions result in the growth of secondary quasi-aligned ZnS nanowires as numerous branches on the shell nanotubes. These hierarchical architectures are attractive for two reasons: (i) the sharp and quasi-aligned ZnS tips of the nanostructures are potential field-emitters and (ii) since In in bulk form is superconducting the synthesis of core In nanowires should now pave the way for further investigations on magnetic versus transport behavior in type-1 superconductors at the nanoscale. The synthesis could be achieved by employing a rapidly heating carbothermal chemical vapor deposition technique and a high reaction temperature. Transmission electron microscopy reveals that the core In nanowires are single crystals, whereas, within a hierarchical shell, the stem and the branches are separated with a crystalline interface. Field-emission measurements demonstrate remarkably large field enhancement which is explained on the basis of a sequential stepwise enhancement mechanism involving the consecutive stem and branch contributions. The present new nanoarchitectures are envisaged to be an important candidate for potential nanoelectronic devices.
Collapse
Affiliation(s)
- Ujjal K Gautam
- International Center for Materials Nanoarchitectonics (MANA), World Premier International Research Center, Japan.
| | | | | | | | | |
Collapse
|