101
|
Nyolczas N, Charwat S, Posa A, Hemetsberger R, Pavo N, Hemetsberger H, Pavo IJ, Glogar D, Maurer G, Gyöngyösi M. Tracking the migration of cardially delivered therapeutic stem cells in vivo: state of the art. Regen Med 2009; 4:407-22. [PMID: 19438316 DOI: 10.2217/rme.09.14] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Cell-based therapy is a promising, novel therapeutic strategy for cardiovascular disease. The rapid transition of this approach from the benchside to clinical trials has left a gap in the understanding of the mechanisms of cell therapy. Monitoring of cell homing and the fate of cardially delivered stem cells is fundamental for clarification of the myocardial regenerative process. Noninvasive imaging techniques allow an in vivo evaluation of the survival, migration and differentiation of implanted stem cells over time, and by this means, can help to answer unresolved questions. The most promising in vivo tracking methods involve the direct, nonspecific labeling of cells including MRI, radionuclide imaging and the use of reporter-gene imaging. This review summarizes the most important results of animal and human studies in which the fate and biodistribution of cardially delivered stem cells are assessed through different in vivo tracking methods.
Collapse
Affiliation(s)
- N Nyolczas
- Department of Cardiology, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Nikolic B, Faintuch S, Goldberg SN, Kuo MD, Cardella JF. Stem Cell Therapy: A Primer for Interventionalists and Imagers. J Vasc Interv Radiol 2009; 20:999-1012. [DOI: 10.1016/j.jvir.2009.04.075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 04/24/2009] [Accepted: 04/28/2009] [Indexed: 02/06/2023] Open
|
103
|
Blackwood KJ, Lewden B, Wells RG, Sykes J, Stodilka RZ, Wisenberg G, Prato FS. In vivo SPECT quantification of transplanted cell survival after engraftment using (111)In-tropolone in infarcted canine myocardium. J Nucl Med 2009; 50:927-35. [PMID: 19470852 DOI: 10.2967/jnumed.108.058966] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
UNLABELLED Current investigations of cell transplant therapies in damaged myocardium are limited by the inability to quantify cell transplant survival in vivo. We describe how the labeling of cells with (111)In can be used to monitor transplanted cell viability in a canine infarction model. METHODS We experimentally determined the contribution of the (111)In signal associated with transplanted cell (TC) death and radiolabel leakage to the measured SPECT signal when (111)In-labeled cells were transplanted into the myocardium. Three groups of experiments were performed in dogs. Radiolabel leakage was derived by labeling canine myocardium in situ with free (111)In-tropolone (n = 4). To understand the contribution of extracellular (111)In (e.g., after cell death), we developed a debris impulse response function (DIRF) by injecting lysed (111)In-labeled cells within reperfused (n = 3) and nonreperfused (n = 5) myocardial infarcts and within normal (n = 3) canine myocardium. To assess the application of the modeling derived from these experiments, (111)In-labeled cells were transplanted into infarcted myocardium (n = 4; 3.1 x 10(7) +/- 5.4 x 10(6) cells). Serial SPECT images were acquired after direct epicardial injection to determine the time-dependent radiolabel clearance. Clearance kinetics were used to correct for (111)In associated with viable TCs. RESULTS (111)In clearance followed a biphasic response and was modeled as a biexponential with a short (T(1/2)(s)) and long (T(1/2)(l)) biologic half-life. The T(1/2)(s) was not significantly different between experimental groups, suggesting that initial losses were due to transplantation methodology, whereas the T(1/2)(l) reflected the clearance of retained (111)In. DIRF had an average T(1/2)(l) of 19.4 +/- 4.1 h, and the T(1/2)(l) calculated from free (111)In-tropolone injected in situ was 882.7 +/- 242.8 h. The measured T(1/2)(l) for TCs was 74.3 h and was 71.2 h when corrections were applied. CONCLUSION A new quantitative method to assess TC survival in myocardium using SPECT and (111)In has been introduced. At the limits, method accuracy is improved if appropriate corrections are applied. In vivo (111)In imaging most accurately describes cell viability half-life if T(1/2)(l) is between 20 h and 37 d.
Collapse
|
104
|
Kuliszewski MA, Fujii H, Liao C, Smith AH, Xie A, Lindner JR, Leong-Poi H. Molecular imaging of endothelial progenitor cell engraftment using contrast-enhanced ultrasound and targeted microbubbles. Cardiovasc Res 2009; 83:653-62. [PMID: 19564152 DOI: 10.1093/cvr/cvp218] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Imaging methods to track the fate of progenitor cells after their delivery would be useful in assessing the efficacy of cell-based therapies. We hypothesized that contrast-enhanced ultrasound (CEU) using microbubbles targeted to a genetically engineered cell-surface marker on endothelial progenitor cells (EPCs) would allow the targeted imaging of vascular engraftment. METHODS AND RESULTS Rodent bone marrow-derived EPCs were isolated, cultured, and transfected to express the marker protein, H-2Kk, on the cell surface. Non-transfected EPCs and EPCs transfected with either null plasmid or Firefly luciferase served as controls. Control microbubbles (MB(C)) and microbubbles targeted to H-2Kk expressed on EPCs (MB(H-2Kk)) were constructed. Binding of targeted microbubbles to EPCs was assessed in vitro using a parallel plate flow chamber system. CEU imaging of EPC-targeted microbubbles was assessed in vivo using subcutaneously implanted EPC-supplemented Matrigel plugs in rats. In flow chamber experiments, there was minimal attachment of microbubbles to plated control EPCs. Although numbers of adhered MB(C) were also low, there was greater and more diffuse attachment of MB(H-2Kk) to plated H-2Kk-transfected EPCs. Targeted CEU demonstrated marked contrast enhancement at the periphery of the H-2Kk-transfected EPC-supplemented Matrigel plug for MB(H-2Kk,) whereas contrast enhancement was low for MB(C). Contrast enhancement was also low for both microbubbles within control mock-transfected EPC plugs. The signal intensity within the H-2Kk-transfected EPC plug was significantly greater for MB(H-2Kk) when compared with MB(C). CONCLUSION Microbubbles targeted to a genetically engineered cell-surface marker on EPCs exhibit specific binding to EPCs in vitro. These targeted microbubbles bind to engrafted EPCs in vivo within Matrigel plugs and can be detected by their enhancement on CEU imaging.
Collapse
Affiliation(s)
- Michael A Kuliszewski
- Division of Cardiology, Keenan Research Centre in the Li Ka Shing Knowledge Institute, 7-052 Bond Wing, St Michael's Hospital, 30 Bond Street, Toronto, Ontario, Canada M5B 1W8
| | | | | | | | | | | | | |
Collapse
|
105
|
Nakamuta JS, Danoviz ME, Marques FLN, dos Santos L, Becker C, Gonçalves GA, Vassallo PF, Schettert IT, Tucci PJF, Krieger JE. Cell therapy attenuates cardiac dysfunction post myocardial infarction: effect of timing, routes of injection and a fibrin scaffold. PLoS One 2009; 4:e6005. [PMID: 19547700 PMCID: PMC2695782 DOI: 10.1371/journal.pone.0006005] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Accepted: 05/06/2009] [Indexed: 01/09/2023] Open
Abstract
Background Cell therapy approaches for biologic cardiac repair hold great promises, although basic fundamental issues remain poorly understood. In the present study we examined the effects of timing and routes of administration of bone marrow cells (BMC) post-myocardial infarction (MI) and the efficacy of an injectable biopolymer scaffold to improve cardiac cell retention and function. Methodology/Principal Findings 99mTc-labeled BMC (6×106 cells) were injected by 4 different routes in adult rats: intravenous (IV), left ventricular cavity (LV), left ventricular cavity with temporal aorta occlusion (LV+) to mimic coronary injection, and intramyocardial (IM). The injections were performed 1, 2, 3, or 7 days post-MI and cell retention was estimated by γ-emission counting of the organs excised 24 hs after cell injection. IM injection improved cell retention and attenuated cardiac dysfunction, whereas IV, LV or LV* routes were somewhat inefficient (<1%). Cardiac BMC retention was not influenced by timing except for the IM injection that showed greater cell retention at 7 (16%) vs. 1, 2 or 3 (average of 7%) days post-MI. Cardiac cell retention was further improved by an injectable fibrin scaffold at day 3 post-MI (17 vs. 7%), even though morphometric and function parameters evaluated 4 weeks later displayed similar improvements. Conclusions/Significance These results show that cells injected post-MI display comparable tissue distribution profile regardless of the route of injection and that there is no time effect for cardiac cell accumulation for injections performed 1 to 3 days post-MI. As expected the IM injection is the most efficient for cardiac cell retention, it can be further improved by co-injection with a fibrin scaffold and it significantly attenuates cardiac dysfunction evaluated 4 weeks post myocardial infarction. These pharmacokinetic data obtained under similar experimental conditions are essential for further development of these novel approaches.
Collapse
Affiliation(s)
- Juliana S. Nakamuta
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Maria E. Danoviz
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Fabio L. N. Marques
- Radiopharmacy Laboratory, Nuclear Medicine Center, University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Leonardo dos Santos
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
- Cardiac Physiology and Pathophysiology Laboratory, Cardiology Division, Federal University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Claudia Becker
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Giovana A. Gonçalves
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Paula F. Vassallo
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Isolmar T. Schettert
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Paulo J. F. Tucci
- Cardiac Physiology and Pathophysiology Laboratory, Cardiology Division, Federal University of Sao Paulo, São Paulo, São Paulo, Brazil
| | - Jose E. Krieger
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
106
|
Abstract
Advances in noninvasive imaging techniques may aid in the understanding of cardiac stem cell therapy. Nuclear imaging enables in vivo evaluation of myocardial perfusion, metabolism, and function, in addition to the stem cell fate. This article summarizes recent clinical and experimental nuclear imaging studies in cardiac stem cell therapy.
Collapse
|
107
|
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
|
108
|
Kraitchman DL, Bulte JWM. In vivo imaging of stem cells and Beta cells using direct cell labeling and reporter gene methods. Arterioscler Thromb Vasc Biol 2009; 29:1025-30. [PMID: 19359666 DOI: 10.1161/atvbaha.108.165571] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cellular transplantation therapy offers a means to stimulate cardiovascular repair either by direct (graft-induced) or indirect (host-induced) tissue regeneration or angiogenesis. Typically, autologous or donor cells of specific subpopulations are expanded exogenously before administration to enrich the cells most likely to participate in tissue repair. In animal models of cardiovascular disease, the fate of these exogenous cells can be determined using histopathology. Recently, methods to label cells with contrast agents or transduce cells with reporter genes to produce imaging beacons has enabled the serial and dynamic assessment of the survival, fate, and engraftment of these cells with noninvasive imaging. Although cell tracking methods for cardiovascular applications have been most studied in stem or progenitor cells, research in tracking of whole islet transplants and particularly insulin producing beta cells has implications to the cardiovascular community attributable to the vascular changes associated with diabetes mellitus. In this review article, we will explore some of the state-of-the art methods for stem, progenitor, and beta cell tracking.
Collapse
Affiliation(s)
- Dara L Kraitchman
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Baltimore, MD, USA.
| | | |
Collapse
|
109
|
Smirnov P. Cellular magnetic resonance imaging using superparamagnetic anionic iron oxide nanoparticles: applications to in vivo trafficking of lymphocytes and cell-based anticancer therapy. Methods Mol Biol 2009; 512:333-53. [PMID: 19347287 DOI: 10.1007/978-1-60327-530-9_19] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
In current cancer research, the application of cytotoxic T lymphocytes with specificity to tumor antigens is regarded as a real therapeutic hope. The objective of imaging is to provide a follow-up of these killer cells in real time, in order to gain a better understanding of the mechanisms and action modes of lymphocytes on the tumor. Magnetic resonance imaging (MRI) has the advantage of the innocuousness of the applied magnetic field. Moreover, it has an exceptional spatial resolution allowing the visualization of anatomical areas without in-depth limitations. These features make MRI particularly adapted for cellular imaging. The use of " (ultrasmall) superparamagnetic iron oxide " particles [(U) SPIO] offers the adequate sensitivity required for cellular imaging. To promote a sufficient capture of these particles in nonphagocytic cells and make the cell of interest " detectable " by MRI after its injection, an important challenge in cellular imaging is to develop improved cell-labeling techniques. Superparamagnetic anionic nanoparticles (iron oxides of 10-nm diameter) are adsorbed in a nonspecific way on the membrane of the majority of cells, allowing their spontaneous internalization in intracellular vesicles. This pathway of cellular labeling confers a particular status to these nanoparticles as MRI contrast agents; the cells labeled in this manner possess magnetic and contrast properties that allow their in vivo detection and follow-up by MRI. This chapter describes the synthesis, the potential use, and the features of cellular labeling with these types of anionic nanoparticles. We also focus on the MRI contrast properties of the labeled cells, as well as on the feasibility of in vivo detection of immunizing circulating cells by MRI, with direct implications in cell-based anticancer therapy using lymphocytes.
Collapse
Affiliation(s)
- Pierre Smirnov
- Centre de Biophysique Moléculaire (CBM), UPR 4301 CNRS, Equipe Imagerie et Spectroscopie par Résonance Magnétique du Petit Animal, Orléans, France
| |
Collapse
|
110
|
Walker PA, Shah SK, Harting MT, Cox CS. Progenitor cell therapies for traumatic brain injury: barriers and opportunities in translation. Dis Model Mech 2009; 2:23-38. [PMID: 19132123 PMCID: PMC2615170 DOI: 10.1242/dmm.001198] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury (TBI) directly affects nearly 1.5 million new patients per year in the USA, adding to the almost 6 million cases in patients who are permanently affected by the irreversible physical, cognitive and psychosocial deficits from a prior injury. Adult stem cell therapy has shown preliminary promise as an option for treatment, much of which is limited currently to supportive care. Preclinical research focused on cell therapy has grown significantly over the last decade. One of the challenges in the translation of this burgeoning field is interpretation of the promising experimental results obtained from a variety of cell types, injury models and techniques. Although these variables can become barriers to a collective understanding and to evidence-based translation, they provide crucial information that, when correctly placed, offers the opportunity for discovery. Here, we review the preclinical evidence that is currently guiding the translation of adult stem cell therapy for TBI.
Collapse
Affiliation(s)
- Peter A. Walker
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - Shinil K. Shah
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - Matthew T. Harting
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | - Charles S. Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA
| |
Collapse
|
111
|
Bonaros N, Sondermejer H, Schuster M, Rauf R, Wang S, Seki T, Skerrett D, Itescu S, Kocher A. CCR3- and CXCR4-mediated interactions regulate migration of CD34+ human bone marrow progenitors to ischemic myocardium and subsequent tissue repair. J Thorac Cardiovasc Surg 2008; 136:1044-53. [DOI: 10.1016/j.jtcvs.2007.12.067] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 12/06/2007] [Accepted: 12/24/2007] [Indexed: 11/30/2022]
|
112
|
Schächinger V, Aicher A, Döbert N, Röver R, Diener J, Fichtlscherer S, Assmus B, Seeger FH, Menzel C, Brenner W, Dimmeler S, Zeiher AM. Pilot Trial on Determinants of Progenitor Cell Recruitment to the Infarcted Human Myocardium. Circulation 2008; 118:1425-32. [DOI: 10.1161/circulationaha.108.777102] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background—
Clinical trials indicate a beneficial effect of intracoronary infusion of progenitor cells on myocardial function in patients with ischemic heart disease. The extent and potential determinants of proangiogenic progenitor cell homing into the damaged myocardium after intracoronary infusion and the underlying mechanisms are still unknown.
Method and Results—
Circulating proangiogenic progenitor cells isolated from peripheral blood and cultivated for 3 days were labeled with radioactive indium oxine (
111
In-oxine). Radiolabeled proangiogenic progenitor cells (7.6±3.0 MBq, mean±SD) were administered to patients with previous myocardial infarction and a revascularized infarct vessel at various stages after infarction (5 days to 17 years). Viability of the infarcted myocardium was determined by
18
F-fluorodeoxyglucose–positron emission tomography and microcirculatory function by intracoronary Doppler measurements. One hour after application of progenitor cells, a mean of 6.9±4.7% (range, 1% to 19%; n=17) of total radioactivity was detected in the heart, which declined to 2±1% after 3 to 4 days. Average activity within the first 24 hours was highest among patients with acute myocardial infarction (≤14 days; 6.3±2.9%; n=8) and progressively decreased in patients treated in an intermediate phase (>14 days to 1 year; 4.5±3.2%; n=4) or a chronic stage (infarct age >1 year; 2.5±1.6%; n=5). Low viability of the infarcted myocardium and reduced coronary flow reserve were significant (
P
<0.05) predictors of proangiogenic progenitor cell homing.
Conclusions—
In patients after myocardial infarction undergoing intracoronary infusion of
111
In-oxine–labeled proangiogenic progenitor cells, a substantial amount of radioactivity is detected for several days in the heart, indicating homing of progenitor cells to the myocardium. The amount of proangiogenic progenitor cells retained in the heart decreased progressively with time after the acute myocardial infarction. Proangiogenic progenitor cells preferentially home to extensive acute myocardial infarcts characterized by low viability and reduced coronary flow reserve.
Collapse
Affiliation(s)
- Volker Schächinger
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Alexandra Aicher
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Natascha Döbert
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Rainer Röver
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Jürgen Diener
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Stephan Fichtlscherer
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Birgit Assmus
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Florian H. Seeger
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Christian Menzel
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Winfried Brenner
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Stefanie Dimmeler
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| | - Andreas M. Zeiher
- From the Department of Medicine III, Division of Cardiology (V.S., A.A., R.R., S.F., B.A., F.H.S., S.D., A.M.Z.), and Department of Nuclear Medicine (N.D., J.D., C.M.), J.W. Goethe University Frankfurt, Frankfurt am Main, and Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg (W.B.), Germany
| |
Collapse
|
113
|
Konoplyannikov AG, Petriev VM, Konoplyannikova OA, Kal’sina SS, Lepechina LA, Smorizanova OA, Semenkova IV, Agaeva EV. Effects of 60Co whole-body γ-irradiation in different doses on the distribution of 188Re-labeled autologous mesenchymal stem cells in Wistar rats after intravenous (systemic) transplantation during different periods after exposure. Bull Exp Biol Med 2008; 145:520-5. [DOI: 10.1007/s10517-008-0132-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
114
|
Mesenchymal stem cell administration at coronary artery reperfusion in the rat by two delivery routes: a quantitative assessment. Life Sci 2008; 83:511-5. [PMID: 18755200 DOI: 10.1016/j.lfs.2008.07.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 07/02/2008] [Accepted: 07/31/2008] [Indexed: 01/06/2023]
Abstract
AIMS Ideally, mesenchymal stem cells (MSC) home to and/or remain at the site of damaged myocardium when administered after myocardial infarction. However, MSC may not remain in the heart, but instead relocate to other areas. We investigated quantitatively the distribution of labeled rat MSC, given by two routes after coronary artery occlusion/reperfusion in rats. MAIN METHODS Rats were subjected to 45 min of coronary artery occlusion and 7 days of reperfusion. Before reperfusion rats received 2 x 10(6) MSC, labeled with europium, injected directly into the ischemic region of the heart (n = 9) or intravenously (n = 8). After 1 week tissues were analyzed for label content together with a standard curve of known quantities of labeled MSC. KEY FINDINGS In rats receiving cells injected directly into the myocardium, 15% of labeled cells were retained in the heart. When the cells were administered intravenously, no MSC were detected in the heart. The route of administration did not affect distribution to other organs, as the number of MSC in liver, spleen and lung was similar with both routes of delivery. SIGNIFICANCE Even with direct intramyocardial injection, only a small proportion of the cells are retained in the heart, instead traveling to other organs. With intravenous injection there was no evidence that cells "homed" to the damaged heart. Although cell delivery to the heart was significantly affected by the route of administration, the distribution of cells to other organs was similar with both routes of administration.
Collapse
|
115
|
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
|
116
|
Meier R, Piert M, Piontek G, Rudelius M, Oostendorp RA, Senekowitsch-Schmidtke R, Henning TD, Wels WS, Uherek C, Rummeny EJ, Daldrup-Link HE. Tracking of [18F]FDG-labeled natural killer cells to HER2/neu-positive tumors. Nucl Med Biol 2008; 35:579-88. [PMID: 18589302 DOI: 10.1016/j.nucmedbio.2008.02.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2007] [Revised: 02/08/2008] [Accepted: 02/28/2008] [Indexed: 12/01/2022]
Abstract
INTRODUCTION The objective of this study was to label the human natural killer (NK) cell line NK-92 with [(18)F]fluoro-deoxy-glucose (FDG) for subsequent in vivo tracking to HER2/neu-positive tumors. METHODS NK-92 cells were genetically modified to NK-92-scFv(FRP5)-zeta cells, which express a chimeric antigen receptor that is specific to the tumor-associated ErbB2 (HER2/neu) antigen. NK-92 and NK-92-scFv(FRP5)-zeta cells were labeled with [(18)F]FDG by simple incubation at different settings. Labeling efficiency was evaluated by a gamma counter. Subsequently, [(18)F]FDG-labeled parental NK-92 or NK-92-scFv(FRP5)-zeta cells were intravenously injected into mice with implanted HER2/neu-positive NIH/3T3 tumors. Radioactivity in tumors was quantified by digital autoradiography and correlated with histopathology. RESULTS The NK-92 and NK-92-scFv(FRP5)-zeta cells could be efficiently labeled with [(18)F]FDG by simple incubation. Optimal labeling efficiencies (80%) were achieved using an incubation period of 60 min and additional insulin (10 IU/ml). After injection of 5x10(6) [(18)F]FDG-labeled NK-92-scFv(FRP5)-zeta cells into tumor-bearing mice, digital autoradiography showed an increased uptake of radioactivity in HER2/neu-positive tumors at 60 min postinjection. Conversely, injection of 5x10(6) NK-92 cells not directed against HER2/neu receptors did not result in increased uptake of radioactivity in the tumors. Histopathology confirmed an accumulation of the NK-92-scFv(FRP5)-zeta cells, but not the parental NK cells, in tumor tissues. CONCLUSION The human NK cell line NK-92 can be directed against HER2/neu antigens by genetic modification. The genetically modified NK cells can be efficiently labeled with [(18)F]FDG, and the accumulation of these labeled NK cells in HER2/neu-positive tumors can be monitored with autoradiography.
Collapse
Affiliation(s)
- Reinhard Meier
- Department of Radiology, University of California-San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Psaltis PJ, Gronthos S, Worthley SG, Zannettino AC. Cellular Therapy for Cardiovascular Disease Part 2—Delivery of Cells and Clinical Experience. Clin Med Cardiol 2008. [DOI: 10.4137/117954682000200001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Peter J Psaltis
- Cardiovascular Research Centre, Royal Adelaide Hospital; Department of Medicine, University of Adelaide, South Australia, 5000
| | - Stan Gronthos
- Division of Haematology, Institute of Medical and Veterinary Science; Department of Medicine, University of Adelaide, South Australia, 5000
| | - Stephen G Worthley
- Cardiovascular Research Centre, Royal Adelaide Hospital; Department of Medicine, University of Adelaide, South Australia, 5000
| | - Andrew Cw Zannettino
- Division of Haematology, Institute of Medical and Veterinary Science; Department of Medicine, University of Adelaide, South Australia, 5000
| |
Collapse
|
118
|
Visualization of antigen-specific human cytotoxic T lymphocytes labeled with superparamagnetic iron-oxide particles. Eur Radiol 2008; 18:1087-95. [DOI: 10.1007/s00330-008-0874-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Revised: 01/08/2008] [Accepted: 01/16/2008] [Indexed: 11/27/2022]
|
119
|
Isostructural fluorescent and radioactive probes for monitoring neural stem and progenitor cell transplants. Nucl Med Biol 2008; 35:159-69. [DOI: 10.1016/j.nucmedbio.2007.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 09/25/2007] [Accepted: 11/02/2007] [Indexed: 01/17/2023]
|
120
|
Seo SB, Yang J, Lee ES, Jung Y, Kim K, Lee SY, Kim D, Suh JS, Huh YM, Haam S. Nanohybrids via a polycation-based nanoemulsion method for dual-mode detection of human mesenchymal stem cells. ACTA ACUST UNITED AC 2008. [DOI: 10.1039/b804544e] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
121
|
Louis DN. Molecular pathology of malignant gliomas. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2007; 2:277-305. [PMID: 18039109 DOI: 10.1146/annurev.pathol.2.010506.091930] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Malignant gliomas, the most common type of primary brain tumor, are a spectrum of tumors of varying differentiation and malignancy grades. These tumors may arise from neural stem cells and appear to contain tumor stem cells. Early genetic events differ between astrocytic and oligodendroglial tumors, but all tumors have an initially invasive phenotype, which complicates therapy. Progression-associated genetic alterations are common to different tumor types, targeting growth-promoting and cell cycle control pathways and resulting in focal hypoxia, necrosis, and angiogenesis. Knowledge of malignant glioma genetics has already impacted clinical management of these tumors, and researchers hope that further knowledge of the molecular pathology of malignant gliomas will result in novel therapies.
Collapse
Affiliation(s)
- David N Louis
- Molecular Pathology Unit, Department of Pathology and Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.
| |
Collapse
|
122
|
Cell therapy for myocardial infarction: Special delivery. J Mol Cell Cardiol 2007; 44:473-6. [PMID: 18234211 DOI: 10.1016/j.yjmcc.2007.10.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 10/30/2007] [Indexed: 01/14/2023]
|
123
|
Manganas LN, Zhang X, Li Y, Hazel RD, Smith SD, Wagshul ME, Henn F, Benveniste H, Djurić PM, Enikolopov G, Maletić-Savatić M. Magnetic resonance spectroscopy identifies neural progenitor cells in the live human brain. Science 2007; 318:980-5. [PMID: 17991865 PMCID: PMC4039561 DOI: 10.1126/science.1147851] [Citation(s) in RCA: 312] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The identification of neural stem and progenitor cells (NPCs) by in vivo brain imaging could have important implications for diagnostic, prognostic, and therapeutic purposes. We describe a metabolic biomarker for the detection and quantification of NPCs in the human brain in vivo. We used proton nuclear magnetic resonance spectroscopy to identify and characterize a biomarker in which NPCs are enriched and demonstrated its use as a reference for monitoring neurogenesis. To detect low concentrations of NPCs in vivo, we developed a signal processing method that enabled the use of magnetic resonance spectroscopy for the analysis of the NPC biomarker in both the rodent brain and the hippocampus of live humans. Our findings thus open the possibility of investigating the role of NPCs and neurogenesis in a wide variety of human brain disorders.
Collapse
Affiliation(s)
- Louis N. Manganas
- SUNY Stony Brook, Stony Brook, NY 11794, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Yao Li
- SUNY Stony Brook, Stony Brook, NY 11794, USA
| | - Raphael D. Hazel
- SUNY Stony Brook, Stony Brook, NY 11794, USA
- Brookhaven National Laboratory, Upton, NY 11719, USA
| | | | | | - Fritz Henn
- Brookhaven National Laboratory, Upton, NY 11719, USA
| | - Helene Benveniste
- SUNY Stony Brook, Stony Brook, NY 11794, USA
- Brookhaven National Laboratory, Upton, NY 11719, USA
| | | | | | - Mirjana Maletić-Savatić
- SUNY Stony Brook, Stony Brook, NY 11794, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| |
Collapse
|
124
|
Agnoletti G, Cargnoni A, Agnoletti L, Di Marcello M, Balzarini P, Gitti L, Martina P, Grigolato PG, Ferrari R. Percutaneous coronary injection of bone marrow cells in small experimental animals: small is not too small. Pathol Res Pract 2007; 203:801-8. [PMID: 17905524 DOI: 10.1016/j.prp.2007.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Revised: 07/04/2007] [Accepted: 07/20/2007] [Indexed: 10/22/2022]
Abstract
Intracoronary infusion of bone marrow cells (BMCs) is thought to induce cardiac regeneration in ischemic heart disease and dilated cardiomyopathy. The aim of our study was to develop a new method to inject BMCs into coronary arteries of small experimental animals. Transient atrioventricular block (AVB) was induced in 25 rats and 39 hamsters by intracarotid injection of adenosine 5'-triphosphate (ATP). Contrast echocardiography was obtained. BMCs (0.2-0.5 ml) were collected through femoral puncture, stained with PKH26 and injected into the carotid artery (CA). Animals were immediately sacrificed or followed for 1 month. To evaluate BMCs transfer from CA to myocardium, AVB and BMCs injections were performed in 10 hamsters subjected to coronary ligation for 30 min. Induction of transient AVB was possible in all animals by injecting 20-30 mg of ATP. Animals recovered a basal cardiac activity spontaneously or by dopamine injection. Flash injection of contrast medium through the CA induced staining of aortic root, coronary arteries, and myocardium. BMCs injection was possible in all cases. No immediate or late ECG changes were observed. Immediately after injection in healthy animals, histological examination showed the presence of BMCs in small coronary arteries and, after 1 month, the absence of infarction. In ischemic hearts, the presence of BMCs in the myocardium was observed 24h after ischemia. ATP-induced AVB block allows for percutaneous intracoronary injection of BMCs in small experimental animals with no immediate or late mortality and morbidity. This method offers new perspectives for the investigation of BMCs coronary infusion and engraftment in heart diseases.
Collapse
|
125
|
Krause U, Harter C, Seckinger A, Wolf D, Reinhard A, Bea F, Dengler T, Hardt S, Ho A, Katus HA, Kuecherer H, Hansen A. Intravenous delivery of autologous mesenchymal stem cells limits infarct size and improves left ventricular function in the infarcted porcine heart. Stem Cells Dev 2007; 16:31-7. [PMID: 17348804 DOI: 10.1089/scd.2006.0089] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Systemic delivery of bone marrow-derived mesenchymal stem cells (MSCs) is a noninvasive approach for myocardial repair. We aimed to test this strategy in a pig model of myocardial infarction. Pigs (n = 8) received autologous MSCs (1 x 10(6)/kg body weight) labeled with fluorescent dye 48 h post proximal left anterior descending artery (LAD) occlusion. Hemodyamics, infarct size, and myocardial function were assessed at baseline and after 1 month. Morphologic analysis revealed that labeled MSCs migrated in the peri-infarct region, resulting in smaller infarct size (32 +/- 7 vs. 19 +/- 7%, p = 0.01), higher fractional area shortening (23 +/- 3 vs. 34.0 +/- 7%, p = 0.001), lower left ventricular end diastolic pressure (18.7 +/- 5 vs. 10.2 +/- 4 mmHg, p = 0.02) and higher +dp/dt (4,570 +/- 540 vs. 6,742 +/- 700 mmHg/s, p = 0.03) during inotropic stimulation. Systemic intravenous delivery of MSCs to pigs limits myocardial infarct size and is an attractive approach for tissue repair.
Collapse
Affiliation(s)
- Ulf Krause
- Department of Hematology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Abstract
Cell-based therapy has been heralded as a promising, novel therapeutic strategy for cardiovascular diseases. Despite a rapid transition from animal studies to clinical trials, there remain numerous unresolved, and at times, controversial issues with respect to underlying molecular mechanisms. In parallel, recent advances in the field of molecular imaging has provided a means to bridge the gap in knowledge through in vivo stem cells tracking. Herein, we review current in vivo imaging techniques and future directions for tracking the effects of cell-based therapy.
Collapse
Affiliation(s)
- Kozo Hoshino
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | | | | | |
Collapse
|
127
|
Wolf D, Reinhard A, Krause U, Seckinger A, Katus HA, Kuecherer H, Hansen A. Stem cell therapy improves myocardial perfusion and cardiac synchronicity: new application for echocardiography. J Am Soc Echocardiogr 2007; 20:512-20. [PMID: 17484992 DOI: 10.1016/j.echo.2006.11.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2006] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Intravenous delivery of mesenchymal stem cell (MSC) is a noninvasive approach for myocardial tissue repair. We aimed to test this strategy in a pig model of myocardial infarction and to examine the usefulness of new echocardiographic applications to monitor cardioprotective effects of stem cell therapy. METHODS Pigs (n = 8) received autologous or allogeneic MSCs (1 x 10(6)/kg body weight) labeled with fluorescent dye 48 hours after proximal left anterior descending coronary artery occlusion. Infarct size, myocardial function, and perfusion (A x beta) were assessed by myocardial contrast echocardiography and standard histologic methods after 1 month. RESULTS Morphologic analysis revealed that labeled MSCs migrated in the peri-infarct region resulting in smaller infarct size by myocardial contrast echocardiography (control vs autologous and allogeneic MSC: 38 +/- 10% vs 25 +/- 5% and 28 +/- 6%, P < .01), higher fractional area shortening (23 +/- 3% vs 34.0 +/- 7% and 28 +/- 2%, P < .01), higher cardiac synchrony (167 +/- 36 vs 68 +/- 17 and 85 +/- 26 milliseconds, P < .003), and improved microvascular flow A x beta in the ischemic border zone (0.18 +/- 0.2 vs 0.56 +/- 0.3 and 0.49 +/- 0.2, P < .03). CONCLUSIONS Systemic delivery of autologous and allogeneic MSCs preserves myocardial viability even in large animals and is, therefore, an attractive approach for tissue repair. Myocardial contrast echocardiography is useful to evaluate microvascular perfusion, which was enhanced by MSCs.
Collapse
Affiliation(s)
- David Wolf
- Department of Cardiology and Angiology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
128
|
Beeres SLMA, Bengel FM, Bartunek J, Atsma DE, Hill JM, Vanderheyden M, Penicka M, Schalij MJ, Wijns W, Bax JJ. Role of imaging in cardiac stem cell therapy. J Am Coll Cardiol 2007; 49:1137-48. [PMID: 17367656 DOI: 10.1016/j.jacc.2006.10.072] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 10/11/2006] [Accepted: 10/23/2006] [Indexed: 12/17/2022]
Abstract
Stem cell therapy has emerged as a potential therapeutic option for cell death-related heart diseases. Preclinical and a number of early phase human studies suggested that cell therapy may augment perfusion and increase myocardial contractility. The rapid translation into clinical trials has left many issues unresolved, and emphasizes the need for specific techniques to visualize the mechanisms involved. Furthermore, the clinical efficacy of cell therapy remains to be proven. Imaging allows for in vivo tracking of cells and can provide a better understanding in the evaluation of the functional effects of cell-based therapies. In this review, a summary of the most promising imaging techniques for cell tracking is provided. Among these are direct labeling of cells with super-paramagnetic agents, radionuclides, and the use of reporter genes for imaging of transplanted cells. In addition, a comprehensive summary is provided of the currently available studies investigating a cell therapy-related effect on left ventricular function, myocardial perfusion, scar tissue, and myocardial viability.
Collapse
Affiliation(s)
- Saskia L M A Beeres
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Abstract
Drugs, surgery, and radiation are the traditional modalities of therapy in medicine. To these are being added new therapies based on cells and viruses or their derivatives. In these novel therapies, a cell or viral vector acts as a drug in its own right, altering the host or a disease process to bring about healing. Most of these advances originate from the significant recent advances in molecular medicine, but some have been around for some time. Blood transfusions and cowpox vaccinations are part of the history of medicine...but nevertheless are examples of cell- and viral-based therapies. This article focuses on the modern molecular incarnations of these therapies, and specifically on how imaging is used to track and guide these novel agents. We survey the literature dealing with imaging these new cell and viral particle therapies and provide a framework for understanding publications in this area. Leading technology of gene modifications are the fundamental modifications applied to make these new therapies amenable to imaging.
Collapse
Affiliation(s)
- Dawid Schellingerhout
- Neuroradiology Section, Department of Radiology and Experimental Diagnostic Imaging, Division of Diagnostic Imaging, M D Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
130
|
Schaller BJ, Modo M, Buchfelder M. Molecular Imaging of Brain Tumors: A Bridge Between Clinical and Molecular Medicine? Mol Imaging Biol 2007; 9:60-71. [PMID: 17203238 DOI: 10.1007/s11307-006-0069-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
As the research on cellular changes has shed invaluable light on the pathophysiology and biochemistry of brain tumors, clinical and experimental use of molecular imaging methods is expanding and allows quantitative assessment. The term molecular imaging is defined as the in vivo characterization and measurement of biologic processes at the cellular and molecular level. Molecular imaging sets forth to probe the molecular abnormalities that are the basis of disease rather than to visualize the end effects of these molecular alterations and, therefore, provides different additional biochemical or molecular information about primary brain tumors compared to histological methods "classical" neuroradiological diagnostic studies. Common clinical indications for molecular imaging contain primary brain tumor diagnosis and identification of the metabolically most active brain tumor reactions (differentiation of viable tumor tissue from necrosis), prediction of treatment response by measurement of tumor perfusion, or ischemia. The interesting key question remains not only whether the magnitude of biochemical alterations demonstrated by molecular imaging reveals prognostic value with respect to survival, but also whether it identifies early disease and differentiates benign from malignant lesions. Moreover, an early identification of treatment success or failure by molecular imaging could significantly influence patient management by providing more objective decision criteria for evaluation of specific therapeutic strategies. Specially, as molecular imaging represents a novel technology for visualizing metabolism and signal transduction to gene expression, reporter gene assays are used to trace the location and temporal level of expression of therapeutic and endogenous genes. Molecular imaging probes and drugs are being developed to image the function of targets without disturbing them and in mass amounts to modify the target's function as a drug. Molecular imaging helps to close the gap between in vitro and in vivo integrative biology of disease.
Collapse
Affiliation(s)
- B J Schaller
- Neuroscience Imaging, Department of Neurological Surgery, University of Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
| | | | | |
Collapse
|
131
|
Grøgaard HK, Sigurjonsson OE, Brekke M, Kløw NE, Landsverk KS, Lyberg T, Eriksen M, Egeland T, Ilebekk A. Cardiac accumulation of bone marrow mononuclear progenitor cells after intracoronary or intravenous injection in pigs subjected to acute myocardial infarction with subsequent reperfusion. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2007; 8:21-7. [PMID: 17293265 DOI: 10.1016/j.carrev.2006.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 09/08/2006] [Accepted: 09/08/2006] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The purpose of the present study was to compare the efficacy of intracoronary and intravenous injection of autologous progenitor cells for homing to the acutely infarcted but reperfused myocardium in pigs. METHODS Myocardial infarction was induced in 11 anesthetized pigs by 60-min balloon inflation in the mid LAD. After balloon deflation, reperfusion was verified and autologous CD31(+) progenitor cells, or bone marrow mononuclear cells, labeled with PKH67, were injected either intracoronarily (n=6) or intravenously (n=3). By autopsy, 4-5 days after induction of infarction, tissue from the heart and other organs was obtained for fluorescence microscopy. RESULTS In the heart, PKH(+) cells were detected throughout the reperfused infarcted myocardium, and the number of PKH(+) cells was significantly higher after intracoronary than after intravenous injection (3.2+/-0.55 vs. 0.33+/-0.17 cells/high-power field/10(6) cells injected, P=.01). Few PKH(+) cells were detected in the spleen, lung, mesenteric lymph node, and bone marrow. In an additional animal with a coil placed in the mid LAD, progenitor cells were not detected in the infarcted myocardium or in the normal myocardium. CONCLUSION Autologous mononuclear and CD31(+) cells from bone marrow accumulated in the infarcted myocardium when injected intracoronarily or intravenously after established reperfusion, and the accumulation of cells was significantly greater after intracoronary injection than after intravenous injection. Accumulation of PKH(+) cells did not appear in the normal myocardium or in the nonreperfused infarcted myocardium. PKH(+) cells were detected in spleen, lung, and bone marrow but to a lesser degree than in the infarcted myocardium.
Collapse
Affiliation(s)
- Haakon K Grøgaard
- Institute for Experimental Medical Research (IEMR), Ulleval University Hospital, N-0407 Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Abstract
Patients with ischemic cardiomyopathy have a poor prognosis despite all pharmacological, interventional and surgical treatment modalities currently applied. Heart transplantation remains the ideal treatment for this group of patients but the scarcity of donors hinders its widespread application. The autologous transplantation of stem cells (SCs) for cardiac repair is emerging as a new therapy for patients with myocardial dysfunction early after an acute infarction or ischemic cardiomyopathy. The rationale of this novel method is the enhancement of the repair mechanisms achieved by tissue-specific and circulating stem/progenitor cells. SCs assist naturally occurring myocardial repair by contributing to increased myocardial perfusion and contractile performance especially in the setting of acute myocardial infarction (AMI), but also in patients with chronic ischemic heart failure and advanced, diffuse coronary artery disease. The exact mechanism of their action has not been fully elucidated. Few studies continue to suggest a formation of few new contractile tissue. The majority if investigators believe that these cells do not persist long in the myocardium but that they secrete vascular growth and other cardioprotective factors.
Collapse
|
133
|
Abstract
Cardiac stem cell therapy is an innovative and promising therapeutic approach for heart failure. However, despite an increasing body of existing experimental and human data, it still presents a substantial challenge for basic scientists and clinical researchers. Several issues concerning biologic mechanisms of therapy remain to be answered, and unequivocal proof of clinical efficacy is needed. The variety of different available cell types and different methods for cell delivery to the myocardium raises further questions about the most useful therapeutic approach. Nuclear imaging not only provides accurate noninvasive information about myocardial perfusion, contractile function and viability, which enables assessment of clinical benefits of therapy. The rapidly developing field of molecular imaging has also brought up more specific tracers targeting cellular and subcellular biologic events, which are expected to shed more light upon mechanisms of cell therapy. Moreover, nuclear imaging is well suited for tracking of transplanted cells by use of direct radionuclide labeling or genetic labeling with reporter genes that can be targeted by radioactive reporter probes. Such a broad spectrum of available in vivo information is expected to significantly impact the future development of cell therapy towards a clinically accepted treatment.
Collapse
Affiliation(s)
- Frank M Bengel
- Division of Nuclear Medicine, Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University Medical Institutions, 601 N Caroline St, JHOC 3225, Baltimore, MD 21287, USA.
| |
Collapse
|
134
|
Nowak B, Weber C, Schober A, Zeiffer U, Liehn EA, von Hundelshausen P, Reinartz P, Schaefer WM, Buell U. Indium-111 oxine labelling affects the cellular integrity of haematopoietic progenitor cells. Eur J Nucl Med Mol Imaging 2006; 34:715-721. [PMID: 17096094 DOI: 10.1007/s00259-006-0275-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Accepted: 08/25/2006] [Indexed: 10/23/2022]
Abstract
PURPOSE Cell-based therapy by transplantation of progenitor cells has emerged as a promising development for organ repair, but non-invasive imaging approaches are required to monitor the fate of transplanted cells. Radioactive labelling with (111)In-oxine has been used in preclinical trials. This study aimed to validate (111)In-oxine labelling and subsequent in vivo and ex vivo detection of haematopoietic progenitor cells. METHODS Murine haematopoietic progenitor cells (10(6), FDCPmix) were labelled with 0.1 MBq (low dose) or 1.0 MBq (high dose) (111)In-oxine and compared with unlabelled controls. Cellular retention of (111)In, viability and proliferation were determined up to 48 h after labelling. Labelled cells were injected into the cavity of the left or right cardiac ventricle in mice. Scintigraphic images were acquired 24 h later. Organ samples were harvested to determine the tissue-specific activity. RESULTS Labelling efficiency was 75 +/- 14%. Cellular retention of incorporated (111)In after 48 h was 18 +/- 4%. Percentage viability after 48 h was 90 +/- 1% (control), 58 +/- 7% (low dose) and 48 +/- 8% (high dose) (p<0.0001). Numbers of viable cells after 48 h (normalised to 0 h) were 249 +/- 51% (control), 42 +/- 8% (low dose) and 32 +/- 5% (high dose) (p<0.0001). Cells accumulated in the spleen (86.6 +/- 27.0% ID/g), bone marrow (59.1 +/- 16.1% ID/g) and liver (30.3 +/- 9.5% ID/g) after left ventricular injection, whereas most of the cells were detected in the lungs (42.4 +/- 21.8% ID/g) after right ventricular injection. CONCLUSION Radiolabelling of haematopoietic progenitor cells with (111)In-oxine is feasible, with high labelling efficiency but restricted stability. The integrity of labelled cells is significantly affected, with substantially reduced viability and proliferation and limited migration after systemic transfusion.
Collapse
Affiliation(s)
- Bernd Nowak
- Department of Nuclear Medicine, University Hospital, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Christian Weber
- Department of Molecular Cardiovascular Research, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Andreas Schober
- Department of Molecular Cardiovascular Research, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Ute Zeiffer
- Department of Molecular Cardiovascular Research, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Elisa A Liehn
- Department of Molecular Cardiovascular Research, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Philipp von Hundelshausen
- Department of Molecular Cardiovascular Research, University Hospital, RWTH Aachen University, Aachen, Germany
| | - Patrick Reinartz
- Department of Nuclear Medicine, University Hospital, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Wolfgang M Schaefer
- Department of Nuclear Medicine, University Hospital, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulrich Buell
- Department of Nuclear Medicine, University Hospital, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
135
|
Mayer-Kuckuk P, Boskey AL. Molecular imaging promotes progress in orthopedic research. Bone 2006; 39:965-977. [PMID: 16843078 DOI: 10.1016/j.bone.2006.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Revised: 04/13/2006] [Accepted: 05/05/2006] [Indexed: 02/03/2023]
Abstract
Modern orthopedic research is directed towards the understanding of molecular mechanisms that determine development, maintenance and health of musculoskeletal tissues. In recent years, many genetic and proteomic discoveries have been made which necessitate investigation under physiological conditions in intact, living tissues. Molecular imaging can meet this demand and is, in fact, the only strategy currently available for noninvasive, quantitative, real-time biology studies in living subjects. In this review, techniques of molecular imaging are summarized, and applications to bone and joint biology are presented. The imaging modality most frequently used in the past was optical imaging, particularly bioluminescence and near-infrared fluorescence imaging. Alternate technologies including nuclear and magnetic resonance imaging were also employed. Orthopedic researchers have applied molecular imaging to murine models including transgenic mice to monitor gene expression, protein degradation, cell migration and cell death. Within the bone compartment, osteoblasts and their stem cells have been investigated, and the organic and mineral bone phases have been assessed. These studies addressed malignancy and injury as well as repair, including fracture healing and cell/gene therapy for skeletal defects. In the joints, molecular imaging has focused on the inflammatory and tissue destructive processes that cause arthritis. As described in this review, the feasibility of applying molecular imaging to numerous areas of orthopedic research has been demonstrated and will likely result in an increase in research dedicated to this powerful strategy. Molecular imaging holds great promise in the future for preclinical orthopedic research as well as next-generation clinical musculoskeletal diagnostics.
Collapse
Affiliation(s)
- Philipp Mayer-Kuckuk
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York 10021, USA; Memorial Sloan-Kettering Cancer Center, New York 10021, USA.
| | - Adele L Boskey
- Bone Cell Biology and Imaging Laboratory, Hospital for Special Surgery, New York 10021, USA
| |
Collapse
|
136
|
Zhou R, Acton PD, Ferrari VA. Imaging stem cells implanted in infarcted myocardium. J Am Coll Cardiol 2006; 48:2094-106. [PMID: 17112999 PMCID: PMC2597078 DOI: 10.1016/j.jacc.2006.08.026] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Revised: 08/14/2006] [Accepted: 08/14/2006] [Indexed: 01/14/2023]
Abstract
Stem cell-based cellular cardiomyoplasty represents a promising therapy for myocardial infarction. Noninvasive imaging techniques would allow the evaluation of survival, migration, and differentiation status of implanted stem cells in the same subject over time. This review describes methods for cell visualization using several corresponding noninvasive imaging modalities, including magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, and bioluminescent imaging. Reporter-based cell visualization is compared with direct cell labeling for short- and long-term cell tracking.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
137
|
Mäkinen S, Kekarainen T, Nystedt J, Liimatainen T, Huhtala T, Närvänen A, Laine J, Jolkkonen J. Human umbilical cord blood cells do not improve sensorimotor or cognitive outcome following transient middle cerebral artery occlusion in rats. Brain Res 2006; 1123:207-15. [PMID: 17070789 DOI: 10.1016/j.brainres.2006.09.056] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 09/11/2006] [Accepted: 09/11/2006] [Indexed: 11/17/2022]
Abstract
The present study investigated effects of human umbilical cord blood (HUCB) cells on sensorimotor, cognitive, and histological outcome in rats subjected to transient middle cerebral artery occlusion (MCAO). Halothane anesthetized adult male Wistar rats were subjected to transient MCAO for 2 h. HUCB cells (mononuclear 1-5x10(7) or Lin(-) cells 1-5x10(5)) were administered intravenously after 24 h recovery. The limb-placing test was performed on postoperative days 2, 4, 6, 9, 12, 16, and 20. In addition, beam-walking and cylinder tests were used to assess sensorimotor function at baseline, and on postoperative days 4, 12, and 20. Morris water-maze was used to assess cognitive performance on postoperative days 22-24. Subsequently, rats were perfused for measurement of infarct volumes and detection of HUCB cells by immunohistochemistry (MAB1281). MCAO rats showed a partial spontaneous recovery in sensorimotor function during the follow-up. However, the recovery profile was similar in MCAO controls and in MCAO rats that received HUCB cells. HUCB did not affect impaired water-maze performance of MCAO rats. Only few human nuclei-specific MAB1281-positive cells were detected in the ipsilateral hemisphere in MCAO rats that received HUCB cells. Infarct volumes did not differ between the experimental groups. A group of additional rats were used to further study biodistribution of intravenously given (111)In-oxine-labelled mononuclear HUCB cells in MCAO and sham-operated rats. SPECT imaging data indicated a high tracer uptake in the lung, liver, spleen, and kidney, but not in the brain immediately after administration or 24 h post-administration. The present study suggests that HUCB cells do not improve functional recovery or histological outcome in MCAO rats after systemic administration because of limited migration of cells in the ischemic brain.
Collapse
Affiliation(s)
- Susanna Mäkinen
- Department of Neuroscience and Neurology, University of Kuopio, P.O. Box 1627, 70211 Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Stodilka RZ, Blackwood KJ, Kong H, Prato FS. A method for quantitative cell tracking using SPECT for the evaluation of myocardial stem cell therapy. Nucl Med Commun 2006; 27:807-13. [PMID: 16969264 DOI: 10.1097/01.mnm.0000237987.31597.cf] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE A promising SPECT-based method for evaluating stem cells therapy uses (111)In-labelled cells, transfected with a reporter gene. Cells are first transplanted to the infarct, and subsequently interrogated for transgenic expression using a systemic injection of an (131)I-labelled reporter probe. The method is impeded by the physical effects of scatter, (131)I/(111)In cross-talk, and attenuation. We hypothesize that correcting for physical effects improves detection of transgenic expression in transplanted cells when (111)In localization is available. METHODS Canine bone marrow mesenchymal cells (BMMCs), radiolabelled and transfected, were injected into infarcted myocardium. Next, a reporter probe was injected systemically, and 22 SPECT scans were acquired over 20 h. Finally, (99m)Tc-sestamibi was injected and imaged. The animal was killed, the heart sectioned, and counted for (131)I and (111)In in a well-counter ('gold standard'). Canine SPECTs were reconstructed in two ways: with corrections for physical effects and without corrections. The first (111)In reconstruction and the (99m)Tc reconstruction were used to define volumes-of-interest over the transplanted BMMC (VBMMC) and normal myocardium (VNM), respectively. RESULTS (131)I reconstructions without corrections for physical effects had negligible differential uptake. With corrections, VBMMC was consistently higher than VNM, demonstrating transgene expression. (131)I had the following VBMMC:VNM activity ratio: without correction for physical effects=0.869; with corrections=1.23; and well-counter=1.21. VNM showed the following (131)I:(111)In activity ratio: without corrections=3.07; with corrections=1.38; and well-counter=1.58. CONCLUSIONS In dual-isotope SPECT, corrections for physical effects were required to detect transgene expression in cells transplanted into an infarction when localization information was available.
Collapse
Affiliation(s)
- Robert Z Stodilka
- Imaging Program, Lawson Health Research Institute, University of Western Ontario, Ontario, Canada.
| | | | | | | |
Collapse
|
139
|
Abstract
Nuclear cardiology has made significant advances since the first reports of planar scintigraphy for the evaluation of left ventricular perfusion and function. While the current "state of the art" of gated myocardial perfusion single-photon emission computed tomographic (SPECT) imaging offers invaluable diagnostic and prognostic information for the evaluation of patients with suspected or known coronary artery disease (CAD), advances in the cellular and molecular biology of the cardiovascular system have helped to usher in a new modality in nuclear cardiology, namely, molecular imaging. In this review, we will discuss the current state of the art in nuclear cardiology, which includes SPECT and positron emission tomographic evaluation of myocardial perfusion, evaluation of left ventricular function by gated myocardial perfusion SPECT and gated blood pool SPECT, and the evaluation of myocardial viability with PET and SPECT methods. In addition, we will discuss the future of nuclear cardiology and the role that molecular imaging will play in the early detection of CAD at the level of the vulnerable plaque, the evaluation of cardiac remodeling, and monitoring of important new therapies including gene therapy and stem cell therapy.
Collapse
|
140
|
Abstract
Cardiovascular disease is a major public health challenge in the western world. Mortality of acute events has improved, but more patients develop HF--a condition affecting up to 22 million people worldwide. Cell transplantation is the first therapy to attempt replacement of lost cardiomyocytes and vasculature to restore lost contractile function. Since the first reported functional repair after injection of autologous skeletal myoblasts into the injured heart in 1998, a variety of cell types have been proposed for transplantation in different stages of cardiovascular disease. Fifteen years of preclinical research and the rapid move into clinical studies have left us with promising results and a better understanding of cells as a potential clinical tool. Cell-based cardiac repair has been the first step, but cardiac regeneration remains the more ambitious goal. Promising new cell types and the rapidly evolving concept of adult stem and progenitor cell fate may enable us to move towards regenerating viable and functional myocardium. Meeting a multidisciplinary consensus will be required to translate these findings into safe and applicable clinical tools.
Collapse
Affiliation(s)
- Harald C Ott
- Massachusetts General Hospital, Department of Surgery, Boston, MA, USA
| | | |
Collapse
|
141
|
Bindslev L, Haack-Sørensen M, Bisgaard K, Kragh L, Mortensen S, Hesse B, Kjaer A, Kastrup J. Labelling of human mesenchymal stem cells with indium-111 for SPECT imaging: effect on cell proliferation and differentiation. Eur J Nucl Med Mol Imaging 2006; 33:1171-7. [PMID: 16763813 DOI: 10.1007/s00259-006-0093-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2005] [Accepted: 02/07/2006] [Indexed: 02/03/2023]
Abstract
PURPOSE Stem cell therapy seems to be a new treatment option within cardiac diseases to improve myocardial perfusion and function. However, the delivery and traceability of the cells represent a problem. Radioactive labelling with 111In could be a method for tracking mesenchymal stem cells (MSCs). However, 111In could influence the viability and differentiation capacity of MSCs, which would limit its use. Therefore, the aim of this study was to evaluate the influence of 111In labelling in doses relevant for SPECT imaging in humans on the viability and differentiation capacity of human MSCs. METHODS AND RESULTS Human MSCs isolated from bone marrow were incubated with 111In-tropolone (15-800 Bq/cell). The labelling efficiency was approximately 25% with 30 Bq/cell 111In. The MSC doubling time was 1.04+/-0.1 days and was not influenced by 111In within the range 15-260 Bq/cell. Using 30 Bq 111In/cell it was possible to label MSCs to a level relevant for clinical scintigraphic use. With this dose, 111In had no effect on characteristic surface and intracellular markers of cultured MSCs analysed both by flow cytometry and by real-time polymerase chain reaction. Further, the labelled MSCs differentiated towards endothelial cells and formed vascular structures. CONCLUSION It is possible to label human MSCs with 111In for scintigraphic tracking of stem cells delivered to the heart in clinical trials without affecting the viability and differentiation capacity of the MSCs. This creates an important tool for the control of stem cell delivery and dose response in clinical cardiovascular trials.
Collapse
Affiliation(s)
- L Bindslev
- Stem Cell Laboratory 9312, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Abstract
The purpose of this study was to characterize the performance of single photon emission computed tomography (SPECT) in tasks associated with tracking transplanted cells. Previous studies identified matters of hardware design, whereas we focus on biological variables impacting system performance, such as cell colony growth and non-specific radiolabelling. Using experimental data, a digital phantom was developed of in vitro 111In-radiolabelled stem cells, transfected with a reporter gene, transplanted into canine infarcted myocardium and interrogated using a peripherally injected 131I-radiolabelled reporter probe. Single- and dual-head SPECT acquisition was simulated. Performance was characterized using an estimation task, where the precision of parameter estimates (111In and 131I radiolabel quantity, cell colony size and location, and background) was tracked as the phantom evolved to simulate 111In-label efflux, cell colony growth and improved reporter probe specificity. In vitro pre-labelling of transplanted cells improved precision of parameter estimates via a priori size and location information. Precision of radiolabel quantity estimates improved with cell colony growth, despite 111In radiolabel dilution; size and location parameters were influenced little. Precision of radiolabel quantity estimates improved with reduced reporter probe non-specific uptake. The performance of SPECT in cell tracking is influenced strongly by biological variables. These should be considered when planning experiments or developing SPECT technology for cell tracking.
Collapse
Affiliation(s)
- Robert Z Stodilka
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada.
| | | | | |
Collapse
|
143
|
Graham JJ, Lederman RJ, Dick AJ. Magnetic resonance imaging and its role in myocardial regenerative therapy. Regen Med 2006; 1:347-55. [PMID: 17465788 DOI: 10.2217/17460751.1.3.347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
There has been extensive interest recently in cardiac stem cell therapy. Current research has been hampered by differences in cell type, methods of delivery and efficacy evaluation. In this article we review the use of magnetic resonance imaging in this growing area and argue that it is well suited to all areas of myocardial regeneration: from patient identification, through cell delivery and tracking of appropriately labeled cells, to evaluation of therapeutic effect. Potential future advances are discussed including magnetic resonance imaging-guided intervention suites and the use of higher field strength magnets for cell tracking.
Collapse
Affiliation(s)
- John J Graham
- Division of Cardiology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | |
Collapse
|
144
|
Stuckey DJ, Carr CA, Martin-Rendon E, Tyler DJ, Willmott C, Cassidy PJ, Hale SJM, Schneider JE, Tatton L, Harding SE, Radda GK, Watt S, Clarke K. Iron particles for noninvasive monitoring of bone marrow stromal cell engraftment into, and isolation of viable engrafted donor cells from, the heart. Stem Cells 2006; 24:1968-75. [PMID: 16627684 DOI: 10.1634/stemcells.2006-0074] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Stem cells offer a promising approach to the treatment of myocardial infarction and prevention of heart failure. We have used iron labeling of bone marrow stromal cells (BMSCs) to noninvasively track cell location in the infarcted rat heart over 16 weeks using cine-magnetic resonance imaging (cine-MRI) and to isolate the BMSCs from the grafted hearts using the magnetic properties of the donor cells. BMSCs were isolated from rat bone marrow, characterized by flow cytometry, transduced with lentiviral vectors expressing green fluorescent protein (GFP), and labeled with iron particles. BMSCs were injected into the infarct periphery immediately following coronary artery ligation, and rat hearts were imaged at 1, 4, 10, and 16 weeks postinfarction. Signal voids caused by the iron particles in the BMSCs were detected in all rats at all time points. In mildly infarcted hearts, the volume of the signal void decreased over the 16 weeks, whereas the signal void volume did not decrease significantly in severely infarcted hearts. High-resolution three-dimensional magnetic resonance (MR) microscopy identified hypointense regions at the same position as in vivo. Donor cells containing iron particles and expressing GFP were identified in MR-targeted heart sections after magnetic cell separation from digested hearts. In conclusion, MRI can be used to track cells labeled with iron particles in damaged tissue for at least 16 weeks after injection and to guide tissue sectioning by accurately identifying regions of cell engraftment. The magnetic properties of the iron-labeled donor cells can be used for their isolation from host tissue to enable further characterization.
Collapse
Affiliation(s)
- Daniel J Stuckey
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, UK, OX1 3PT
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
The discovery of adult progenitor cells capable of generating new vascular and myocardial tissue offers the promise of salvage of ischaemically threatened or irreversibly damaged cardiac tissue. Not surprisingly, great interest has focused on the use of a variety of cell types to treat both acute myocardial infarction and chronic ischaemic heart disease. This review focuses on the treatment of these two categories of disease, the cell types being considered, our understanding of timing and methods of cellular administration, and possible mechanisms of myocardial salvage.
Collapse
Affiliation(s)
- Thomas J Povsic
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Box 3126, Durham, NC 27710, USA.
| | | |
Collapse
|
146
|
Tran N, Poussier S, Franken PR, Maskali F, Groubatch F, Vanhove C, Antunes L, Karcher G, Villemot JP, Marie PY. Feasibility of in vivo dual-energy myocardial SPECT for monitoring the distribution of transplanted cells in relation to the infarction site. Eur J Nucl Med Mol Imaging 2006; 33:709-15. [PMID: 16572303 DOI: 10.1007/s00259-006-0075-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 01/02/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Cell therapy using bone marrow mesenchymal stem cells (BMSCs) shows promise in the treatment of myocardial infarction (MI) but accurate cell delivery within MI areas remains critical. In the present study, we tested the feasibility of in vivo pinhole SPECT imaging for monitoring the sites of intramyocardial implanted BMSCs in relation to targeted MI areas in rats. METHODS BMSCs were labelled with (111)In-oxine and injected within the fibrotic areas of 3-month-old MI in ten rats. Two days later, dual (111)In/(99m)Tc-sestamibi pinhole SPECT was recorded for localisation of (111)In-BMSCs on a 15-segment left ventricular (LV) division. Additional (99m)Tc-sestamibi pinhole SPECT had been performed 1 month earlier and on the day before transplantation. In vitro counting on histological sections was used to validate the pinhole SPECT determination of (111)In-BMSC activity within LV segments. RESULTS The underperfused MI area (segments with <70% uptake) was stable between the (99m)Tc-sestamibi SPECT study recorded at 1 month (4.6+/-1.9 segments) and at 1 day (4.7+/-2.3 segments) before transplantation. (111)In-BMSCs were detected by dual-energy SPECT in 56 segments: 33 (59%) were underperfused MI segments but 23 (41%) were not (14 adjacent and nine remote segments). Finally, (111)In-labelled BMSCs were not detected in 14 out of the 47 (30%) underperfused MI segments. CONCLUSION When BMSCs are injected within MI areas in rats, sites of early cell retention do not always match the targeted MI areas. The dual-energy pinhole SPECT technique may be used for monitoring the sites of early retention of implanted BMSCs and the data obtained may have critical importance when analysing the effects of cardiac cell therapy.
Collapse
Affiliation(s)
- Nguyen Tran
- Laboratory of Surgery School, Faculty of Medicine, UHP-Nancy, Avenue de la forêt de Haye, 54500, Vandoeuvre-lès-Nancy, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Ma B, Hankenson KD, Dennis JE, Caplan AI, Goldstein SA, Kilbourn MR. A simple method for stem cell labeling with fluorine 18. Nucl Med Biol 2006; 32:701-5. [PMID: 16243645 DOI: 10.1016/j.nucmedbio.2005.04.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2005] [Revised: 04/22/2005] [Accepted: 04/23/2005] [Indexed: 10/25/2022]
Abstract
Hexadecyl-4-[(18)F]fluorobenzoate ([(18)F]HFB), a long chain fluorinated benzoic acid ester, was prepared in a one-step synthesis by aromatic nucleophilic substitution of [(18)F]fluoride ion on hexadecyl-4-(N,N,N-trimethylammonio)benzoate. The radiolabeled ester was obtained in good yields (52% decay corrected) and high purity (97%). [(18)F]HFB was used to radiolabel rat mesenchymal stem cells (MSCs) by absorption into cell membranes. MicroPET imaging of [(18)F]HFB-labeled MSCs following intravenous injection into the rat showed the expected high and persistent accumulation of radioactivity in the lungs. [(18)F]HFB is thus simple to prepare and uses labeling agent for short-term distribution studies of injected stem cells.
Collapse
Affiliation(s)
- Bing Ma
- Department of Radiology, Division of Nuclear Medicine, University of Michigan Medical School, Ann Arbor, 48109, USA
| | | | | | | | | | | |
Collapse
|
148
|
Abstract
Cardiovascular cell therapy offers the first real potential to treat the underlying injuries associated with cardiac and vascular disease. By delivering appropriate exogenous cells to an injury site, the potential exists to mitigate injury or even to begin to reverse damage. Based on their inordinate pre-clinical promise as myogenic or angiogenic precursors, skeletal myoblasts and bone marrow or blood-derived mesenchymal and hematopoietic progenitor cells have all rapidly moved from bench to early clinical studies. From these parallel paths we are learning a number of useful lessons and have begun to visualize the hurdles to be overcome as we move these therapies forward. It is now obvious that cell-based cardiac and vascular repair are feasible-both early and later in the disease process. In fact, cell therapy may offer an unparalleled opportunity for improvement to millions of individuals living with cardiovascular disease. However, many questions about the technology remain. The mechanisms associated with cardiovascular repair remain unclear. Whether a best cell type, delivery method, or route of administration exists is unknown. And, whether cellbased disease prevention is feasible is still unanswerable. Now is the time to delve deeply into the questions of cell-based myocardial and vascular repair-even as we cautiously proceed clinically. Only by understanding these issues will we be able to decrease unanticipated clinical effects and to fulfill the potential promise of the most exciting opportunity yet to treat CVD. As we do so, we must prevent uncontrolled, poorly planned studies and until we understand cell therapy's potential, we must limit "too good to be true" promises. Only by addressing unanswered questions, carefully limiting our promises, and rigorously performing pre-clinical and clinical studies can we provide the surest opportunity for safely moving the field forward.
Collapse
Affiliation(s)
- H C Ott
- Center for Cardiovascular Repair, University of Minnesota, Minnesota 55455, USA
| | | | | |
Collapse
|
149
|
Bunnell BA, Deng W, Robinson CM, Waldron PR, Bivalacqua TJ, Baber SR, Hyman AL, Kadowitz PJ. Potential application for mesenchymal stem cells in the treatment of cardiovascular diseases. Can J Physiol Pharmacol 2006; 83:529-39. [PMID: 16091779 DOI: 10.1139/y05-043] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Stem cells isolated from various sources have been shown to vary in their differentiation capacity or pluripotentiality. Two groups of stem cells, embryonic and adult stem cells, may be capable of differentiating into any desired tissue or cell type, which offers hope for the development of therapeutic applications for a large number of disorders. However, major limitations with the use of embryonic stem cells for human disease have led researchers to focus on adult stem cells as therapeutic agents. Investigators have begun to examine postnatal sources of pluripotent stem cells, such as bone marrow stroma or adipose tissue, as sources of mesenchymal stem cells. The following review focuses on recent research on the use of stem cells for the treatment of cardiovascular and pulmonary diseases and the future application of mesenchymal stem cells for the treatment of a variety of cardiovascular disorders.
Collapse
Affiliation(s)
- Bruce A Bunnell
- Department of Pharmacology, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Bengel FM, Schachinger V, Dimmeler S. Cell-based therapies and imaging in cardiology. Eur J Nucl Med Mol Imaging 2005; 32 Suppl 2:S404-16. [PMID: 16205898 DOI: 10.1007/s00259-005-1898-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cell therapy for cardiac repair has emerged as one of the most exciting and promising developments in cardiovascular medicine. Evidence from experimental and clinical studies is increasing that this innovative treatment will influence clinical practice in the future. But open questions and controversies with regard to the basic mechanisms of this therapy continue to exist and emphasise the need for specific techniques to visualise the mechanisms and success of therapy in vivo. Several non-invasive imaging approaches which aim at tracking of transplanted cells in the heart have been introduced. Among these are direct labelling of cells with radionuclides or paramagnetic agents, and the use of reporter genes for imaging of cell transplantation and differentiation. Initial studies have suggested that these molecular imaging techniques have great potential. Integration of cell imaging into studies of cardiac cell therapy holds promise to facilitate further growth of the field towards a broadly clinically useful application.
Collapse
Affiliation(s)
- Frank M Bengel
- Nuklearmedizinische Klinik und Poliklinik, Technische Universität München, Ismaninger Strasse 22, 81675, Munich, Germany.
| | | | | |
Collapse
|