You are on page 1of 8

T.K. Indira and P.K.

Lakshmi : Magnetic Nanoparticles – A Review 1035

International Journal of Pharmaceutical Sciences and Nanotechnology

Volume 3 • Issue 3 • October - December 2010


Review Article
Magnetic Nanoparticles – A Review

T.K. Indira, P.K. Lakshmi


G. Pulla Reddy College of Pharmacy, Hyderabad, India.

ABSTRACT: In the recent past, the targeted drug delivery has gained more attention for various advantages. Amongst
the plethora of avenues explored for targeted drug delivery, magnetic nanoparticles backed foremost attention offering local
drug delivery, reduced side effects and controlled drug release for prolonged period of time addressing problems of healthy
tissue damage, drug wastage. An approach was made herein to review the history of magnetic guidance, concept of
magnetic nanoparticles, their advantages, methods of preparation, characterization, applications explored in various fields
of drug delivery. This review also deals up with the disadvantages of magnetic nanoparticles, the alternative way to
overcome. It also focuses on exemplifying different drug formulations formulated into magnetic nanoparticles.
Future prospectives, challenges faced in drug delivery and scope of magnetic nanoparticles have also been addressed.

KEY WORDS: magnetic nanoparticles; magnetic drug targeting; drug targeting; super paramagnetism

risk of particle aggregation (Ruiz-Hernandez et al., 2008).


Introduction First, they have sizes that place them at dimensions
The concept of magnetic drug targeting (MDT) has been comparable to those of a virus (20±500 nm), a protein
around for over 30 years and simply entails retaining (5±50 nm) or a gene (2 nm wide and 10±100 nm long)
specially designed magnetic drug carrier particles (Tartaj et al., 2004). The magnetic nanoparticles, which
(MDCPs) at a specific site in the body using an externally used in bio-applications are usually made from
applied magnetic field which was first used in early 1940s biocompatible materials such as magnetite (Fe3O4) for
as a new methodology in waste water treatment (Arias et which susceptibility is large (Sachin Shaw et al., 2009).
al., 2001). Nanotechnology is at the leading edge of the
Targeting of drugs by nanoparticles is intended to
rapidly developing new therapeutic and diagnostic
reduce drug wastage, frequency of drug administration,
concepts in all areas of medicine (Veronica et al., 2008).
side effects providing prolonged, sustained drug delivery
Magnetic nanoparticles (MNPs) are a class of engineered
to desired targeted organ (Ece Simsek and Mehmet Akif
particulate materials of <100nm that can be manipulated
Kilic, 2005). Iron oxide magnetic nanoparticles present a
under the influence of an external magnetic field. MNPs
higher performance in terms of chemical stability and
are commonly composed of magnetic elements, such as
biocompatibility compared with metallic nanoparticles.
iron, nickel, cobalt and their oxides like magnetite
(Ece Simsek and Mehmet Akif Kilic, 2005). Nanoparticles
((Fe3O4), maghemite (٧-Fe2O3), cobalt ferrite (Fe2CoO4),
have a large surface that may be properly modified to
chromium di-oxide (CrO2) (Yang et al., 2006).
attach biological agents (Tartaj et al., 2004).
Advantages of Magnetic Nanoparticles Magnetic Nanoparticles: Magnetic property
There had been a growing interest on the properties and
The classification of a material's magnetic properties is
broad range of applications of colloids in recent years.
based on its magnetic susceptibility (χ), which is defined
Magnetic biomaterials provide the ability to be directed
by the ratio of the induced magnetization (M) to the
and concentrated within the target tissue by means of
applied magnetic field (H). In ferri- and ferromagnetic
external magnetic field and to be removed once therapy
materials, magnetic moments align parallel to H, coupling
was completed.
interactions between the electrons of the material result in
Magnetic nanoparticles display the phenomenon of ordered magnetic states. The susceptibilities of these
superparamagnetism, not keeping magnetized after the materials depend on their temperature, external field H and
action of magnetic filed, offering advantage of reducing atomic structures. At small sizes (in the order of tens of

1035
1036 International Journal of Pharmaceutical Sciences and Nanotechnology Volume 3 • Issue 3 • October-December 2010

nanometers), ferri-or ferro-magnetic materials, such as 2000). Both parameters affect the chemical composition of
MNPs, become a single magnetic domain and therefore the surface and consequently, the electrostatic surface
maintain one large magnetic moment. However, at charge of the particles. Under these conditions, magnetite
sufficiently high temperatures (i.e., blocking temperature, particles are formed by aggregation of primary particles
TB) thermal energy is sufficient to induce free rotation of formed within a Fe(OH)2 gel. This ordered aggregation
the particle resulting in a loss of net magnetization in the gives rise to spherical crystalline particles (Sugimoto T and
absence of an external field (Conroy Sun et al., 2008). Matijevic E, 1980). The smallest particles can also be
Lack of remnant magnetization after removal of external generated after adding polyvinyl alcohol (PVA) to the iron
fields enables the particles to maintain their colloidal salts (Lee J et al., 1996). Modifications of this method
stability and avoids aggregation making it feasible for their allow for synthesis in the presence of dextran or any other
use in biomedical applications. The coupling interactions substance that renders the magnetic nanoparticles
in single magnetic domains result in much higher magnetic biocompatible and thus make this method especially
susceptibilities than paramagnetic materials (Lu et al., appropriate for in vivo applications (Molday R S and
2007). Mackenze D, 1982; Palmacci S and Josephson L, 1993).
Nanosized magnetic particles can be obtained by
Synthetic Routes of Preparation of Magnetic transferring an acidic iron (II)/iron (III) salt solution into
nanoparticles: iron (II, III)-carbonate by adding equivalent amounts of
Precipitation from solution alkaline carbonate, followed by thermal oxidation
(Bergemann C, 1996). The size of the particles can be
Uniform particles are usually prepared via homogeneous controlled by the thermal reaction velocity and
precipitation reactions, a process that involves the concentration of the iron salts. This method was used
separation of the nucleation and growth of the nuclei widely for magnetic Fe3O4 nanoparticles for waste water
(Sugimoto T, 2000). purification where three samples were prepared by
Co-precipitation different methods in which first sample was prepared by
this method (Shen et al., 2009). There was evidence of
This method may be the most promising one because of its direct binding of YADH (yeast alcohol dehydrogenase)
simplicity and productivity (ZHAO Yuanbi et al., 2008). It magnetite nanoparticles via carbodiimide activation using
is widely used for biomedical applications because of ease this method (Dong-Hwang Chen, Min-Hung Liao, 2002).
of implementation and need for less hazardous materials Iron oxide nanoparticles prepared by the displacement of
and procedures (Patnaik, 2004). Co-precipitation is air by N2 gas during preparation-prevented oxidation of
specifically the precipitation of an unbound "antigen along ferrous ion in the aqueous solution and controlled the
with an antigen-antibody complex" in terms of medicine particle size (Aslam Khan, 2008). By the use of magnetic
(Patnaik, 2004).The reaction principle is simply as: nanoparticles (Fe3O4) and measurement of magnetic
Fe2+ + 2Fe3+ + 8OH– ⇔ Fe (OH)2 + 2Fe(OH)3 → susceptibility, a new UF (ultra filtration) membrane
integrity test is presented and evaluated (Guo et al., 2009).
Fe3O4 + 4H2O (Guo et al., 2009).
Development of novel magnetic nano-adsorbent using iron
There are two main methods for the synthesis in oxide nanoparticles as cores and polyacrylic acid (PAA) as
solution of magnetite spherical particles in the nanometer ionic exchange groups (Liao MH, Chen DH, 2002)
range. In the first, ferrous hydroxide suspensions are possessed a high ion-exchange capacity and was quite
partially oxidized with different oxidizing agents effective for the enzyme recovery by the adsorption of
(Sugimoto T and Matijevic E, 1980). For example, methylene blue (MB) from an aqueous solution by
spherical magnetite particles of narrow size distribution polyacrylic acid-bound iron oxide magnetic nanoparticles
with mean diameters between 30 and 100 nm can be (Sou-Yee Mak, 2004). Preparation of magnetic
obtained from Fe(II) salt, a base and a mild oxidant (nitrate nanoparticles Fe3O4, coating with amino silane for
ions) (Sugimoto T and Matijevic E, 1980). immobilizing them with the pseudomonas putida esterase
The other method consists in ageing stoichiometric via glutaraldehyde coupling reaction was reported (Shyh-
mixtures of ferrous and ferric hydroxides in aqueous Yu Shaw, 2006). Fe3O4 magnetic nanoparticles with
media, yielding spherical magnetite particles homogeneous narrow size distribution can be obtained by controlling the
in size (Massart R and Cabuil V, 1987). It has been shown titrating rate of ammonium hydroxide (Zhao Yuanbi et al.,
that by adjusting the pH and the ionic strength of the 2008). The effect of Fe3O4 content in Fe3O4/ZnO
precipitation medium, it is possible to control the mean core/shell magnetic nanoparticles on the photocatalytic
size of the particles over one order of magnitude (from 15 degradation was studied (R.Y. Hong et al., 2008). It is well
to 2 nm) (Jolivet J P, 2000). The size decreases as the pH known that the NH2 group on chitosan molecules may
and the ionic strength in the medium increases (Jolivet J P, interact with Fe2+in aqueous solution, So, for the purpose
T.K. Indira and P.K. Lakshmi : Magnetic Nanoparticles – A Review 1037

of lipase immobilization, chitosan nanoparticles were as an oxidizer and the adsorption of Saccharomyces
firstly prepared by cross-linking with TPP in HCl solution cerevisiae mandelated dehydrogenase (SCMD) protein on
then oxidizing in aqueous solution (Yujun Wang et al., the surface-modified magnetic nanoparticles coated with
2009).Using the above method, ultra-stable biocompatible chitosan studied in a batch adsorption system has been
magnetic fluids using citrate-coated cobalt ferrite reported. Functionalization of surface-modified magnetic
nanoparticles to evaluate colloidal stability using four particles have been performed by the covalent binding of
different biocompatible magnetic fluid samples, namely chitosan onto the surface of magnetic Fe3O4 nanoparticles
S25P2, S25P5, S60P2, and S60P5 which showed a long- and kinetics of adsorption were studied (Gui-Yin Li et al.,
term colloidal Stability, though sample S25P5 revealed a 2008). Saccharomyces cerevisiae alcohol dehydrogenase
higher fluctuation on the nanoparticle concentration over (SCAD) immobilized to the magnetic Fe3O4-chitosan
the time were reported (Morais et al., 2006). nanoparticles via glutaraldehyde coupling reaction has
The difficulty in preparing Fe3O4 magnetic been reported The size, structure and magnetic property of
nanoparticles by chemical co-precipitation is the tendency the particles before and after binding to the SCAD, the
of agglomeration of particles because of extremely small kinetic constants, thermal stability and reusability of
particle size leading to great specific surface area and high SCAD immobilized on the magnetic Fe3O4-chitosan
surface energy, consideration of effect of alkali, emulsifier, nanoparticles using phenylglyoxylic acid as substrate were
and reaction temperature are the decisions making factors also reported (Gui Yin Li et al., 2008). Fe3O4 particles and
of final product (ZHAO Yuanbi et al., 2008). Controlled Fe3O4–chitosan nanocomposites prepared by this method
syntheses of magnetic Fe3O4–chitosan nanoparticles under were regular, sphere with a mean diameter of 23 nm and
UV irradiation in aqueous system were also reported where 25 nm (Gui Yin Li et al., 2008) and covalent binding of
irradiation under UV light did not result in a phase change alpha-ketoglutaric acid chitosan (KCTS) onto the surface
and average size in aqueous solution measured was 64 nm of Fe3O4 magnetic nanoparticles via carbodiimide
(Lian-ying Zhang et al., 2009). Immobilized protease on activation were also reported (Gui Yin Li et al., 2008). The
the magnetic nanoparticles was recently investigated for colloidal dispersion of magnetite nanoparticles, of triiron
the hydrolysis of rapeseed meals (Xin Jin, Ju-Fang Li et a., dodecacarbonyl (Fe3(CO)12) in diethyleneglycol
2010). diethylether as a continuous phase and oleic acid as a
stabilizer were reported. The obtained nanoparticles
High-temperature decomposition of annealed at 300, 700 and 9000C and the annealing
organic precursors temperature allowed control of size and size distribution of
the particles, as well as their composition and magnetic
The decomposition of iron precursors in the presence of properties (Daniel Amara et al., 2009). Carbon-
hot organic surfactants has yielded markedly improved encapsulated iron (Fe@C) nanoparticles with core/shell
samples with good size control, narrow size distribution structure have been synthesized by detonation method,
and good crystallinity of individual and dispersible using a composite explosive precursor where the
magnetic iron oxide nanoparticles (Hyeon et al., 2003). For detonation reaction was ignited by a non-electric detonator
example, injecting solutions of FeCup3 (Cup:N- in nitrogen gas in an explosion vessel and results showed
nitrosophenylhydroxylamine) in octylamine into long- that detonation products were made up of the body
chain amines at 250–300˚C yielded nanocrystals of centered cubic iron core and the graphitic carbon core
maghemite ranging from 4 to 10 nm in diameter, are diameter in the range of 15–50 nm (Luo Ning et al.,
crystalline, and are dispersible in organic solvents 2009). Epithelial internalization of superparamagnetic
(Rockenberger J et al., 1999). Monodispersed magnetite nanoparticles and response to external magnetic field were
nanoparticles with sizes from 3-20 nm by the high- reported by using modified precipitation method (Kenneth
temperature (265˚C) reaction of iron (III) acetylacetonate Dormer et al., 2005).
in phenyl ether in the presence of alcohol, oleic acid, and
oleylamine were prepared (Shen S and Zeng H, 2002). In Other solution techniques
particular, magnetite nanoparticles around 4 nm were
Methods for the production of magnetic nanoparticles for
obtained by the thermal decomposition of the iron
in vivo applications were reported. A variety of protein
precursor but to obtain diameters up to 20 nm a seed-
components that function as carriers or storage devices for
mediated growth method was required (Pedro Tartaj et al.,
metal components is been developed by nature. Of these
2003). For preparation of maghemite nanoparticles of 13
systems, the iron-storage protein ferritin is probably the
nm, Fe(CO)5 was injected into a solution containing
most intensively studied and best understood (Chasteen N
surfactants and a mild oxidant (trimethylamine oxide)
D and Harrison P M, 1999). Ferritin consists of a central
(Hyeon T et al., 2001). Fe3O4 nanoparticles prepared by
core of hydrated iron (III) oxide encapsulated with a multi
hydrothermal method with a ferrous complex using H2O2
subunit protein shell. As a result of the inner diameter of
1038 International Journal of Pharmaceutical Sciences and Nanotechnology Volume 3 • Issue 3 • October-December 2010

the nanoreactors, magnetite (Meldrum F C et al., 1992) and also reported (Boutonnet M, 1982). β-FeOOH
magnetite/maghemite nanoparticles (Wong K K W et al., nanoparticles prepared in a microemulsion system with
1998) of about 6–7 nm in diameter. The nonionic surfactant polyoxyethylene(4)nonylphenylether
magnetite/maghemite particles were generated by with 20–30 nm length nanorods with a crystal structure,
oxidation of apoferritin (empty ferritin) with where the size and shapes of β-FeOOH nanoparticles can
trimethylamino-N-oxide, which was loaded with various be manipulated by the surfactant has been reported
amounts of iron (II) ions. recently (Y.X. Yang et al., 2008). In situ preparation of
Use of dendrimers as templating hosts for the magnetic chitosan/Fe3O4 composite nanoparticles in tiny
production of magnetic nanoparticles for in vivo pools of water-in-oil microemulsion where the chitosan
applications were reported by using carboxylated poly particle size varied from 10 nm to 80 nm with different
(amidoamine) PAMAM dendrimers (generation 4.5) for molecular weight of chitosan has been reported (Zhi Jia et
the synthesis and stabilization of ferrimagnetic iron oxide al., 2006). Magnetically responsive micro gel that consists
nanoparticles where oxidation of Fe (II) at slightly elevated of a small iron oxide magnetic nanoparticles (∼15 nm in
pH and temperature resulted in the formation of highly diameter) embedded in a biocompatible micro gel varying
soluble nanocomposites of iron oxides and dendrimer, from ∼65 to ∼110 nm in diameter has been reported
which are stable under a wide range of temperatures and
pH were reported (Strable E et al., 2001). FePt (Aslam Khan, 2006). The chitosan-coated magnetic
nanoparticles with narrow size-distribution were prepared nanoparticles (CSMNPs) prepared as carriers of 5-
by simple chemical reduction method at room temperature Fluorouracil (CS–5-FuMNPs) through a reverse
using PAMAM-OH as dendrimers in which metal ions are microemulsion method were spherical in shape with an
coordinated in, and NaBH4 as reductant. The as-made FePt average size of 100±20 nm, low aggregation and good
magnetic responsivity (Long zhang Zhu et al., 2009).
nanoparticles are spherical with an average size of ∼3 nm
and have the chemically disordered face-center-cubic (fcc) Polyols
structure. The FePt nanoparticles can be transformed to the
Fine metallic particles can be obtained reduction of
face-center-tetragonal (fct) phase after annealing at 7000C
dissolved metallic salts and direct metal precipitation from
for 1 h. The prepared fcc FePt nanoparticles are found to
a solution containing a polyol (Matijevic E, 1989).
be superparamagnetic at room temperature. After
annealing in a reducing atmosphere, they transformed to In the polyol process, the liquid polyol acts as the
ferromagnetic and showed high covercivity (Litao Bai et solvent of the metallic precursor, the reducing agent and in
al., 2009). some cases as a complexing agent for the metallic cations.
The metal precursor can be highly or only slightly soluble
Microemulsions in the polyol. The solution is stirred and heated to a given
temperature reaching the boiling point of the polyol for
Water-in-oil (W/O) microemulsions systems, a fine micro
less reducible metals. By controlling the kinetic of the
droplets of the aqueous phase trapped within assemblies of
precipitation, nonagglomerated metal particles with well-
surfactant molecules dispersed in a continuous oil phase.
defined shape and size can be obtained. In vitro
(Pedro Tartaj et al., 2003). The surfactant-stabilized micro
cytotoxicity assessment, and in vivo visualization of
cavities (typically in the range of 10 nm) provide a
multimodal, RITC-labeled, silica-coated magnetic
confinement effect that limits particle nucleation, growth,
nanoparticles for labeling human cord blood–derived
and agglomeration (Pileni M P, 1993). Nanosized magnetic
mesenchymal stem cells have been reported (Ki-Soo Park
particles were prepared with average sizes from 4 to 12 nm
et al., 2010). Fe3O4 magnetic nanoparticles prepared with
by using ferrous dodecyl sulfate, Fe(DS)2, micellar
the co-precipitation method combining a surface
solution to produce nanosized magnetic particles whose
decoration process, and the polyol process, and the factors
size is controlled by the surfactant concentration and by
influencing the adsorption of metal ions, e.g., pH,
temperature (Feltin N and Pileni M P, 1997). Magnetite
temperature, amount of adsorbent, and contacting time
nanoparticles around 4 nm in diameter have been prepared
were been reported (Shen et al., 2009).
by the controlled hydrolysis with ammonium hydroxide of
FeCl2 and FeCl3 aqueous solutions within the reverse
Aerosol/vapor methods
micelle nanocavities generated by using Aerosol Optical
Thickness (AOT) as surfactant and heptane as the Spray and laser pyrolysis have been shown to be excellent
continuous oil phase (Lopez-Quintela and Rivas, 1993). techniques for the direct and continuous production of
Reverse micelle reaction carried out using Cetyl Trimethyl well-defined magnetic nanoparticles under exhaustive
Ammonium Bromide (CTAB) as the surfactant, octane as control of the experimental conditions (Tartaj et al., 2003).
the oil phase, and aqueous reactants as the water phase was Spray pyrolysis is a process in which a solid is obtained by
T.K. Indira and P.K. Lakshmi : Magnetic Nanoparticles – A Review 1039

spraying a solution into a series of reactors where the copolymers. Determination of magnetic drug loading and
aerosol droplets undergo evaporation of the solvent and encapsulation efficiency is done by calculating the weight
solute condensation within the droplet, followed by drying of drug encapsulated in the nanoparticles, gross weight of
and thermolysing of the precipitated particle at higher the nanoparticles with that of total weight of the feeding
temperature (Messing et al., 1993). Recently, this method drug.
has been used to prepare colloidal aggregates of
Invitro drug release studies were determined using an
superparamagnetic maghemite nanoparticles in the form of
in vitro apparatus-simulating human circulatory to test the
hollow or dense spheres and with the possibility of having,
retentions of ferrofluids under various flow speeds of
surface enriched in silica (Tartaj et al., 2001; Tartaj et al.,
medium, various magnetic fields with different magnitudes
2002; Tartaj et al, 2003; Tartaj et al., 2004). Of special
and gradient. The retention of ferrofluids in the target was
interest is that the method also allows the preparation of
obtained through measuring the content of magnetic
air-stable superparamagnetic α-Fe metallic and FeCo
nanoparticles in the beaker by an atomic absorption
nanomagnets encapsulated in silica or alumina (Tarta et al.,
spectrometer (Hua Xu et al., 2005).
2004; Tarta et al., 2004). The laser pyrolysis method
involves heating a flowing mixture of gases with a In vivo drug release was determined by using Healthy
continuous wave carbon dioxide laser, which initiates and male Sprague–Dawley rats of 280–300g (Hua Xu et al.,
sustains a chemical reaction (Cannon et al., 1982; Bomati- 2005), Sheep (Katja Schulze et al., 2005), mice as
Miguel et al., 2002). Above a certain pressure and laser experimental animals between 8 and 9 weeks of age (Ki-
power, a critical concentration of nuclei is reached in the Soo Park et al., 2010).
reaction zone, which leads to homogeneous nucleation of
particles that are further transported to a filter by an inert Applications of Magnetic Nanoparticles in
gas. Three characteristics of this method must be drug targeting
emphasized: (a) the small particle size, (b) the narrow Magnetic nanoparticles have attracted the attention of
particle size distribution, and (c) the nearly absence of scientific community as potential materials for carriers of
aggregation. Biocompatible magnetic dispersions have drug or gene delivery, DNA, biomolecules separation,
been prepared from ۷-Fe2O3 nanoparticles (5 nm) hypothermal treatment of tumours, contrast agents for
synthesized by continuous laser pyrolysis of Fe(CO)5 magnetic resonance imaging, information storage, catalysis
vapors (Bautista et al., 2004; Veintemillas-Verdaguer et and biomedical uses.
al., 2004). The laser pyrolysis technique seems to be a
good alternative to the coprecipitation method for The different applications of magnetic nanoparticles
producing MRI contrast agents with the advantage of being include:
a continuous synthesis method that leads to uniform • Use of superparamagnetic iron oxide
particles capable of being dispersed and therefore nanoparticles+, coated with polyvinyl alcohol,
transformed in a biocompatible magnetic liquid (Pedro (PVA-SPION) and its fluorescently-functionalized
Tartaj et al., 2003). analogue (amino-PVA-Cy3.5-SPION) were
evaluated in the treatment of inflammatory joint
Characterization of magnetic nanoparticles: diseases (Katja Schulze et al., 2005).
Magnetic nanoparticles are characterized for its size and • Effective targeting of selective tissue markers has
shape using TEM, (Hong et al., 2008). Structural been demonstrated using derivatized super
characterization using XRD using Cu Kα radiation source paramagnetic nanoparticles. For example, Annexin
generated at 40 kV and 30 mA (Nan Wang et al., 2010). V-SPIO successfully targeted atherosclerotic
Particle size distribution was measured by dynamic laser plaque, Hepama-1 (humanized monoclonal
scattering analyzer (DLS) (ZHAO Yuanbi et al., 2008). antibody)-SPIO targeted liver cancer and HIV-1 tat
The magnetic density of ferromagnet was measured by peptide-SPOI have demonstrated efficacy for
SJZP-1 Gauss/Tesla-Meters (ZHAO Yuanbi et al., 2008). intracellular magnetic labeling and non-invasive
Chemical composition determination using atomic tracking of a large number of cell types. Targeting
absorption spectroscopy (Morais et al., 2006). Magnetite galactose-terminal-ASPIO (amino-functionalized
content determination is done by TGA, Inductively SPIO) to the cell-surface of asialoglycoprotein
coupled plasma atomic emission spectrometer (ICP-AES) receptor (ASGPR) expressed by hepatocytes has
(Misara Hamoudeh et al., 2007) and magnetic property been reported (Guifang Huang et al., 2008).
using vibrating sample magnetometer (Lian-ying Zhang et • Biomedical applications of magnetic nanoparticles
al., 2010). Differential scanning calorimetry (DSC) is used includes cellular labeling/cell separation,
to analyze the structure of the copolymer block and to detoxification of biological fluids, tissue repair,
determine the glass transition temperature (Tg) of the
1040 International Journal of Pharmaceutical Sciences and Nanotechnology Volume 3 • Issue 3 • October-December 2010

drug delivery, magnetic resonance imaging, of using a ferromagnetic implant, in conjunction with the
hyperthermia, magnetofection (Arun Kumar et al., multi drug carrier particles (MDCPs) and an external
2009) were well addressed (Ajay Kumar Gupta, magnetic field, has been receiving considerable attention
Mona Gupta, 2005). in forth coming era termed as Implant assisted Magnetic
• Magnetic resonance imaging (MRI) (Conroy Sun et drug Targeting (Misael O. Aviles et al., 2008; P.J. Cregg et
al., 2008) properties of novel oleic acid-coated iron al., 2009).
oxide and pluronic-stabilized MNPs of doxorubicin
(Jian-Bo Sun et al., 2007; Munnier et al., 2008; Conclusion
Babita Gaihre et al., 2009; Kayal, Ramanujan,
2010), Magnetite/poly (alkylcyanoacrylate) Magnetic Nanoparticles offer to open new vistas in the
(core/shell) nanoparticles of 5-fluorouracil, area of drug delivery, and they promises as a prudent tactic
gemcitabine and cisplatin (Yang et al., 2006) to overcome the drug delivery related problems when the
delivery systems for active targeting (Jose L. Arias problems of toxicity, localization, cost are addressed.
et al., 2008) and paclitaxel demonstrated highly
synergistic antiproliferative activity in MCF-7 References
breast cancer cells (Tapan K. Jain et al., 2008).
Abdalla, M.O.; Aneja, R.; Dean, D.; Rangari, V.; Russell, A.;
• Chemical conjugation of urokinase to magnetic Jaynes, J.; Yates, C.; Turner, T. Synthesis and
nanoparticles for targeted thrombolysis was studied characterization of noscapine loaded magnetic polymeric
(Feng Bi et al., 2009). nanoparticles. J. Magn. Magn. Mater. 2010, 322, 190-196.
• Development of hyaluronic acid–Fe2O3 hybrid
Amara, D.; Felner, I.; Nowik, I.; Margel, S. Synthesis and
magnetic nanoparticles for targeted delivery of characterization of Fe and Fe3O4 nanoparticles by thermal
peptides was reported (Arun Kumar et al., 2007). decomposition of triiron dodecacarbonyl. Colloids Surf. A:
• Cell targeting with multifunctional magnetic Physicochem. Eng. Aspects 2009, 339, 106-110.
nanoparticles was studied in Gene therapy (Akira
Arias, J.L.; Gallardo, V.; Ruiz, M.A.; Delgado, A.V.;
Ito et al., 2005).
Magnetite/poly (alkylcyanoacrylate) (core/shell)
• Antitumour immunity induction by hyperthermia nanoparticles as 5-Fluorouracil delivery systems for active
using magnetite nanoparticles were explored targeting. Eur. J. Pharm. Biopharm. 2008, 69, 54-63.
(Helene Rahn et al., 2009).
Aslam, K. Preparation and characterization of magnetic
• Transferrin derivatised particles to localize to the nanoparticles embedded in microgels. Mater. Lett. 2008, 62,
cell membrane without instigating receptor- 898–902.
mediated endocytosis, and also induce up-
regulation in the cells for many genes, particularly Aviles, M.O.; Mangual, J.O.; Ebner, A.D.; Ritter, J.A. Isolated
in the area of cytoskeleton and cell signaling has swine heart ventricle perfusion model for implant assisted-
been investigated (Catherine C. Berry et al., 2004). magnetic drug targeting. Int. J. Pharm. 2008, 361, 202-208.

• Non-invasively targeting respiratory tract Bai, L.; Wan, H.; Shane Street, C. Preparation of ultrafine FePt
deposition of high aspect ratio aerosols loaded with nanoparticles by chemical reduction in PAMAM-OH
magnetic nanoparticles by controlling particle template. Colloids Surf. A: Physicochem. Eng. Aspects 2009,
orientations through magnetic field alignment to 349, 23-28.
specific locations within the lung (Andrew R. Berry, C.C.; Charles, S.; Wells, S.; Dalby, M.J.; Curtis, A.S.G.
Martin, Warren H. Finlay, 2008). The influence of transferrin stabilised magnetic nanoparticles
• Folic acid–Pluronic F127 magnetic nanoparticle on human dermal fibroblasts in culture. Int. J. Pharm. 2004,
clusters for combined targeting, diagnosis (Jia-Jyun 269, 211–225.
Lin et al., 2009) and therapeutical applications have Berry, C.C.; Wells, S.; Charles, S.; Curtis, A.S.G. Dextran and
been studied recently. albumin derivatised iron oxide nanoparticles: influence on
fibroblasts in vitro. Biomaterials 2003, 24, 4551–4557.
Recent advances in Magnetic Nanoparticles
Bi, F.; Zhang, J.; Su, Y.; Tang, Y.C.; Liu, J.N. Chemical
Due to magnetic forces, generally being short ranged and conjugation of urokinase to magnetic nanoparticles for
underwhelming compared to hydrodynamic forces in the targeted thrombolysis. Biomaterials 2009, 30, 5125–5130.
body, the targeted collection of magnetic nanoparticles has
met with limited success. So, an attempt to overcome its Chen, D.H.; Liao, M.H. Preparation and characterization of
YADH-bound magnetic nanoparticles. J. Mol. Catal. B
shortcomings, the notion
Enzymatic 2002, 16, 283–291.
T.K. Indira and P.K. Lakshmi : Magnetic Nanoparticles – A Review 1041

Chen, J.; Wu, H.; Han, D.; Xie, C. Using anti-VEGF McAb and Jia, Z.; Yujun, W.; Yangcheng, L.; Jingyu, M.; Guangsheng, L. In
magnetic nanoparticles as double-targeting vector for the situ preparation of magnetic chitosan/Fe3O4 composite
radioimmunotherapy of liver cancer. Cancer Lett. 2006, 231, nanoparticles in tiny pools of water-in-oil microemulsion.
169-175. React. Funct. Polym. 2006, 66, 1552–1558.
Conroy, S.; Jerry Lee, S.H.; Zhang, M. Magnetic nanoparticles in Lee, D.K.; Kang, Y.S. Preparation and characterization of
MR imaging and drug delivery. Adv. Drug Deliv. Rev. 2008, magnetic nanoparticles by ٧-irradiation. Mater. Sci. Eng. C
60, 1252–1265. 2004, C 24, 107-111.
Cregg, P.J.; Murphy, K.; Mardinoglu, A. Inclusion of magnetic Li, G.Y.; Huang, K.L.; Jiang, Y.R.; Ding, P.; Yang, D.L.
dipole–dipole and hydrodynamic interactions in Implant- Preparation and characterization of carboxyl functionalization
assisted magnetic drug targeting. J. Magn. Magn. Mater. of chitosan derivative magnetic nanoparticles. Biochem. Eng.
2009, 321, 3893–3898. J. 2008, 40, 408–414.
Dormer, K.; Seeney, C.; Lewelling, K.; Lian, G.; Gibson, D.;
Li, G.Y.; Huang, K.L.; Jiang, Y.R.; Ding, P.; Yang, D.L.
Johnson, M. Epithelial internalization of superparamagnetic
Preparation and characterization of Saccharomyces cerevisiae
nanoparticles and response to external magnetic field.
alcohol dehydrogenase immobilized on magnetic
Biomaterials 2005, 26, 2061–2072.
nanoparticles. Int. J. Biol. Macromol. 2008, 42, 405–412.
Du, X.; Inokuchi, M.; Toshima, N. Preparation and
Li, G.Y.; Jiang, Y.R.; Huang, K.L.; Ding, P.; Chen, J. Preparation
characterization of Co–Pt bimetallic magnetic nanoparticles.
and properties of magnetic Fe3O4–chitosan nanoparticles. J.
J. Magn. Magn. Mater. 2006, 299, 21-28.
Alloys Compd. 2008, 466, 451-456.
Fu, W.; Yang, H.; Bala, H.; Liu, S.; Li, M.; Zou, G. Preparation
and magnetic characterization of core–shell structure stainless Li, G.Y.; Jiang, Y.R.; Huang, K.L.; Ding, P.; Yao, L.L. Kinetics
steel/silica nanoparticles. Mater. Lett. 2006, 60: 1728–1732. of adsorption of Saccharomyces cerevisiae mandelated
dehydrogenase on magnetic Fe3O4–chitosan nanoparticles.
Gaihrea, B.; Khil, MS.; Lee, D.R.; Kim, H.Y. Gelatin-coated Colloids Surf. A: Physicochem. Eng. Aspects 2008, 320, 11-
magnetic iron oxide nanoparticles as carrier system: Drug 18.
loading and in vitro drug release study. Int. J. Pharm. 2009,
365, 180-189. Ma, H.L.; Qi, X.R.; Maitani, Y.; Nagai, T. Preparation and
characterization of superparamagnetic iron oxide
Garcia-Cerda, L.A.; Torres-Garcia, V.A.; Matutes-Aquino, J.A.;
nanoparticles stabilized by alginate. Int. J. Pharm. 2007, 333,
Ayala-Valenzuela, O.E. Magnetic nanocomposites:
177-186.
preparation and characterization of Co-ferrite nanoparticles in
a silica matrix. J. Alloys Compd. 2004, 369, 148–151. Mak, S.Y.; Chen, D.H. Fast adsorption of methylene blue on
polyacrylic acid-bound iron oxide magnetic nanoparticles.
Gupta, A.K.; Gupta, M. Synthesis and surface engineering of iron
Dyes and Pigments 2004, 61, 93–98.
oxide nanoparticles for biomedical applications. Biomaterials
2005, 26, 3995–4021. Martin, A.R.; Finlay, W.H. Enhanced deposition of high aspect
Hong, R.Y.; Feng, B.; Chen, L.L.; Liu, G.H.; Li, H.Z.; Zheng, Y.; ratio aerosols in small airway bifurcations using magnetic
Wei, D.G. Synthesis, characterization and MRI application of field alignment. J. Aerosci. 2008, 39, 679 – 690.
dextran-coated Fe3O4 magnetic nanoparticles. Biochem. Eng. Morais, P.C.; Santos, R.L.; Pimenta, AC.M.; Azevedo, R.B.;
J. 2008, 42, 290-300. Lima, E.C.D. Preparation and characterization of ultra-stable
Hong, R.Y.; Zhang, S.Z.; Di, G.Q.; Li, H.Z.; Zheng, Y.; Ding, J.; biocompatible magnetic fluids using citrate-coated cobalt
Wei, D.G.; Preparation, characterization and application of ferrite nanoparticles. Thin Solid Films 2006, 515, 266-270.
Fe3O4/ZnO core/shell magnetic nanoparticles. Mater. Res. Munnier, E.; Jonathan, S.C.; Linassier, C.; Eyrolles, L.D.;
Bull. 2008, 43, 2457–2468. Marchais, H.; Herve, S.K.; Dubois, P.; Chourpa, I. Novel
Huang, G.; Diakur, J.; Xu, Z.; Wiebe, L.I. Asialoglycoprotein method of doxorubicin–SPION reversible association for
receptor-targeted superparamagnetic iron oxide nanoparticles. magnetic drug targeting. Int. J. Pharm. 2008, 363, 170–176.
Int. J. Pharm. 2008, 360, 197–203.
Ning, L.; Xiaojie, L.; Xiaohong, W.; Honghao, Y.; Fei, M.; Wei,
Ito, A.; Shinkai, M.; Honda, H.; Kobayashi, T. Medical S.; Preparation and magnetic behavior of carbon-encapsulated
Application of Functionalized Magnetic Nanoparticles. J. iron nanoparticles by detonation method. Comp. Sci.
Biosci. Bioeng. 2005, 100, 1-11. Technol. 2009, 69: 2554–2558.
Jain, T.K.; Richey, J.; Strand, M.; Pelecky, D.L.L.; Flask, C.A.; Qiu, J.D.; Xiong, M.; Liang, R.P.; Peng, H.P.; Liu, F. Synthesis
Labhasetwar, V. Magnetic nanoparticles with dual functional and characterization of ferrocene modified Fe3O4@Au
properties: Drug delivery and magnetic resonance imaging. magnetic nanoparticles and its application. Biosens.
Biomaterials 2008, 29, 4012–4021. Bioelectron. 2009, 24, 2649–2653.
1042 International Journal of Pharmaceutical Sciences and Nanotechnology Volume 3 • Issue 3 • October-December 2010

Schulze, K.; Koch, A.; Schopf, B.; Petri, A.; Steitz, B.; Xu, H.; Song, T.; Bao, X.; Hu, L. Site-directed research of
Chastellain, M.; Hofmann, M.; Hofmann, H.; Rechenberg, magnetic nanoparticles in magnetic drug targeting. J. Magn.
B.V.; Intraarticular application of superparamagnetic Magn. Mater. 2005, 293, 514–519.
nanoparticles and their uptake by synovial membrane—an Xu, J.; Yang, H.; Fu, W.; Du, K.; Sui, Y.; Chen, J.; Zeng, Y.; Li,
experimental study in sheep. J. Magn. Magn. Mater. 2005, M.; Zou, G.; Preparation and magnetic properties of
293, 419–432. magnetite nanoparticles by sol–gel method. J. Magn. Magn.
Shaw, S.Y.; Chen, Y.J.; Ou, J.J.; Ho, L. Preparation and Mater. 2007, 309, 307-311.
characterization of Pseudomonas putida esterase immobilized Yamaura, M.; Camilo, R.L.; Sampaio, L.C.; Macedo, M.A.;
on magnetic nanoparticles. Enzyme Microb. Technol. 2006, Nakamura, M.; Toma, H.E. Preparation and characterization
39, 1089–1095. of (3-aminopropyl) triethoxysilane-coated magnetite
nanoparticles. J. Magn. Magn. Mater. 2004, 279, 210-217.
Shen, Y.F.; Tang, J.; Nie, Z.H.; Wang, Y.D.; Renc, Y.; Zuo, L.
Preparation and application of magnetic Fe3O4 nanoparticles Yang, J.; Park, S.B.; Geun Yoon H.; Huh, Y.M.; Haam, S.
for wastewater purification. Sep. Purif. Technol. 2009, 68, Preparation of poly ε-caprolactone nanoparticles containing
312–319. magnetite for magnetic drug carrier. Int. J. Pharm. 2004, 324,
185–190.
Sun, J.B.; Duan, J.H.; Dai, S.L; Ren, J.; Zhang, Y.D.; Tian, J.S.;
Li, Y. In vitro and in vivo antitumor effects of doxorubicin Yang, Y.X.; Liu, M.L.; Zhu, H.; Chen, Y.R.; Mu, G.J.; Liu, X.N.;
loaded with bacterial magnetosomes (DBMs) on H22 cells: Jia, Y.Q. Preparation, characterization, magnetic property,
The magnetic bio-nanoparticles as drug carriers. Cancer Lett. and Mossbauer spectra of the β-FeOOH nanoparticles
2007, 258, 109-117. modified by nonionic surfactant. J. Magn. Magn. Mater.
2008, 320, L132– L136.
Tartaj, P.; Morales, M.P.; Veintemillas-Verdaguer, S.; Teresita
Yong, Y.; Bai, O.; Li, Y.; Lin, L.; Cui, Y.; Xi, C. Preparation and
Gonzalez-Carreno, T.; Carlos Serna, J. The preparation of
application of polymer-grafted magnetic nanoparticles for
magnetic nanoparticles for applications in biomedicine. J.
lipase immobilization. J. Magn. Magn. Mater. 2008, 320,
Phys. D: Appl. Phys. 2003, 36, R182–R197.
2350– 2355.
Vekas, L.; Bica, D.; Avdeev, M.V. Magnetic nanoparticles and
Zhang, J.; Misra, R.D.K. Magnetic drug-targeting carrier
concentrated magnetic nanofluids: Synthesis, properties and encapsulated with thermosensitive smart polymer: Core–shell
some applications. China Particu. 2007, 5, 43-49. nanoparticle carrier and drug release response. Acta
Wang, S.; Yang, Z.; Sun, W. Preparation and characterization of Biomater. 2007, 3, 838–850.
antifouling thermosensitive magnetic nanoparticles for Zhang, L.Y.; Zhu, X.J.; Sun, H.W.; Chi, G.R.; Xu, J.X.; Sun,
applications in biomedicine, Mater. Sci. Engin. C 2009, C Y.L. Control synthesis of magnetic Fe3O4–chitosan
(29), 1196–1200. nanoparticles under UV irradiation in aqueous system. Curr.
Wang, X.; Wang, L.; He, X.; Zhang, Y.; Chen, L. A molecularly Appl. Phys. 2010, 10, 828–833.
imprinted polymer-coated nanocomposite of magnetic Zhao, Y.; Qiu, Z.; Huang, J. Preparation and Analysis of Fe3O4
nanoparticles for estrone recognition. Talanta. 2009, 78, 327– Magnetic Nanoparticles Used as Targeted-drug Carriers.
332. Chinese J. Chem. Eng. 2008, 16 (3), 451-455.
Wu, Y.; Wang, Y.; Luo, G.; Dai, Y. In situ preparation of Zhu, L.; Ma, J.; Jia, N.; Zhao, Y.; Shen, H.; Chitosan-coated
magnetic Fe3O4-chitosan nanoparticles for lipase magnetic nanoparticles as carriers of 5-Fluorouracil:
immobilization by cross-linking and oxidation in aqueous Preparation, characterization and cytotoxicity studies.
solution. BioRes. Technol. 2009, 100, 3459–3464. Colloids Surf. B: Biointerfaces 2009, 68, 1-6.

You might also like