Targeted iron oxide particles for in vivo magnetic resonance detection of atherosclerotic lesions with antibodies directed to oxidation-specific epitopes

Karen C Briley-Saebo, Young Seok Cho, Peter X Shaw, Sung Kee Ryu, Venkatesh Mani, Stephen Dickson, Ehsan Izadmehr, Simone Green, Zahi A Fayad, Sotirios Tsimikas, Karen C Briley-Saebo, Young Seok Cho, Peter X Shaw, Sung Kee Ryu, Venkatesh Mani, Stephen Dickson, Ehsan Izadmehr, Simone Green, Zahi A Fayad, Sotirios Tsimikas

Abstract

Objectives: The aim of this study was to determine whether iron oxide particles targeted to oxidation-specific epitopes image atherosclerotic lesions.

Background: Oxidized low-density lipoprotein plays a major role in atherosclerotic plaque progression and destabilization. Prior studies indicate that gadolinium micelles labeled with oxidation-specific antibodies allow for in vivo detection of vulnerable plaques with magnetic resonance imaging (MRI). However, issues related to biotransformation/retention of gadolinium might limit clinical translation. Iron oxides are recognized as safe and effective contrast agents for MRI. Because the efficacy of passively targeted iron particles remains variable, it was hypothesized that iron particles targeted to oxidation-specific epitopes might increase the utility of this platform.

Methods: Lipid-coated ultra-small superparamagnetic iron particles (LUSPIOs) (<20 nm) and superparamagnetic iron particles (<40 nm) were conjugated with antibodies targeted to either malondialdehyde-lysine or oxidized phospholipid epitopes. All formulations were characterized, and their in vivo efficacy evaluated in apolipoprotein E deficient mice 24 h after bolus administration of a 3.9-mg Fe/kg dose with MRI. In vivo imaging data were correlated with the presence of oxidation-specific epitopes with immunohistochemistry.

Results: MRI of atherosclerotic lesions, as manifested by signal loss, was observed after administration of targeted LUSPIOs. Immunohistochemistry confirmed the presence of malondialdehyde-epitopes and iron particles. Limited signal attenuation was observed for untargeted LUSPIOs. Additionally, no significant arterial wall uptake was observed for targeted or untargeted lipid-coated superparamagnetic iron oxide particles, due to their limited ability to penetrate the vessel wall.

Conclusions: This study demonstrates that LUSPIOs targeted to oxidation-specific epitopes image atherosclerotic lesions and suggests a clinically translatable platform for the detection of atherosclerotic plaque.

Copyright © 2011 American College of Cardiology Foundation. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1. Diagram
Figure 1. Diagram
Schematic diagram of the lipid-coated iron oxide particles formed.
Figure 2. TEM
Figure 2. TEM
Transmission electron microscopy (TEM) and negative TEM of untargeted lipid-coated ultrasmall superparamagnetic iron oxide particle (LUSPIO) and lipid-coated superparamagnetic iron oxide particle (LSPIO) formations. The red asterisks indicate the iron core. The red arrow points to the lipid layer associated with the particles.
Figure 3. Nuclear Magnetic Relaxation Dispersion Profiles
Figure 3. Nuclear Magnetic Relaxation Dispersion Profiles
The longitudinal (r1) and transverse (r2) nuclear magnetic relaxation dispersion profiles of untargeted LSPIOs and LUSPIOs. Abbreviations as in Figure 2.
Figure 4. Macrophage Uptake of Iron Oxide…
Figure 4. Macrophage Uptake of Iron Oxide Particles
In vitro J774A.1 macrophage uptake of untargeted lipid-coated iron particles (negative control), targeted lipid-coated iron particles, and Feridex (Berlex, Inc.) (positive control). All cells were incubated with 1 mmol/l Fe for 24 h. Both Perl’s staining for iron oxide uptake and confocal microscopy (rhodamine-labeled LUSPIOs/LSPIOs) were performed. In addition, iron oxide particle uptake was determined with inductively coupled plasma mass spectrometry and reported in pg Fe/cell. Abbreviations as in Figure 2.
Figure 5. LUSPIO MRI of ApoE−/− Mice
Figure 5. LUSPIO MRI of ApoE−/− Mice
(A) Representative in vivo LUSPIO magnetic resonance images obtained before (PRE) and 24 h after (POST) administration of a 3.9-mg Fe/kg dose in apolipoprotein E deficient (apoE−/−) mice. The yellow arrows indicate the position of the lumen associated with the abdominal aorta. All gradient echo (GRE) images shown were obtained with an echo time of 7 ms. Corresponding white marker gradient echo acquisition for superparamagnetic particles with positive contrast (GRASP) images are also shown. The apoE−/− mice were administered free excess MDA2 antibody at the time of MDA2-labeled LUSPIO injection, for the competitive inhibition results. Matched histology sections show iron deposition (blue) within foam cells associated with the arterial wall. Relative percentage change in the R2* values associated with the arterial wall after administration of the LUSPIO formulations are also shown (B). The error bars reflect the SD associated with mean value obtained for mice receiving untargeted LUSPIOs and antibody-labeled LUSPIOs. MRI = magnetic resonance imaging; other abbreviations as in Figures 2 and 4.
Figure 6. Ultrasmall Iron Oxide Particle MRI…
Figure 6. Ultrasmall Iron Oxide Particle MRI of ApoE−/− Mice
Representative in vivo LSPIO magnetic resonance images obtained before and 24 h after administration of a 3.9-mg Fe/kg dose in apoE−/− mice. Only the untargeted and E06 formulations are shown in A. The yellow arrows indicate the position of the lumen associated with the abdominal aorta. The GRE images shown were obtained with an echo time of 7 ms. Corresponding white marker GRASP images are also shown. Matched histology sections show limited iron deposition (blue) within foam cells associated with the arterial wall. Relative percentage change in the R2* values associated with the arterial wall after administration of the LUSPIO formulations are also shown (B). The error bars reflect the SD associated with mean value obtained for mice receiving untargeted and antibody-labeled LSPIOs. Abbreviations as in Figures 2, 4, and 5.
Figure 7. Immunohistochemistry
Figure 7. Immunohistochemistry
Immunohistochemistry (A) and confocal microscopy (B) of MDA2-labeled LUSPIOs in the arterial wall of apoE−/− and wild-type mice. The arterial wall was stained for MDA-lysine (MDA3 staining), macrophages (Mac3), and iron oxide (Perl’s) deposition. Confocal microscopy with a multi-channel laser so that colocalization between the cell nuclei (blue), macrophages (green), and MDA2-labeled LUSPIOs (red) could be performed. Abbreviations as in Figures 2, 4, and 5.

References

    1. Virmani R, Burke AP, Farb A, Kolodgie FD. Pathology of the vulnerable plaque. J Am Coll Cardiol. 2006;47:C13–8.
    1. Waxman S, Ishibashi F, Muller JE. Detection and treatment of vulnerable plaques and vulnerable patients: novel approaches to prevention of coronary events. Circulation. 2006;114:2390–411.
    1. Tsimikas S, Shortal BP, Witztum JL, Palinski W. In vivo uptake of radiolabeled MDA2, an oxidation-specific monoclonal antibody, provides an accurate measure of atherosclerotic lesions rich in oxidized LDL and is highly sensitive to their regression. Arterioscler Thromb Vasc Biol. 2000;20:689–97.
    1. Hartvigsen K, Chou MY, Hansen LF, et al. The role of innate immunity in atherogenesis. J Lipid Res. 2009;50 (Suppl):S388–93.
    1. Tsimikas S. Oxidative biomarkers in the diagnosis and prognosis of cardiovascular disease. Am J Cardiol. 2006;98:9P–17P.
    1. Tsimikas S, Aikawa M, Miller FJ, Jr, et al. Increased plasma oxidized phospholipid:apolipoprotein B-100 ratio with concomitant depletion of oxidized phospholipids from atherosclerotic lesions after dietary lipid-lowering: a potential biomarker of early atherosclerosis regression. Arterioscler Thromb Vasc Biol. 2007;27:175–81.
    1. Tsimikas S, Brilakis ES, Miller ER, et al. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N Engl J Med. 2005;353:46–57.
    1. Tsimikas S, Mallat Z, Talmud PJ, et al. Oxidation-specific biomarkers, lipoprotein(a), and risk of fatal and nonfatal coronary events. J Am Coll Cardiol. 2010;56:946–55.
    1. Cai J, Hatsukami TS, Ferguson MS, et al. In vivo quantitative measurement of intact fibrous cap and lipid-rich necrotic core size in atherosclerotic carotid plaque: comparison of high-resolution, contrast-enhanced magnetic resonance imaging and histology. Circulation. 2005;112:3437–44.
    1. Desai MY, Lima JA. Imaging of atherosclerosis using magnetic resonance: state of the art and future directions. Curr Atheroscler Rep. 2006;8:131–9.
    1. Briley-Saebo KC, Shaw PX, Mulder WJ, et al. Targeted molecular probes for imaging atherosclerotic lesions with magnetic resonance using antibodies that recognize oxidation-specific epitopes. Circulation. 2008;117:3206–15.
    1. Ide M, Kuwamura M, Kotani T, Sawamoto O, Yamate J. Effects of gadolinium chloride (GdCl(3)) on the appearance of macrophage populations and fibrogenesis in thioacetamide-induced rat hepatic lesions. J Comp Pathol. 2005;133:92–102.
    1. Vander Elst L, Van Haverbeke Y, Goudemant JF, Muller RN. Stability assessment of gadolinium complexes by P-31 and H-1 relaxometry. Magn Reson Med. 1994;31:437–44.
    1. Zhu D, Lu XL, Hardy PA, Leggas M, Jay M. Nanotemplate-engineered nanoparticles containing gadolinium for magnetic resonance imaging of tumors. Investig Radiol. 2008;43:129–40.
    1. Pedersen M. Safety update on the possible causal relationship between gadolinium-containing MRI agents and nephrogenic systemic fibrosis. J Magn Reson Imaging. 2007;25:881–3.
    1. Trivedi RA, U-King-Im JM, Graves MJ, et al. In vivo detection of macrophages in human carotid atheroma: temporal dependence of ultrasmall superparamagnetic particles of iron oxide-enhanced MRI. Stroke. 2004;35:1631–5.
    1. Schmitz SA, Taupitz M, Wagner S, Wolf KJ, Beyersdorff D, Hamm B. Magnetic resonance imaging of atherosclerotic plaques using superparamagnetic iron oxide particles. J Magn Reson Imaging. 2001;14:355–61.
    1. Kooi ME, Cappendijk VC, Cleutjens KB, et al. Accumulation of ultrasmall superparamagnetic particles of iron oxide in human atherosclerotic plaques can be detected by in vivo magnetic resonance imaging. Circulation. 2003;107:2453–8.
    1. Briley-Saebo K, Bjornerud A, Grant D, Ahlstrom H, Berg T, Kindberg GM. Hepatic cellular distribution and degradation of iron oxide nanoparticles following single intravenous injection in rats: implications for magnetic resonance imaging. Cell Tissue Res. 2004;316:315–23.
    1. Tang T, Howarth SP, Miller SR, et al. Assessment of inflammatory burden contralateral to the symptomatic carotid stenosis using high-resolution ultrasmall, superparamagnetic iron oxide-enhanced MRI. Stroke. 2006;37:2266–70.
    1. Briley-Saebo KC, Mani V, Hyafil F, Cornily JC, Fayad ZA. Fractionated Feridex and positive contrast: in vivo MR imaging of atherosclerosis. Magn Reson Med. 2008;59:721–30.
    1. Mani V, Adler E, Briley-Saebo KC, et al. Serial in vivo positive contrast MRI of iron oxide-labeled embryonic stem cell-derived cardiac precursor cells in a mouse model of myocardial infarction. Magn Reson Med. 2008;60:73–81.
    1. Mani V, Briley-Saebo KC, Itskovich VV, Samber DD, Fayad ZA. Gradient echo acquisition for superparamagnetic particles with positive contrast (GRASP): sequence characterization in membrane and glass superparamagnetic iron oxide phantoms at 1.5T and 3T. Magn Reson Med. 2006;55:126–35.
    1. Kellar KE, Fujii DK, Gunther WH, et al. NC100150 injection, a preparation of optimized iron oxide nanoparticles for positive-contrast MR angiography. J Magn Reson Imaging. 2000;11:488–94.
    1. Roch A, Muller RN, Gillis P. Water relaxation by SPM particles: neglecting the magnetic anisotropy? A caveat J Magn Reson Imaging. 2001;14:94–6.
    1. Briley-Saebo KC, Johansson LO, Hustvedt SO, et al. Clearance of iron oxide particles in rat liver: effect of hydrated particle size and coating material on liver metabolism. Invest Radiol. 2006;41:560–71.
    1. Xu XP, Meisel SR, Ong JM, et al. Oxidized low-density lipoprotein regulates matrix metalloproteinase-9 and its tissue inhibitor in human monocyte-derived macrophages. Circulation. 1999;99:993–8.
    1. Burke A, Cresswell N, Kolodgie FD, Virmani R, Tsimikas S. Increased expression of oxidation-specific epitopes and Lp(a) reflect unstable plaques in human coronary arteries (abstr) J Am Coll Cardiol. 2008;49 (Suppl A):A299.
    1. Tsimikas S, Palinski W, Halpern SE, Yeung DW, Curtiss LK, Witztum JL. Radiolabeled MDA2, an oxidation-specific, monoclonal antibody, identifies native atherosclerotic lesions in vivo. J Nucl Cardiol. 1999;6:41–53.
    1. Briley-Saebo KC, Amirbekian V, Mani V, et al. Gadolinium mixed-micelles: Effect of the amphiphile on in vitro and in vivo efficacy in apolipoprotein E knockout mouse models of atherosclerosis. Magnetic Resonance in Medicine. 2006;56:1336–46.
    1. Russell S, Cancel LM, Tarbell JM, Rumschitzki DS. A protein diffusion model of the sealing effect. Chem Eng Sci. 2009;64:4504–14.
    1. Pouliquen D, Perroud H, Calza F, Jallet P, Le Jeune JJ. Investigation of the magnetic properties of iron oxide nanoparticles used as contrast agent for MRI. Magn Reson Med. 1992;24:75–84.
    1. Majumdar S, Zoghbi SS, Gore JC. Pharmacokinetics of superpara-magnetic iron-oxide MR contrast agents in the rat. Invest Radiol. 1990;25:771–7.
    1. Seneterre E, Weissleder R, Jaramillo D, et al. Bone marrow: ultrasmall superparamagnetic iron oxide for MR imaging. Radiology. 1991;179:529–33.
    1. Tsimikas S. Noninvasive imaging of oxidized low-density lipoprotein in atherosclerotic plaques with tagged oxidation-specific antibodies. Am J Cardiol. 2002;90:22L–7L.

Source: PubMed

3
Abonnieren