Superparamagnetic iron oxide nanoparticles: diagnostic magnetic resonance imaging and potential therapeutic applications in neurooncology and central nervous system inflammatory pathologies, a review

Jason S Weinstein, Csanad G Varallyay, Edit Dosa, Seymur Gahramanov, Bronwyn Hamilton, William D Rooney, Leslie L Muldoon, Edward A Neuwelt, Jason S Weinstein, Csanad G Varallyay, Edit Dosa, Seymur Gahramanov, Bronwyn Hamilton, William D Rooney, Leslie L Muldoon, Edward A Neuwelt

Abstract

Superparamagnetic iron oxide nanoparticles have diverse diagnostic and potential therapeutic applications in the central nervous system (CNS). They are useful as magnetic resonance imaging (MRI) contrast agents to evaluate: areas of blood-brain barrier (BBB) dysfunction related to tumors and other neuroinflammatory pathologies, the cerebrovasculature using perfusion-weighted MRI sequences, and in vivo cellular tracking in CNS disease or injury. Novel, targeted, nanoparticle synthesis strategies will allow for a rapidly expanding range of applications in patients with brain tumors, cerebral ischemia or stroke, carotid atherosclerosis, multiple sclerosis, traumatic brain injury, and epilepsy. These strategies may ultimately improve disease detection, therapeutic monitoring, and treatment efficacy especially in the context of antiangiogenic chemotherapy and antiinflammatory medications. The purpose of this review is to outline the current status of superparamagnetic iron oxide nanoparticles in the context of biomedical nanotechnology as they apply to diagnostic MRI and potential therapeutic applications in neurooncology and other CNS inflammatory conditions.

Figures

Figure 1
Figure 1
(A) 1–8: Adapted with permission from Varallyay et al (2002). Patient with anaplastic oligodendroglioma. A.1–2: nonenhanced (A.1) and gadolinium-enhanced (A.2) SE T1-weighted images of a right temporal tumor. The gadolinium-enhanced image shows strong, lobulated peripheral enhancement with a central nonenhancing region. A.3: at 24 h after ferumoxtran-10 (Combidex) infusion, SE T1-weighted image shows high-signal intensity in a similar distribution, but with less peripheral lobulation, compared with the gadolinium-enhanced image. Also note that the nongadolinium-enhancing central zone became isointense to white matter, suggesting some ferumoxtran accumulation. A.4–6: fast SE T2-weighted image obtained before ferumoxtran-10 infusion (A.4) and fast SE T2-weighted (A.5) and GRE T2*-weighted (A.6) images obtained 24 h after ferumoxtran-10 infusion show a heterogeneous tumor mass with peripheral decreased signal intensity that is more prominent on the GRE T2*-weighted image. The distribution of low-signal intensity is similar to that of the high-signal intensity areas on the SE T1-weighted image (A.3). A.7–8: photomicrographs of pathologic specimens stained for iron (DAB-enhanced Perls stain). A.7: (original magnification—7.5 × ; bar indicates 1 mm), tumor (T) and reactive brain interface (RB) show intense staining for iron at the periphery of the tumor. A.8: (original magnification—100 × ; bar indicates 0.1 mm), cellular iron staining at the tumor-reactive brain interface shows iron uptake by parenchymal cells with fibrillar processes (arrows) rather than within the round tumor cells (T) themselves. (B) Adapted with permission from Muldoon et al (2005). Iron particle imaging of human small-cell lung cancer (LX-1 cell line) xenografts in a rodent model. B.1–8: T1 MRI scans (1.5 T) comparing ferumoxtran-10 (top), ferumoxytol (middle), and ferumoxides (bottom) 24 h after intravenous administration in nude rats with LX-1 intracerebral tumors. All animals were imaged at 9 days after tumor cell implantation except #3 and #4, which were imaged on day 6. B.9–10: iron histochemistry photomicrographs. Intense iron staining was found at the LX-1 tumor/reactive brain interface 24 h after administration of ferumoxtran-10 or ferumoxytol.
Figure 2
Figure 2
(A) MRI showing cerebral vasculature at 7 T. Minimum intensity projection (mIP) susceptibility-weighted images were obtained without contrast agent (A.1), with GBCA (gadoteridol, 0.1 mmol/kg, A.2), and immediately after USPIO injection (ferumoxytol 510 mg, A.3). The USPIO enhancement results in superior visualization of small intracerebral vessels. (B) 3D MPRAGE T1-weighted MRI in a patient at 3 T and 7 T without contrast (B.1 and B.4), with GBCA (B.2 and B.5), and with ferumoxytol (510 mg, 24 h after infusion—B.3 and B.6). GBCA associated T1-weighted signal intensity increases at higher magnetic field strength because of the increased nominal brain tissue T1 relaxation time constant, whereas ferumoxytol enhancement signal intensity is reduced because of increased T2* contributions at 7 T. (C) Adapted with permission from Neuwelt et al (2007). Time course of ferumoxytol enhancement in a patient with recurrent GBM. GBCA-enhanced T1-weighted (C.1), T2-weighted (C.2), and post-ferumoxytol T1-weighted (C.3–7) MRI scans were obtained using a 1.5 T MRI at five time points (C.3=4–6 h; C.4=6–20 h; C.5=24–28 h; C.6=48–52 h; C.7=∼72 h). Peak intensity is observed at the 24 to 28 h time point; much later than that observed with GBCA (not shown), which enhances maximally 3.5 to 25 mins after injection. The figure shows comparison of ferumoxytol-enhancement intensity curves at 1.5 T and 3 T MRI. Maximum ferumoxytol T1 enhancement is greater at 1.5 T than on 3 T.
Figure 3
Figure 3
(A) Cartoon depiction of cerebral blood volume (CBV) and mean transit time (MTT) calculation using DSC first-pass MRI. The first-pass effect of a contrast agent bolus in brain tissue is monitored using a series of T2*-weighted MR images from the same brain region, which is scanned repeatedly. The susceptibility effect of a paramagnetic (GBCA), or superparamagnetic (USPIO), contrast agent causes decreased signal intensity that can be converted into a contrast agent concentration. This concentration can then be converted into quantitative measurements of CBV proportional to the area under the curve (hatched area) assuming that the longitudinal relaxation time T1 of bulk tissue remains constant during bolus passage. This assumption holds true if the contrast agent is confined to the vascular space and the vascular volume fraction is so small that its contribution to the overall signal intensity may be neglected. (B) Comparison of gadodiamide versus ferumoxytol perfusion imaging in a highly vascular and permeable human glioma (U87 MG cell line) xenograft at 12 T. The curves depicted (B.1 and B.4) show the signal intensity—time course using standard rapid T2*-weighted gradient echo MR sequences during gadodiamide (B.1) and ferumoxytol (B.4) first-pass boli. Regions of interest were defined on the T2-weighted anatomical images (B.2 and B.5) in the tumor (red) and normal-appearing brain tissue (blue). Because of extravasation of the smaller molecular weight gadodiamide from the vasculature, CBV calculations (area under the curve proportional to area of DSC dip) will be inappropriately low. The CBV parametric maps (B.3 and B.6) show the discrepancy between CBV calculations using these two agents; ferumoxytol-based perfusion imaging correctly delineates elevated CBV in this aggressive tumor model. Adapted with permission from Neuwelt et al (2007) and Varallyay et al (2009).
Figure 4
Figure 4
(A) Discordance of rCBV calculated using ferumoxytol versus GBCA enhancement suggestive of pseudoprogression. At the middle of radiation time point (middle row), rCBV with ferumoxytol clearly shows increased blood flow (yellow/green area on rCBV with Fe parametric map marked with small red arrow) in a second posterior frontal lobe lesion (this lesion was not initially radiated after the patient's first resection of a left frontal GBM). After completion of radiation (bottom row), the area of GBCA enhancement is increased; however, DSC-MRI with ferumoxytol (third column) shows decreased rCBV compared with surrounding normal brain. (B) A 19-year-old man with GBM imaged pre- and postoperatively, after standard radiochemotherapy (RCT) and at multiple time points after treatment with bevacizumab. B.1: GBCA-enhanced, T1-weighted MRI before surgery shows a large ring enhancing lesion in the right parietooccipital lobe with mass effect and midline shift; B.2: postoperative image, before initiating RCT. B.3: MRI 1 month after completion of RCT revealed increased area of GBCA enhancement on T1-weighted images (see white arrow) with predominantly low blood volume except for a thin area of high blood volume at the periphery of the Gd- and Fe-rCBV parametric maps (B.4 and B.5). Note that the peripheral rCBV is more prominent (intense green/yellow circle marked by white arrow in image 5; the red area in image 4 is likely artifact) using ferumoxytol versus gadoteridol. Adjuvant temozolomide treatment was continued and bevacizumab treatment was then initiated. B.6–7: GBCA-enhanced, T1-weighted MRIs—after first and second courses of treatment with bevacizumab show significant improvements in mass effect and diminished contrast enhancement. B.8–10: after six treatments with bevacizumab and temozolomide, there is a slight increase in GBCA enhancement, but decreased rCBV on both gadoteridol and ferumoxytol perfusion imaging.
Figure 5
Figure 5
(A) Reprinted with permission from Saleh et al (2007). The USPIO-enhanced MRI of a 54-year-old man with infarction involving the left middle cerebral artery distribution reveals the differential development of USPIO-related signal changes over time. Compared with the nonenhanced, T1-weighted image 5 days after stroke (A.1), T1-weighted images 24 h (A.2), and 48 h (A.3) after USPIO infusion show increasing hyperintense signal enhancement in the periphery of the infarcted parenchyma. Nonenhanced, T2*-weighted image (A.4) displays hyperintense demarcation of the infarcted territory. T2*-weighted images 24 h (A.5) and 48 h (A.6) after USPIO infusion show signal change from hyperintense to hypointense attributable to USPIO perfusion. (B) Ferumoxytol MR angiography at 3 T of the supraaortic arteries in a kidney transplant patient with a glomerular filtration rate (GFR) <60 mL/mins/1.73 m2. Large plaque causes significant luminal narrowing at the origin of the innominate artery (red arrows). A significant stenosis was also observed at the origin of the left subclavian artery (not shown). Carotid bulb shows moderate stenosis on both sides (white arrows). Note the difference in diameter between the left and right carotid as well as the vertebral arteries. The color reproduction of this figure is available on the html full text version of the manuscript.
Figure 6
Figure 6
Adapted with permission from Manninger et al (2005). Patient with ADEM. Axial, T1-weighted images without (1), and with (2) gadolinium (inset, coronal) show faint, subtle enhancement in multiple brain stem lesions. Six days later, significant and more prominent enhancement can be seen at the same sites (3) using ferumoxtran-10 (inset, coronal). Three months later, the lesions no longer enhance on T1-weighted images with gadolinium (4).
Figure 7
Figure 7
(A) Pre- and postferumoxtyol (15 mins, middle row; 24 h, bottom row) enhanced 12 T MRIs after controlled cortical injury in a rat model of traumatic brain injury. At 72 h after injury, there is marked signal loss visualized on T2* (Fe 24 h images); (B) Phagocytic inflammatory cells, seen on H&E (B.1) and stained with CD68 (B.2) and fibronectin (B.3). (C) The inflammatory cells are at least in part derived from peripheral circulating monocytes as illustrated in panels (C.1) and (C.2), which show quantum dot (Invitrogen, Carlsbad, CA)-labeled peripheral mononuclear cells infiltrating around the cortical injury (#1; 800 × magnification) or within blood vessels on the uninjured side (C.2; 800 × magnification).

References

    1. Akeson P, Nordstrom CH, Holtas S. Time-dependency in brain lesion enhancement with gadodiamide injection. Acta Radiol. 1997;38:19–24.
    1. Akhtari M, Bragin A, Cohen M, Moats R, Brenker F, Lynch MD, Vinters HV, Engel J., Jr Functionalized magnetonanoparticles for MRI diagnosis and localization in epilepsy. Epilepsia. 2008;49:1419–1430.
    1. Arbab AS, Yocum GT, Kalish H, Jordan EK, Anderson SA, Khakoo AY, Read EJ, Frank JA. Efficient magnetic cell labeling with protamine sulfate complexed to ferumoxides for cellular MRI. Blood. 2004a;104:1217–1223.
    1. Arbab , Yocum GT, Wilson LB, Parwana A, Jordan EK, Kalish H, Frank JA. Comparison of transfection agents in forming complexes with ferumoxides, cell labeling efficiency, and cellular viability. Mol Imaging. 2004b;3:24–32.
    1. Aronen HJ, Gazit IE, Louis DN, Buchbinder BR, Pardo FS, Weisskoff RM, Harsh GR, Cosgrove GR, Halpern EF, Hochberg FH, et al. Cerebral blood volume maps of gliomas: comparison with tumor grade and histologic findings. Radiology. 1994;191:41–51.
    1. Audebert HJ, Rott MM, Eck T, Haberl RL. Systemic inflammatory response depends on initial stroke severity but is attenuated by successful thrombolysis. Stroke. 2004;35:2128–2133.
    1. Baeten K, Hendriks JJ, Hellings N, Theunissen E, Vanderlocht J, Ryck LD, Gelan J, Stinissen P, Adriaensens P. Visualisation of the kinetics of macrophage infiltration during experimental autoimmune encephalomyelitis by magnetic resonance imaging. J Neuroimmunol. 2008;195:1–6.
    1. Banks WA. Physiology and pathology of the blood-brain barrier: implications for microbial pathogenesis, drug delivery and neurodegenerative disorders. J Neurovirol. 1999;5:538–555.
    1. Bareyre FM. Neuronal repair and replacement in spinal cord injury. J Neurol Sci. 2008;265:63–72.
    1. Barnett PA, Roman-Goldstein S, Ramsey F, McCormick CI, Sexton G, Szumowski J, Neuwelt EA. Differential permeability and quantitative MR imaging of a human lung carcinoma brain xenograft in the nude rat. Am J Pathol. 1995;146:436–449.
    1. Bendszus M, Bartsch A, Stoll G. Is the disruption of the blood-brain barrier a prerequisite for cellular infiltration in autoimmune encephalitis. Brain. 2005;128:E25.
    1. Bitsch A, Schuchardt J, Bunkowski S, Kuhlmann T, Bruck W. Acute axonal injury in multiple sclerosis. Correlation with demyelination and inflammation. Brain. 2000;123 (Pt 6:1174–1183.
    1. Blockley NP, Jiang L, Gardener AG, Ludman CN, Francis ST, Gowland PA. Field strength dependence of R1 and R2* relaxivities of human whole blood to ProHance, Vasovist, and deoxyhemoglobin. Magn Reson Med. 2008;60:1313–1320.
    1. Bourrinet P, Bengele HH, Bonnemain B, Dencausse A, Idee JM, Jacobs PM, Lewis JM. Preclinical safety and pharmacokinetic profile of ferumoxtran-10, an ultrasmall superparamagnetic iron oxide magnetic resonance contrast agent. Invest Radiol. 2006;41:313–324.
    1. Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent MJ. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9:453–461.
    1. Brekke C, Williams SC, Price J, Thorsen F, Modo M. Cellular multiparametric MRI of neural stem cell therapy in a rat glioma model. Neuroimage. 2007;37:769–782.
    1. Brochet B, Deloire MS, Touil T, Anne O, Caille JM, Dousset V, Petry KG. Early macrophage MRI of inflammatory lesions predicts lesion severity and disease development in relapsing EAE. Neuroimage. 2006;32:266–274.
    1. Cameron DG, Bensley EH, Wood P, Grayston V. Treatment of iron deficiency anaemia with saccharated iron oxide given by the intravenous route. Can Med Assoc J. 1951;64:27–30.
    1. Cao Y, Tsien CI, Nagesh V, Junck L, Ten Haken R, Ross BD, Chenevert TL, Lawrence TS. Survival prediction in high-grade gliomas by MRI perfusion before and during early stage of RT [corrected] Int J Radiat Oncol Biol Phys. 2006;64:876–885.
    1. Cappendijk VC, Kessels AG, Heeneman S, Cleutjens KB, Schurink GW, Welten RJ, Mess WH, van Suylen RJ, Leiner T, Daemen MJ, van Engelshoven JM, Kooi ME. Comparison of lipid-rich necrotic core size in symptomatic and asymptomatic carotid atherosclerotic plaque: initial results. J Magn Reson Imaging. 2008;27:1356–1361.
    1. Cha S, Knopp EA, Johnson G, Wetzel SG, Litt AW, Zagzag D. Intracranial mass lesions: dynamic contrast-enhanced susceptibility-weighted echo-planar perfusion MR imaging. Radiology. 2002;223:11–29.
    1. Chang KH, Ra DG, Han MH, Cha SH, Kim HD, Han MC.1994Contrast enhancement of brain tumors at different MR field strengths: comparison of 0.5 T and 2.0 T AJNR Am J Neuroradiol 151413–1419.discussion 1420–1413
    1. Chenevert TL, Pipe JG, Williams DM, Brunberg JA. Quantitative measurement of tissue perfusion and diffusion in vivo. Magn Reson Med. 1991;17:197–212.
    1. Chertok B, Moffat BA, David AE, Yu F, Bergemann C, Ross BD, Yang VC. Iron oxide nanoparticles as a drug delivery vehicle for MRI monitored magnetic targeting of brain tumors. Biomaterials. 2008;29:487–496.
    1. Claes A, Gambarota G, Hamans B, van Tellingen O, Wesseling P, Maass C, Heerschap A, Leenders W. Magnetic resonance imaging-based detection of glial brain tumors in mice after antiangiogenic treatment. Int J Cancer. 2008;122:1981–1986.
    1. Corot C, Petry KG, Trivedi R, Saleh A, Jonkmanns C, Le Bas JF, Blezer E, Rausch M, Brochet B, Foster-Gareau P, Baleriaux D, Gaillard S, Dousset V. Macrophage imaging in central nervous system and in carotid atherosclerotic plaque using ultrasmall superparamagnetic iron oxide in magnetic resonance imaging. Invest Radiol. 2004;39:619–625.
    1. Corot C, Robert P, Idee JM, Port M. Recent advances in iron oxide nanocrystal technology for medical imaging. Adv Drug Deliv Rev. 2006;58:1471–1504.
    1. Daldrup-Link HE, Rudelius M, Oostendorp RA, Settles M, Piontek G, Metz S, Rosenbrock H, Keller U, Heinzmann U, Rummeny EJ, Schlegel J, Link TM. Targeting of hematopoietic progenitor cells with MR contrast agents. Radiology. 2003;228:760–767.
    1. del Zoppo G, Ginis I, Hallenbeck JM, Iadecola C, Wang X, Feuerstein GZ. Inflammation and stroke: putative role for cytokines, adhesion molecules and iNOS in brain response to ischemia. Brain Pathol. 2000;10:95–112.
    1. Denes A, Vidyasagar R, Feng J, Narvainen J, McColl BW, Kauppinen RA, Allan SM. Proliferating resident microglia after focal cerebral ischaemia in mice. J Cereb Blood Flow Metab. 2007;27:1941–1953.
    1. Donahue KM, Krouwer HG, Rand SD, Pathak AP, Marszalkowski CS, Censky SC, Prost RW. Utility of simultaneously acquired gradient-echo and spin-echo cerebral blood volume and morphology maps in brain tumor patients. Magn Reson Med. 2000;43:845–853.
    1. Dousset V, Brochet B, Deloire MS, Lagoarde L, Barroso B, Caille JM, Petry KG. MR imaging of relapsing multiple sclerosis patients using ultra-small-particle iron oxide and compared with gadolinium. AJNR Am J Neuroradiol. 2006;27:1000–1005.
    1. Enochs WS, Harsh G, Hochberg F, Weissleder R. Improved delineation of human brain tumors on MR images using a long-circulating, superparamagnetic iron oxide agent. J Magn Reson Imaging. 1999;9:228–232.
    1. Essig M, Weber MA, von Tengg-Kobligk H, Knopp MV, Yuh WT, Giesel FL. Contrast-enhanced magnetic resonance imaging of central nervous system tumors: agents, mechanisms, and applications. Top Magn Reson Imaging. 2006;17:89–106.
    1. Fleige G, Nolte C, Synowitz M, Seeberger F, Kettenmann H, Zimmer C. Magnetic labeling of activated microglia in experimental gliomas. Neoplasia. 2001;3:489–499.
    1. Floris S, Blezer EL, Schreibelt G, Dopp E, van der Pol SM, Schadee-Eestermans IL, Nicolay K, Dijkstra CD, de Vries HE. Blood-brain barrier permeability and monocyte infiltration in experimental allergic encephalomyelitis: a quantitative MRI study. Brain. 2004;127:616–627.
    1. Gore JC, Majumdar S. Measurement of tissue blood flow using intravascular relaxation agents and magnetic resonance imaging. Magn Reson Med. 1990;14:242–248.
    1. Groothuis DR, Vriesendorp FJ, Kupfer B, Warnke PC, Lapin GD, Kuruvilla A, Vick NA, Mikhael MA, Patlak CS. Quantitative measurements of capillary transport in human brain tumors by computed tomography. Ann Neurol. 1991;30:581–588.
    1. Harpur ES, Worah D, Hals PA, Holtz E, Furuhama K, Nomura H. Preclinical safety assessment and pharmacokinetics of gadodiamide injection, a new magnetic resonance imaging contrast agent. Invest Radiol. 1993;28 (Suppl 1:S28–S43.
    1. Hartman KB, Wilson LJ, Rosenblum MG. Detecting and treating cancer with nanotechnology. Mol Diagn Ther. 2008;12:1–14.
    1. Henning EC, Ruetzler CA, Gaudinski MR, Hu TC, Latour LL, Hallenbeck JM, Warach S. Feridex preloading permits tracking of CNS-resident macrophages after transient middle cerebral artery occlusion. J Cereb Blood Flow Metab. 2009;29:1229–139.
    1. Herschman HR. Molecular imaging: looking at problems, seeing solutions. Science. 2003;302:605–608.
    1. Hoehn M, Wiedermann D, Justicia C, Ramos-Cabrer P, Kruttwig K, Farr T, Himmelreich U. Cell tracking using magnetic resonance imaging. J Physiol. 2007;584:25–30.
    1. Hunt MA, Bago AG, Neuwelt EA. Single-dose contrast agent for intraoperative MR imaging of intrinsic brain tumors by using ferumoxtran-10. AJNR Am J Neuroradiol. 2005;26:1084–1088.
    1. Idiyatullin D, Corum C, Park JY, Garwood M. Fast and quiet MRI using a swept radiofrequency. J Magn Reson. 2006;181:342–349.
    1. Jander S, Schroeter M, Saleh A. Imaging inflammation in acute brain ischemia. Stroke. 2007;38:642–645.
    1. Juhasz C, Chugani DC, Padhye UN, Muzik O, Shah A, Asano E, Mangner TJ, Chakraborty PK, Sood S, Chugani HT. Evaluation with alpha-[11C]methyl-L-tryptophan positron emission tomography for reoperation after failed epilepsy surgery. Epilepsia. 2004;45:124–130.
    1. Ke Y, Ming Qian Z. Iron misregulation in the brain: a primary cause of neurodegenerative disorders. Lancet Neurol. 2003;2:246–253.
    1. Khoo VS, Joon DL. New developments in MRI for target volume delineation in radiotherapy. Br J Radiol. 2006;79 (Spec No 1:S2–S15.
    1. Knopp EA, Cha S, Johnson G, Mazumdar A, Golfinos JG, Zagzag D, Miller DC, Kelly PJ, Kricheff II. Glial neoplasms: dynamic contrast-enhanced T2*-weighted MR imaging. Radiology. 1999;211:791–798.
    1. Korosoglou G, Tang L, Kedziorek D, Cosby K, Gilson WD, Vonken EJ, Schar M, Sosnovik D, Kraitchman DL, Weiss RG, Weissleder R, Stuber M. Positive contrast MR-lymphography using inversion recovery with ON-resonant water suppression (IRON) J Magn Reson Imaging. 2008;27:1175–1180.
    1. Kraemer DF, Fortin D, Doolittle ND, Neuwelt EA.2001Association of total dose intensity of chemotherapy in primary central nervous system lymphoma (human non-acquired immunodeficiency syndrome) and survival Neurosurgery 481033–1040.discussion 1040–1031
    1. Kremer S, Grand S, Remy C, Esteve F, Lefournier V, Pasquier B, Hoffmann D, Benabid AL, Le Bas JF. Cerebral blood volume mapping by MR imaging in the initial evaluation of brain tumors. J Neuroradiol. 2002;29:105–113.
    1. Kremer S, Pinel S, Vedrine PO, Bressenot A, Robert P, Bracard S, Plenat F. Ferumoxtran-10 enhancement in orthotopic xenograft models of human brain tumors: an indirect marker of tumor proliferation. J Neurooncol. 2007;83:111–119.
    1. Kroll RA, Neuwelt EA.1998Outwitting the blood-brain barrier for therapeutic purposes: osmotic opening and other means Neurosurgery 421083–1099.discussion 1099–1100
    1. Kulbatski I, Mothe AJ, Nomura H, Tator CH. Endogenous and exogenous CNS derived stem/progenitor cell approaches for neurotrauma. Curr Drug Targets. 2005;6:111–126.
    1. Landry R, Jacobs PM, Davis R, Shenouda M, Bolton WK. Pharmacokinetic study of ferumoxytol: a new iron replacement therapy in normal subjects and hemodialysis patients. Am J Nephrol. 2005;25:400–410.
    1. Laurent S, Forge D, Port M, Roch A, Robic C, Vander Elst L, Muller RN. Magnetic iron oxide nanoparticles: synthesis, stabilization, vectorization, physicochemical characterizations, and biological applications. Chem Rev. 2008;108:2064–2110.
    1. Lev MH, Ozsunar Y, Henson JW, Rasheed AA, Barest GD, Harsh GR, IV, Fitzek MM, Chiocca EA, Rabinov JD, Csavoy AN, Rosen BR, Hochberg FH, Schaefer PW, Gonzalez RG. Glial tumor grading and outcome prediction using dynamic spin-echo MR susceptibility mapping compared with conventional contrast-enhanced MR: confounding effect of elevated rCBV of oligodendrogliomas (corrected) AJNR Am J Neuroradiol. 2004;25:214–221.
    1. Long DM. Capillary ultrastructure in human metastatic brain tumors. J Neurosurg. 1979;51:53–58.
    1. Lyons SA, O'Neal J, Sontheimer H. Chlorotoxin, a scorpion-derived peptide, specifically binds to gliomas and tumors of neuroectodermal origin. Glia. 2002;39:162–173.
    1. Macdonald DR, Cascino TL, Schold SC, Jr, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277–1280.
    1. Maier-Hauff K, Rothe R, Scholz R, Gneveckow U, Wust P, Thiesen B, Feussner A, von Deimling A, Waldoefner N, Felix R, Jordan A. Intracranial thermotherapy using magnetic nanoparticles combined with external beam radiotherapy: results of a feasibility study on patients with glioblastoma multiforme. J Neurooncol. 2007;81:53–60.
    1. Manninger SP, Muldoon LL, Nesbit G, Murillo T, Jacobs PM, Neuwelt EA. An exploratory study of ferumoxtran-10 nanoparticles as a blood-brain barrier imaging agent targeting phagocytic cells in CNS inflammatory lesions. AJNR Am J Neuroradiol. 2005;26:2290–2300.
    1. Matuszewski L, Persigehl T, Wall A, Schwindt W, Tombach B, Fobker M, Poremba C, Ebert W, Heindel W, Bremer C. Cell tagging with clinically approved iron oxides: feasibility and effect of lipofection, particle size, and surface coating on labeling efficiency. Radiology. 2005;235:155–161.
    1. McAteer MA, Sibson NR, von Zur Muhlen C, Schneider JE, Lowe , Warrick N, Channon KM, Anthony DC, Choudhury RP. In vivo magnetic resonance imaging of acute brain inflammation using microparticles of iron oxide. Nat Med. 2007;13:1253–1258.
    1. McDonald RP, Horsburgh KJ, Graham DI, Nicoll JA. Mitochondrial DNA deletions in acute brain injury. Neuroreport. 1999;10:1875–1878.
    1. Metz S, Bonaterra G, Rudelius M, Settles M, Rummeny EJ, Daldrup-Link HE. Capacity of human monocytes to phagocytose approved iron oxide MR contrast agents in vitro. Eur Radiol. 2004;14:1851–1858.
    1. Miki Y, Grossman RI, Udupa JK, Samarasekera S, van Buchem MA, Cooney BS, Pollack SN, Kolson DL, Constantinescu C, Polansky M, Mannon LJ. Computer-assisted quantitation of enhancing lesions in multiple sclerosis: correlation with clinical classification. AJNR Am J Neuroradiol. 1997;18:705–710.
    1. Miki Y, Grossman RI, Udupa JK, van Buchem MA, Wei L, Phillips MD, Patel U, McGowan JC, Kolson DL. Differences between relapsing-remitting and chronic progressive multiple sclerosis as determined with quantitative MR imaging. Radiology. 1999;210:769–774.
    1. Modo M, Hoehn M, Bulte JW. Cellular MR imaging. Mol Imaging. 2005;4:143–164.
    1. Moseley ME, Kucharczyk J, Mintorovitch J, Cohen Y, Kurhanewicz J, Derugin N, Asgari H, Norman D. Diffusion-weighted MR imaging of acute stroke: correlation with T2-weighted and magnetic susceptibility-enhanced MR imaging in cats. AJNR Am J Neuroradiol. 1990;11:423–429.
    1. Mulder WJ, Douma K, Koning GA, van Zandvoort MA, Lutgens E, Daemen MJ, Nicolay K, Strijkers GJ. Liposome-enhanced MRI of neointimal lesions in the ApoE-KO mouse. Magn Reson Med. 2006;55:1170–1174.
    1. Muldoon LL, Sandor M, Pinkston KE, Neuwelt EA.2005Imaging, distribution, and toxicity of superparamagnetic iron oxide magnetic resonance nanoparticles in the rat brain and intracerebral tumor Neurosurgery 57785–796.discussion 785–796
    1. Muldoon LL, Tratnyek PG, Jacobs PM, Doolittle ND, Christoforidis GA, Frank JA, Lindau M, Lockman PR, Manninger SP, Qiang Y, Spence AM, Stupp SI, Zhang M, Neuwelt EA. Imaging and nanomedicine for diagnosis and therapy in the central nervous system: report of the eleventh annual Blood-Brain Barrier Disruption Consortium meeting. AJNR Am J Neuroradiol. 2006;27:715–721.
    1. Muldoon LL, Varallyay P, Kraemer DF, Kiwic G, Pinkston K, Walker-Rosenfeld SL, Neuwelt EA. Trafficking of superparamagnetic iron oxide particles (Combidex) from brain to lymph nodes in the rat. Neuropathol Appl Neurobiol. 2004;30:70–79.
    1. Neuwelt EA, Hamilton BE, Varallyay CG, Rooney WR, Edelman RD, Jacobs PM, Watnick SG. Ultrasmall superparamagnetic iron oxides (USPIOs): a future alternative magnetic resonance (MR) contrast agent for patients at risk for nephrogenic systemic fibrosis (NSF) Kidney Int. 2008;75:465–474.
    1. Neuwelt EA, Varallyay CG, Manninger S, Solymosi D, Haluska M, Hunt MA, Nesbit G, Stevens A, Jerosch-Herold M, Jacobs PM, Hoffman JM.2007The potential of ferumoxytol nanoparticle magnetic resonance imaging, perfusion, and angiography in central nervous system malignancy: a pilot study Neurosurgery 60601–611.discussion 611–602
    1. Neuwelt EA, Varallyay P, Bago AG, Muldoon LL, Nesbit G, Nixon R. Imaging of iron oxide nanoparticles by MR and light microscopy in patients with malignant brain tumours. Neuropathol Appl Neurobiol. 2004;30:456–471.
    1. Neuwelt EA, Weissleder R, Nilaver G, Kroll RA, Roman-Goldstein S, Szumowski J, Pagel MA, Jones RS, Remsen LG, McCormick CI, et al. Delivery of virus-sized iron oxide particles to rodent CNS neurons. Neurosurgery. 1994;34:777–784.
    1. Nighoghossian N, Wiart M, Cakmak S, Berthezene Y, Derex L, Cho TH, Nemoz C, Chapuis F, Tisserand GL, Pialat JB, Trouillas P, Froment JC, Hermier M. Inflammatory response after ischemic stroke: a USPIO-enhanced MRI study in patients. Stroke. 2007;38:303–307.
    1. Oh J, Henry RG, Pirzkall A, Lu Y, Li X, Catalaa I, Chang S, Dillon WP, Nelson SJ. Survival analysis in patients with glioblastoma multiforme: predictive value of choline-to-N-acetylaspartate index, apparent diffusion coefficient, and relative cerebral blood volume. J Magn Reson Imaging. 2004;19:546–554.
    1. Ostergaard L. Principles of cerebral perfusion imaging by bolus tracking. J Magn Reson Imaging. 2005;22:710–717.
    1. Oude Engberink RD, van der Pol SM, Dopp EA, de Vries HE, Blezer EL. Comparison of SPIO and USPIO for in vitro labeling of human monocytes: MR detection and cell function. Radiology. 2007;243:467–474.
    1. Rausch M, Hiestand P, Baumann D, Cannet C, Rudin M. MRI-based monitoring of inflammation and tissue damage in acute and chronic relapsing EAE. Magn Reson Med. 2003;50:309–314.
    1. Rausch M, Hiestand P, Foster CA, Baumann DR, Cannet C, Rudin M. Predictability of FTY720 efficacy in experimental autoimmune encephalomyelitis by in vivo macrophage tracking: clinical implications for ultrasmall superparamagnetic iron oxide-enhanced magnetic resonance imaging. J Magn Reson Imaging. 2004;20:16–24.
    1. Rausch M, Sauter A, Frohlich J, Neubacher U, Radu EW, Rudin M. Dynamic patterns of USPIO enhancement can be observed in macrophages after ischemic brain damage. Magn Reson Med. 2001;46:1018–1022.
    1. Robinson SP, Howe FA, Griffiths JR, Ryan AJ, Waterton JC. Susceptibility contrast magnetic resonance imaging determination of fractional tumor blood volume: a noninvasive imaging biomarker of response to the vascular disrupting agent ZD6126. Int J Radiat Oncol Biol Phys. 2007;69:872–879.
    1. Rohrer M, Bauer H, Mintorovitch J, Requardt M, Weinmann HJ. Comparison of magnetic properties of MRI contrast media solutions at different magnetic field strengths. Invest Radiol. 2005;40:715–724.
    1. Rooney WD, Johnson G, Li X, Cohen ER, Kim SG, Ugurbil K, Springer CS., Jr Magnetic field and tissue dependencies of human brain longitudinal 1H2O relaxation in vivo. Magn Reson Med. 2007;57:308–318.
    1. Ros PR, Freeny PC, Harms SE, Seltzer SE, Davis PL, Chan TW, Stillman AE, Muroff LR, Runge VM, Nissenbaum MA, et al. Hepatic MR imaging with ferumoxides: a multicenter clinical trial of the safety and efficacy in the detection of focal hepatic lesions. Radiology. 1995;196:481–488.
    1. Rose SE, Janke AL, Strudwick MW, McMahon KL, Chalk JB, Snyder P, De zubicaray GI. Assessment of dynamic susceptibility contrast cerebral blood flow response to amphetamine challenge: a human pharmacological magnetic resonance imaging study at 1.5 and 4 T. Magn Reson Med. 2006;55:9–15.
    1. Rosen BR, Belliveau JW, Vevea JM, Brady TJ. Perfusion imaging with NMR contrast agents. Magn Reson Med. 1990;14:249–265.
    1. Ruehm SG, Corot C, Vogt P, Cristina H, Debatin JF. Ultrasmall superparamagnetic iron oxide-enhanced MR imaging of atherosclerotic plaque in hyperlipidemic rabbits. Acad Radiol. 2002;9 (Suppl 1:S143–S144.
    1. Ruiz-Cabello J, Walczak P, Kedziorek DA, Chacko VP, Schmieder AH, Wickline SA, Lanza GM, Bulte JW. In vivo ‘hot spot' MR imaging of neural stem cells using fluorinated nanoparticles. Magn Reson Med. 2008;60:1506–1511.
    1. Saleh A, Schroeter M, Jonkmanns C, Hartung HP, Modder U, Jander S. In vivo MRI of brain inflammation in human ischaemic stroke. Brain. 2004a;127:1670–1677.
    1. Saleh A, Schroeter M, Ringelstein A, Hartung HP, Siebler M, Modder U, Jander S. Iron oxide particle-enhanced MRI suggests variability of brain inflammation at early stages after ischemic stroke. Stroke. 2007;38:2733–2737.
    1. Saleh A, Wiedermann D, Schroeter M, Jonkmanns C, Jander S, Hoehn M. Central nervous system inflammatory response after cerebral infarction as detected by magnetic resonance imaging. NMR Biomed. 2004b;17:163–169.
    1. Schaefer PW, Barak ER, Kamalian S, Gharai LR, Schwamm L, Gonzalez RG, Lev MH. Quantitative assessment of core/penumbra mismatch in acute stroke. CT and MR perfusion imaging are strongly correlated when sufficient brain volume is imaged. Stroke. 2008;39:2986–2992.
    1. Schilling M, Besselmann M, Leonhard C, Mueller M, Ringelstein EB, Kiefer R. Microglial activation precedes and predominates over macrophage infiltration in transient focal cerebral ischemia: a study in green fluorescent protein transgenic bone marrow chimeric mice. Exp Neurol. 2003;183:25–33.
    1. Schroeter M, Franke C, Stoll G, Hoehn M. Dynamic changes of magnetic resonance imaging abnormalities in relation to inflammation and glial responses after photothrombotic cerebral infarction in the rat brain. Acta Neuropathol. 2001;101:114–122.
    1. Schroeter M, Jander S, Huitinga I, Witte OW, Stoll G. Phagocytic response in photochemically induced infarction of rat cerebral cortex. The role of resident microglia. Stroke. 1997;28:382–386.
    1. Soroceanu L, Gillespie Y, Khazaeli MB, Sontheimer H. Use of chlorotoxin for targeting of primary brain tumors. Cancer Res. 1998;58:4871–4879.
    1. Spinowitz BS, Schwenk MH, Jacobs PM, Bolton WK, Kaplan MR, Charytan C, Galler M. The safety and efficacy of ferumoxytol therapy in anemic chronic kidney disease patients. Kidney Int. 2005;68:1801–1807.
    1. Stoll G, Jander S, Schroeter M. Inflammation and glial responses in ischemic brain lesions. Prog Neurobiol. 1998;56:149–171.
    1. Stroke Recommendations on stroke prevention, diagnosis, and therapy. Report of the WHO Task Force on Stroke and Other Cerebrovascular Disorders. Stroke. 1989;20:1407–1431.
    1. Stuber M, Gilson WD, Schar M, Kedziorek DA, Hofmann LV, Shah S, Vonken EJ, Bulte JW, Kraitchman DL. Positive contrast visualization of iron oxide-labeled stem cells using inversion-recovery with ON-resonant water suppression (IRON) Magn Reson Med. 2007;58:1072–1077.
    1. Sugahara T, Korogi Y, Kochi M, Ikushima I, Hirai T, Okuda T, Shigematsu Y, Liang L, Ge Y, Ushio Y, Takahashi M. Correlation of MR imaging-determined cerebral blood volume maps with histologic and angiographic determination of vascularity of gliomas. AJR Am J Roentgenol. 1998;171:1479–1486.
    1. Sykova E, Jendelova P. In vivo tracking of stem cells in brain and spinal cord injury. Prog Brain Res. 2007;161:367–383.
    1. Tanaka R, Komine-Kobayashi M, Mochizuki H, Yamada M, Furuya T, Migita M, Shimada T, Mizuno Y, Urabe T. Migration of enhanced green fluorescent protein expressing bone marrow-derived microglia/macrophage into the mouse brain following permanent focal ischemia. Neuroscience. 2003;117:531–539.
    1. Tang T, Howarth SP, Miller SR, Trivedi R, Graves MJ, King-Im JU, Li ZY, Brown AP, Kirkpatrick PJ, Gaunt ME, Gillard JH. Assessment of inflammatory burden contralateral to the symptomatic carotid stenosis using high-resolution ultrasmall, superparamagnetic iron oxide-enhanced MRI. Stroke. 2006;37:2266–2270.
    1. Thorek DL, Chen AK, Czupryna J, Tsourkas A. Superparamagnetic iron oxide nanoparticle probes for molecular imaging. Ann Biomed Eng. 2006;34:23–38.
    1. Uematsu H, Maeda M. Double-echo perfusion-weighted MR imaging: basic concepts and application in brain tumors for the assessment of tumor blood volume and vascular permeability. Eur Radiol. 2006;16:180–186.
    1. van Landeghem FK, Maier-Hauff K, Jordan A, Hoffmann KT, Gneveckow U, Scholz R, Thiesen B, Bruck W, von Deimling A. Post-mortem studies in glioblastoma patients treated with thermotherapy using magnetic nanoparticles. Biomaterials. 2009;30:52–57.
    1. Varallyay CG, Muldoon LL, Gahramanov S, Wu YJ, Goodman JA, Li X, Pike MM, Neuwelt EA. Dynamic MRI using iron oxide nanoparticles to assess early vascular effects of antiangiogenic versus corticosteroid treatment in a glioma model. J Cereb Blood Flow Metab. 2009;29:853–860.
    1. Varallyay P, Nesbit G, Muldoon LL, Nixon RR, Delashaw J, Cohen JI, Petrillo A, Rink D, Neuwelt EA. Comparison of two superparamagnetic viral-sized iron oxide particles ferumoxides and ferumoxtran-10 with a gadolinium chelate in imaging intracranial tumors. AJNR Am J Neuroradiol. 2002;23:510–519.
    1. Vellinga MM, Oude Engberink RD, Seewann A, Pouwels PJ, Wattjes MP, van der Pol SM, Pering C, Polman CH, de Vries HE, Geurts JJ, Barkhof F. Pluriformity of inflammation in multiple sclerosis shown by ultra-small iron oxide particle enhancement. Brain. 2008;131:800–807.
    1. Verdijk P, Scheenen TW, Lesterhuis WJ, Gambarota G, Veltien AA, Walczak P, Scharenborg NM, Bulte JW, Punt CJ, Heerschap A, Figdor CG, de Vries IJ. Sensitivity of magnetic resonance imaging of dendritic cells for in vivo tracking of cellular cancer vaccines. Int J Cancer. 2007;120:978–984.
    1. Villringer A, Rosen BR, Belliveau JW, Ackerman JL, Lauffer RB, Buxton RB, Chao YS, Wedeen VJ, Brady TJ. Dynamic imaging with lanthanide chelates in normal brain: contrast due to magnetic susceptibility effects. Magn Reson Med. 1988;6:164–174.
    1. Weber R, Ramos-Cabrer P, Hoehn M. Present status of magnetic resonance imaging and spectroscopy in animal stroke models. J Cereb Blood Flow Metab. 2006;26:591–604.
    1. Weissleder R, Elizondo G, Wittenberg J, Rabito CA, Bengele HH, Josephson L. Ultrasmall superparamagnetic iron oxide: characterization of a new class of contrast agents for MR imaging. Radiology. 1990;175:489–493.
    1. Willmore LJ, Sypert GW, Munson JB. Recurrent seizures induced by cortical iron injection: a model of posttraumatic epilepsy. Ann Neurol. 1978;4:329–336.
    1. Wu X, Hu J, Zhou L, Mao Y, Yang B, Gao L, Xie R, Xu F, Zhang D, Liu J, Zhu J. In vivo tracking of superparamagnetic iron oxide nanoparticle-labeled mesenchymal stem cell tropism to malignant gliomas using magnetic resonance imaging. Laboratory investigation. J Neurosurg. 2008;108:320–329.
    1. Wu YJ, Muldoon LL, Varallyay C, Markwardt S, Jones RE, Neuwelt EA. In vivo leukocyte labeling with intravenous ferumoxides/protamine sulfate complex and in vitro characterization for cellular magnetic resonance imaging. Am J Physiol Cell Physiol. 2007;293:C1698–C1708.
    1. Yuh WT, Crain MR, Loes DJ, Greene GM, Ryals TJ, Sato Y. MR imaging of cerebral ischemia: findings in the first 24 hours. AJNR Am J Neuroradiol. 1991;12:621–629.

Source: PubMed

3
Iratkozz fel