701
Views
63
CrossRef citations to date
0
Altmetric
Review Article

Magnetic nanoparticles as contrast agents in biomedical imaging: recent advances in iron- and manganese-based magnetic nanoparticles

, , , , &
Pages 142-154 | Received 24 Sep 2013, Accepted 13 Dec 2013, Published online: 22 Apr 2014

References

  • Achraf AF, Katarzyna C, Ghislaine L, et al. (2009). In vivo imaging of carbon nanotube biodistribution using magnetic resonance imaging. Nano Lett 9:1023–1027
  • Alger JR, Frank JA. (1992). The utilization of magnetic resonance imaging in physiology. Annu Rev Physiol 54:827–846
  • Arruebo M, Fernández-Pacheco R, Ibarra MR, Santamaría J. (2007). Magnetic nanoparticles for drug delivery. Nano Today 2:22–32
  • Babes L, Denizot B, Tanguy G, et al. (1999). Synthesis of iron oxide nanoparticles used as MRI contrast agents: A parametric study. J Colloid Interf Sci 212:474–482
  • Bae KH, Lee K, Kim C, Park G. (2011). Surface functionalized hollow manganese oxide nanoparticles for cancer targeted siRNA delivery and magnetic resonance imaging. Biomaterials 32:176–184
  • Bartolazzi A, Peach R, Aruffo A, Stamenkovic I. (1994). Interaction between CD44 and hyaluronate is directly implicated in the regulation of tumor development. J Exp Med 180:53–66
  • Bhirde AA, Patel V, Gavard JL, et al. (2009). Targeted killing of cancer cells in vivo and in vitro with EGF-directed carbon nanotube-based drug delivery. ACS Nano 3:307–316
  • Boutry S, Laurent S, Elst LV, Muller RN. (2006). Specific E-selection targeting with a superparamagnetic MRI contrast agent. Contrast Media Mol I 1:15–22
  • Burns AA, Vider J, Ow H, et al. (2009). Fluorescent silica nanoparticles with efficient urinary excretion for nanomedicine. Nano Lett 9:442–448
  • Chan CK, Peng H, Liu G, et al. (2007). High-performance lithium battery anodes using silicon nanowires. Nat Nanotechnol 3:31–35
  • Choi JH, Nguyen FT, Barone PW, et al. (2007). Multimodal biomedical imaging with asymmetric single-walled carbon nanotube/iron oxide nanoparticle complexes. Nano Lett 7:861–867
  • Chomoucka J, Drbohlavova J, Huska D, et al. (2010). Magnetic nanoparticles and targeted drug delivering. Pharmaco Res 62:144–149
  • Chouly C, Pouliquen D, Lucet I, et al. (1996). Development of superparamagnetic nanoparticles for MRI: Effect of particle size, charge and surface nature on biodistribution. J Microencapsul 13:245–255
  • Chung HJ, Lee H, Bae K, et al. (2011). Facile synthetic route for surface-functionalized magnetic nanoparticles: Cell labeling and magnetic resonance imaging studies. ACS Nano 5:4329–4336
  • Colombo M, Corsi F, Foschi D, et al. (2010). HER2 targeting as a two-sided strategy for breast cancer diagnosis and treatment: Outlook and recent implications in nanomedical approaches. Pharmacol Res 62:150–165
  • Conlan R, Ernst R, Hahn EL, et al. (2002). A life-saving window on the mind and body: The development of magnetic resonance imaging. Nat Acad Sci 1:1–8
  • Cromer Berman SM, Walczak P, Bulte JW. (2011). Tracking stem cells using magnetic nanoparticles. Wiley Interdiscip Rev Nanomed Nanobiotechnol 3:343–355
  • Da Silva FR, Erdtmann B, Dalpiaz T, et al. (2010). Effects of dermal exposure to Nicotiana tabacum (Jean Nicot, 1560) leaves in mouse evaluated by multiple methods and tissues. J Agric Food Chem 58:9868–9874
  • Dalsin JL, Lin L, Tosatti S, et al. (2005). Protein resistance of titanium oxide surfaces modified by biologically inspired mPEG-DOPA. Langmuir 21:640–646
  • Dillon AC, Jones KM, Bekkedahl TA, et al. (1997). Storage of hydrogen in single-walled carbon nanotubes. Nature 386:377–379
  • El-Dakdouki MH, El-Boubbou K, Zhu DC, Huang, X. (2011). A simple method for the synthesis of hyaluronic acid coated magnetic nanoparticles for highly efficient cell labeling and in vivo imaging. RSC Adv 1:1449–1452
  • Elizondo G, Fretz CJ, Stark DD, et al. (1991). Preclinical evaluation of MnDPDP: New paramagnetic hepatobliliary contrast agent for MR imaging. Radiology 178:73–78
  • Escribano E, Fernández-Pacheco R, Valdivia JG, et al. (2012). Effect of magnet implant on iron biodistribution of Fe@C nanoparticles in the mouse. Arch Pharm Res 35:93–100
  • Gossuin Y, Gillis P, Hocq A, et al. (2009). Magnetic resonance relaxation properties of superparamagnetic particles. Wileys Interdiscipl Rev: Nanomed Nanobiotech 1:299–310
  • Gossuin Y, Roch A, Muller Robert N, Gillis P. (2002). An evaluation of the contributions of diffusion and exchange in relaxation enhancement by MRI contrast agents. J Magnet Res 158:36–42
  • Guo Q, Liu Y, Xu K, et al. (2013). Mouse lymphatic endothelial cell targeted probes: Anti-LYVE-1 antibody-based magnetic nanoparticles. Int J Nanomed 8:2273–2284
  • Gupta AK, Gupta M. (2005). Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. Biomaterials 26:3995–4021
  • Hong SC, Lee JH, Lee J, et al. (2011). Subtle cytotoxicity and genotoxicity differences in superparamagnetic iron oxide nanoparticles coated with various functional groups. Int J Nanomed 6:3219–3231
  • Huang CC, Khu NH, Yeh CS. (2010). The characteristics of sub 10 nm manganese oxide T1 contrast agents of different nanostructured morphologies. Biomaterials 31:4073–4078
  • Jemal A, Siegel R, Ward E, et al. (2009). Cancer statistics. CA Cancer J Clin 59:225–249
  • Jeyarama SA, Michael LM, Annie MT, et al. (2009). Single-walled carbon nanotube materials as T2-weighted MRI contrast agents. J Phys Chem C 113:19369–19372
  • Jia NQ, Lian Q, Shen HB, et al. (2007). Intracellular delivery of quantum dots tagged antisense oligodeoxynucleotides by funcationalized multiwalled carbon nanotubes. Nano Lett 7:2976–2980
  • Kamaly N, Kalber T, Thanou M, et al. (2009). Folate receptor targeted bimodal liposomes for tumor magnetic resonance imaging. Bioconjugate Chem 20:648–655
  • Karmakar A, Bratton SM, Dervishi E, et al. (2011a). Ethylenediamine functionalized-single-walled nanotube (f-SWNT)-assisted in vitro delivery of the oncogene suppressor p53 gene to breast cancer MCF-7 cells. Int J Nanomed 6:1045–1055
  • Karmakar A, Iancu C, Bartos DM, et al. (2012). Raman spectroscopy as a detection and analysis tool for in vitro specific targeting of pancreatic cancer cells by EGF-conjugated, single-walled carbon nanotubes. J App Toxicol 32:365–375
  • Karmakar A, Xu Y, Mahmood MW, et al. (2011b). Radio-frequency induced in vitro thermal ablation of cancer cells by EGF functionalized carbon-coated magnetic nanoparticles. J Mater Chem 21:12761–12769
  • Kievit FM, Stephen ZR, Veiseh O, et al. (2012). Targeting of primary breast cancers and metastases in a transgenic mouse model using rationally designed multifunctional SPIONs. ACS Nano 6:2591–2601
  • Kievit FM, Zhang M. (2011). Cancer nanoteheranostics: Improving imaging and therapy by targeted delivery across biological barriers. Avd Mater 23:H217–H247
  • Kim T, Momin E, Choi J, et al. (2011). Mesoporous silica-coated hollow manganese oxide nanoparticles as positive T1 contrast agents for labeling and MRI tracking of adipose-derived mesenchymal stem cells. J Am Chem Soc 133:2955–2961
  • Kim, T, Reis L, Rajan K, Shima M. (2005). Magnetic behavior of iron oxide nanoparticle biomolecule assembly. J Magn Magn Mater 295:132–138
  • Krishnan KM. (2010). Biomedical nanomagnetics: A spin through possibilities in imaging, diagnostics, and therapy. IEEE Tran Magn 46:2523–2558
  • Lalatonne Y, Richardi J, Pileni M. (2004). Van der Waals versus dipolar forces controlling mesoscopic organizations of magnetic nanocrystals. Nan Mater 3:121–125
  • Lapcik LJ, Lapcik L, De Smedt S, et al. (1998). Hyaluronan: Preparation, structure, properties, and applications. Chem Rev 98:2663–2683
  • Lauffer RB. (1987). Paramagnetic metal complexes as water proton relaxation agents for NMR imaging: Theory and design. Chem Rev 87:901–927
  • Lee H, Dellatore SM, Miller WM, Messersmith PB. (2007a). Mussel-inspired surface chemistry for multifunctional coatings. Science 318:426–430
  • Lee H, Yu MK, Park S, et al. (2007b). Thermally cross-linked superparamagnetic iron oxide nanoparticles: Synthesis and application as a dual imaging probe for cancer in vivo. J Am Chem Soc 129:12739–12745
  • Lee JH, Huh YM, Jun YW, et al. (2007c). Artificially engineered magnetic nanoparticle for ultra-sensitive molecular imaging. Nat Med 13:95–99
  • Li H, El-Dakdouki MH, Zhu DC, et al. (2012). Synthesis of β-cyclodextrin conjugated superparamagnetic iron oxide nanoparticles for selective binding and detection of cholesterol crystals. Chem Commun 48:3385–3387
  • Liu Z, Tabakman SM, Chen Z, Dai HJ. (2009). Preparation of carbon nanotube bioconjugates for biomedical applications. Nat Prot 4:1372–1382
  • Lu CW, Hung Y, Hsiao JK, et al. (2007). Bifunctional magnetic silica nanoparticles for highly efficient human stem cell labeling. Nano Lett 7:149–154
  • Massart R. (1981). Preparation of aqueous magnetic liquids in alkaline and acidic media. IEEE Trans Magn 17:1247–1248
  • McBain S, Yiu H, Dobson J. (2008). Magnetic nanoparticles for gene and drug delivery. Int J Nanomed 3:159–180
  • McCarthy JR, Weissleder R. (2008). Multifunctional magnetic nanoparticles for targeted imaging and therapy. Adv Drug Deliv Rev 60:1241–1251
  • Mikhaylov G, Vasiljeva O. (2011). Promising approaches in using magnetic nanoparticles in oncology. Biol Chem 392:955–960
  • Misra S, Heldin P, Hascall VC, et al. (2011). Hyaluronan–CD44 interactions as potential targets for cancer therapy. FEBS J 278:1429–1443
  • Miyawaki J, Yudasak M, Imai H, et al. (2006). In vivo magnetic resonance imaging of single-walled carbon nanohorns by labeling with magnetite nanoparticles. Adv Mater 18:1010–1014
  • Modo M, Hoehn M, Bulte JW. (2005). Cellular MR imaging. Mol Imaging 4:143–164
  • Moore A, Weissleder R, Bogdanov A. (1997). Uptake of dextran-coated monocrystalline iron oxides in tumore cells and macrophages. J Magn Reson Imaging 7:1140–1145
  • Mulder WJM, Strijkers GJ, van Tilborg GAF, et al. (2006). Lipid-based nanoparticles for contrast-enhanced MRI and molecular imaging. NMR Biomed 19:142–164
  • Müller RH, Jakobs C, Kayser O. (2001). Nanosuspensions as particulate drug formulations in therapy—Rational for development and what we can expect for the future. Adv Drug Deliv Rev 47:3–19
  • Namiki Y, Namiki T, Yoshida H, et al. (2009). A novel magnetic crystal-lipid nanostructure for magnetically guided in vivo gene delivery. Nat Nanotechnol 4:598–606
  • Nasongkla N, Bey E, Ren J, et al. (2006). Multifunctional polymeric micelles as cancer-targeted, MRI-ultrasensitive drug delivery systems. Nano Lett 6:2427–2430
  • Pankhurst Q, Connolly J, Joes S, Dobson J. (2003). Applications of magnetic nanoparticles in biomedicine. J Phys D: Appl Phys 36:167--181
  • Park J, An K, Hwang Y, et al. (2004). Ultra-large-scale syntheses of monodisperse nanocrystals. Nat Mater 3:891–895
  • Patolsky F, Zheng G, Lieber CM. (2006). Nanowire sensors for medicine and the life sciences. Nanomedicine-UK 1:51–65
  • Pawelczyk E, Jordan EK, Balakumaran A, et al. (2009). In vivo transfer of intracellular labels from locally implanted bone marrow stromal cells to resident tissue macrophages. PLoS One 4:e6712
  • Piao Y, Burns A, Kim J, et al. (2008). Designed fabrication of silica-based nanostructured particle systems for nanomedicine applications. Adv Funct Mat 18:3745–3758
  • Prato M, Kostarelos K, Bianco A. (2008). Functionalized carbon nanotbues in drug design and discovery. Acc Chem Res 41:60–68
  • Qiao R, Yang C, Gao M. (2009). Superparamagnetic iron oxide nanoparticles: From preparations to in vivo MRI applications. J Mater Chem 19:6274–6293
  • Reiser MF, Semmler W, Hricak H. (2008). Magnetic resonance tomography. Berlin, Heidelberg: Springer, 92–113
  • Richard C, Doan BT, Beloeil JC, et al. (2008). Noncovalent functionalization of carbon nanotubes with amphiphilic Gd3+ chelates: Toward powerful T1 and T2 MRI contrast agents. Nano Lett 8:232–236
  • Roduner E. (2006). Size matters: Why nanomaterials are different. Chem Soc Rev 35:583–592
  • Rose-Pehersson SL, Pehersson PE. (2004). Environmental applications: Sensor and sensor systems: Overview. In: Karn B, Masciangioli T, Zhang W-x, et al., eds. Nanotechnology and the environment: Applications and implications. Washington (DC): Oxford University Press, 154–156
  • Selvan ST, Patra PK, Ang CY, Ying JY. (2007). Synthesis of silica-coated semiconductor and magnetic quantum dots and their use in the imaging of live cells. Angew Chem Int Ed 46:2448–2452
  • Senyei A, Widder K, Czerlinski G. (1978). Magnetic guidance of drug carrying microspheres. J Appl Phys 49:3578–3583
  • Shin J, Anisur RM, Ko MK, et al. (2009). Hollow manganese oxide nanoparticles as multifunctional agents for magnetic resonance imaging and drug delivery. Angew Chem Int Ed 48:321–324
  • Silva AKA, Wilhelm C, Kolosnjaj-Tabi J, et al. (2012). Cellular transfer of magnetic nanoparticles via cell microvesicles: Impact on cell tracking by magnetic resonance imaging. Pharm Res 29:1392–1403
  • Singh NP, McCoy MT, Tice RR, Schneider EL. (1988). A simple technique for quantitation of low levels of DNA damage in individual cells. Exp Cell Res 175:184–191
  • Sitharaman B, Kissell KR, Hartman KB, et al. (2005). Superparamagnetic gadonanotubes are high-performance MRI contrast agents. Chem Commun 31:3915–3917
  • Sonvico F, Mornet S, Vasseur S, et al. (2006). Folate-conjugated iron oxide nanoparticles for solid tumor targeting as potential specific magnetic hyperthermia mediators: Synthesis, physicochemical characterization, and in vitro experiments. Bioconjugate Chem 16:1181–1188
  • Stöber W, Fink A, Bohn E. (1968). Controlled growth of monodisperse silica spheres in the micron size range. J Colloid Interf Sci 26:62–69
  • Sun SH, Zeng H, Robinson DB, et al. (2004). Monodisperse MFe2O4 (M = Fe, Co, Mn) nanoparticles. J Am Chem Soc 126:273–279
  • Telgmann L, Faber H, Jahn S, et al. (2012). Identification and quantification of potential metabolites of Gd-based contrast agents by electrochemistry/separation/mass spectrometry. J Chromatogr A 1240:147–155
  • Tran TD, Caruthers SD, Hughes M, et al. (2007). Clinical applications of perfluorocarbon nanoparticles for molecular imaging and targeted therapeutics. Int J Nanomed 2:515–526
  • Verity MA. (1999). Manganese neurotoxicity: A mechanistic hypothesis. Neurotoxicology 20:489–497
  • Wang Y, Wong JF, Teng X, et al. (2003). “Pulling” nanoparticles into water: Phase transfer of oleic acid stabilized monodisperse nanoparticles into aqueous solutions of α-cyclodextrin. Nano Lett 3:1555–1559
  • Widder KJ, Senyei AE, Scarpelli DG. (1978). Magnetic microspheres: A model system for site specific drug delivery in vivo. Exp Biol Med 158:141
  • Wong N, Kam S, Dai HJ. (2005). Carbon nanotubes as intracellular protein transporters: Generality and biological funcationality. J Am Chem Soc 127:6021–6026
  • Xu Y, Karmakar A, Heberlein WE, et al. (2012). Multifunctional magnetic nanoparticles for synergistic enhancement of cancer treatment by combinatorial radio frequency thermolysis and drug delivery. Adv Healthcare Mater 1:493–501
  • Yang H, Zhao F, Li Y, et al. (2013). VCAM-1-targeted core/shell nanoparticles for selective adhesion and deliversy to endothelial cells with lipopolysaccharide-induced inflammation under shear flow and cellular magnetic resonance imaging in vitro. INT J Nanomed 8:1897–1906
  • Yellen BB, Forbes ZG, Halverson DS, et al. (2005). Targeted drug delivery to magnetic implants for therapeutic applications. J Magn Magn Mater 293:647–654
  • Yi DK, Selvan ST, Lee SS, et al. (2005). Silica-coated nanocomposites of magnetic nanoparticles and quantum dots. J Am Chem Soc 127:4990–4991
  • Yin M, Wang M, Miao F, et al. (2012). Water-dispersible multiwalled carbon nanotube/iron oxide hybrids as contrast agents for cellular magnetic resonance imaging. Carbon 50:2162–2170
  • Yiu HHP, Pickard MR, Olariu CI, et al. (2011). Fe3O4-PEI-RITC magnetic nanoparticles with imaging and gene transfer capability: Development of a tool for neural cell transplantation therapies. Pharm Res 29:1328–1343
  • Yoo, JW. (2012). Toward improved selectivity of targeted delivery: The potential of magnetic nanoparticles. Arch Pharm Res 35:1–2
  • Yoon T-J, Kim JS, Kim BG, et al. (2005). Multifunctional nanoparticles possessing a “Magnetic Motor Effect” for drug or gene delivery. Angew Chem Int Ed 44:1068–1071
  • Zhang L, Gu FX, Chan JM, et al. (2008). Nanoparticles in medicine: Therapeutic applications and developments. Clin Pharmocol Ther 83:761–769
  • Zhao V, Nie W, Liu X, et al. (2008). Shape- and size-controlled synthesis and dependent magnetic properties of nearly monodisperse Mn3O4 nanocrystals. Small 4:77–81
  • Zhdanov RI, Podobed OV, Vlassov VV. (2002). Cationic lipid-DNA complexes-lipoplexes-for gene transfer and therapy. Bioelectrochemistry 58:53–64

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.