You are on page 1of 20

Advanced Drug Delivery Reviews 63 (2011) 789–808

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a d d r

Magnetic nanomaterials for hyperthermia-based therapy and controlled drug delivery


Challa S.S.R. Kumar a,⁎, Faruq Mohammad a,b
a
Center for Advanced Microstructures & Devices, Louisiana State University, 6980 Jefferson Highway, Baton Rouge, LA 70806, USA
b
Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA 70813, USA

a r t i c l e i n f o a b s t r a c t

Article history: Previous attempts to review the literature on magnetic nanomaterials for hyperthermia-based therapy focused
Received 12 November 2010 primarily on magnetic fluid hyperthermia (MFH) using mono metallic/metal oxide nanoparticles. The term
Accepted 22 March 2011 “hyperthermia” in the literature was also confined only to include use of heat for therapeutic applications.
Available online 5 April 2011
Recently, there have been a number of publications demonstrating magnetic nanoparticle-based hyperthermia
to generate local heat resulting in the release of drugs either bound to the magnetic nanoparticle or
Keywords:
Hyperthermia
encapsulated within polymeric matrices. In this review article, we present a case for broadening the meaning of
Hyperthermia-based therapy the term “hyperthermia” by including thermotherapy as well as magnetically modulated controlled drug
Hyperthermia-based controlled drug delivery delivery. We provide a classification for controlled drug delivery using hyperthermia: Hyperthermia-based
Core–shell magnetic nanoparticles controlled drug delivery through bond breaking (DBB) and hyperthermia-based controlled drug delivery
Theranostics through enhanced permeability (DEP). The review also covers, for the first time, core–shell type magnetic
nanomaterials, especially nanoshells prepared using layer-by-layer self-assembly, for the application of
hyperthermia-based therapy and controlled drug delivery. The highlight of the review article is to portray
potential opportunities for the combination of hyperthermia-based therapy and controlled drug release
paradigms -towards successful application in personalized medicine.
© 2011 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 790
2. Mechanism of magnetic nanomaterials-based hyperthermia: implications for therapy and controlled drug delivery . . . . . . . . . . . . . 792
3. Magnetic nanomaterials for hyperthermia-based therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 793
3.1. Size and size-distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 793
3.2. Carrier viscosity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
3.3. Magnetic anisotropy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
3.4. Stabilizing ligand . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
3.5. Ability for tumor size reduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
3.6. Self-regulated hyperthermia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
3.7. Multi-functionality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
4. Magnetic nanomaterials for hyperthermia-based controlled drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 796
4.1. Hyperthermia-based controlled drug delivery through bond breaking (DBB) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 796
4.2. Hyperthermia-based controlled drug delivery through enhanced permeability (DEP) . . . . . . . . . . . . . . . . . . . . . . . . 796
5. Core–shell magnetic nanomaterials for hyperthermia-based therapy and controlled drug delivery . . . . . . . . . . . . . . . . . . . . . 798
5.1. Why core–shell magnetic nanoparticles? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 798
5.2. Core–shell magnetic nanomaterials for hyperthermia-based therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 799
5.2.1. Prevent anisotropic magnetic dipolar attraction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 799
5.2.2. Provide oxidative stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 799
5.2.3. Enhanced hyperthermia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 799
5.2.4. Reduce toxicity and increase biocompatibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 800
5.3. Core–shell magnetic nanomaterials for hyperthermia-based controlled drug delivery . . . . . . . . . . . . . . . . . . . . . . . . 800
6. MNP-based hyperthermia for therapy and controlled drug delivery: current challenges and opportunities . . . . . . . . . . . . . . . . . . 801
6.1. Maximizing specific absorption rate (SAR) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 801
6.2. Tailoring the properties of magnetic nanomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 801

⁎ Corresponding author.
E-mail address: ckumar1@lsu.edu (C.S.S.R. Kumar).

0169-409X/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2011.03.008
790 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

6.3. Eliminating or minimizing toxicity from NPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 801


6.4. Biocompatibility, catabolism and clearance issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 801
6.5. Induce and sustain therapeutic temperatures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 801
6.6. Monitoring temperature distribution during heating . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802
6.7. Visualization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802
6.8. Theranostics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802
6.9. Thermo tolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802
6.10. Tolerance of magnetic field and frequency of oscillation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802
6.11. Self-regulation of heating . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802
6.12. Practical targeted hyperthermia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803
6.13. Antitumor immunity after hyperthermia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803
6.14. Combination of treatments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803
6.15. Multi-therapeutic and multi-functional modalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 803
6.16. Drug loading and stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 804
6.17. Influence of nature and size of the shell . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805

1. Introduction infrared radiation, magnetically excitable thermoseeds, and tubes with


hot water. However, each of these methods suffers from its own
The etymological meaning of the word “hyperthermia” is generation limitations [10]. Oncologists often use the heat treatment in combina-
of heat and with respect to cancer therapy, the term is used to imply tion with radiotherapy or chemotherapy or both. The combined
treatment based on generation of heat at the tumor site [1]. The approach results in eliminating many cancer cells in addition to making
approach involves raising the temperature of local environment of a the resistant cancer cells more vulnerable to other treatments. Some of
tumor resulting in changing the physiology of diseased cells finally the challenges in traditional hyperthermia treatment are [11]: 1) un-
leading to apoptosis [2]. This treatment modality complements avoidable heating of healthy tissue resulting in burns, blisters and
currently available treatments including chemotherapy, radiation discomfort, 2) limited penetration of heat into body tissues by
therapy, surgery, gene therapy, and immunotherapy for cancer. microwave, laser and ultrasound energy, and 3) thermal under-dosage
Depending on the degree of temperature raise, hyperthermia treatment in the target region, a nearly unsolved problem in the case of bone of
can be classified into different types. In thermo ablation, a tumor is pelvis or scull which shield deep tissues; often yielding recurrent tumor
subjected to high temperatures of heat N46 °C (up to 56 °C) causing cells growth.
to undergo direct tissue necrosis, coagulation or carbonization. With the possibility to convert dissipated magnetic energy into
Moderate hyperthermia (41 °C b Tb46 °C) has various effects both at thermal energy, the application of magnetic materials for hyperther-
the cellular and tissue levels. Diathermia uses lower temperatures mia treatment of cancer was first proposed in 1957 [12]. Since then the
(Tb 41 °C) for the treatment of rheumatic diseases in physiotherapy. approach evolved into a well-researched field due to the introduction
During moderate hyperthermia, which is traditionally termed as of magnetic nanoparticles (MNPs). MNP-based hyperthermia treat-
hyperthermia treatment, cells undergo heat stress in the temperature ment has a number of advantages compared to conventional hyper-
range of 41–46 °C resulting in activation and/or initiation of many intra thermia treatment. These, schematically highlighted in Fig. 1, are:
and extracellular degradation mechanisms like protein denaturation, 1) cancer cells absorb MNPs thereby increasing the effectiveness of
protein folding, aggregation and DNA cross linking. With a single heat hyperthermia by delivering therapeutic heat directly to them, 2) MNPs
treatment, permanent irreversible protein damage can occur resulting can be targeted through cancer-specific binding agents making the
in protein aggregation and/or inhibition of many cellular functions [3]. treatment much more selective and effective, 3) the frequencies of
The other cellular effects of moderate hyperthermia include induction oscillating magnetic fields generally utilized pass harmlessly through
and regulation of apoptosis, signal transduction, multidrug resistance the body and generate heat only in tissues containing MNPs [13],
and heat shock protein (HSP) expression. The tissue level effects include 4) MNPs can also effectively cross blood-brain barrier (BBB) and hence
pH changes, perfusion and oxygenation of tumor micro environment can be used for treating brain tumors, 5) effective and externally
[4–6]. The effectiveness of any hyperthermia treatment greatly depends stimulated heating can be delivered at cellular levels through
on the temperatures generated at the targeted sites of action, duration of alternating magnetic field (AMF) [14], 6) with the possibility to obtain
exposure and characteristics of particular cancer cells [7]. stable colloids using MNPs, they can be administered through a number
Hyperthermia is also categorized into local, regional and whole body of drug delivery routes [15], 7) MNPs used for hyperthermia are only
hyperthermia depending on the location of disease. Local hyperthermia few tens of nanometer in size and therefore, allows easy passage into
involves subjection of heat only to a small area of interest such as a several tumors whose pore sizes are in 380–780 nm range [16],
tumor. Regional hyperthermia involves heat subjection to larger areas 8) compared to macroscopic implants, MNP-based heat generation is
such as whole tissue and organ. Whole body hyperthermia is applied to much more efficient and homogeneous [17], 9) MNP-based hyperther-
treat metastatic cancer cells when spread throughout the body. The mia treatment may induce antitumoral immunity [18], and 10) last but
challenge here is to heat only the tumor cells without damaging most important aspect is that MNP-based hyperthermia can also be
the healthy tissues. Of these, local hyperthermia is gaining much more utilized for controlled delivery of drugs (Sections 4-6) and first such
attention due to intracellular heat subjection within specified region of nanoconstruct for this purpose was made using layer-by-layer self-
interest [8,9]. assembly approach [19]. This additional feature opens up possibilities
Traditionally, hyperthermia treatment was administered by using for the development of multifunctional and multi-therapeutic ap-
external devices to transfer energy to tissues either by irradiation with proaches for treating a number of diseases.
light or electromagnetic waves. Currently available techniques for There have been an increasing number of investigations, in the
induction of hyperthermia are ultrasound, radiofrequency, microwaves, past two decades, into various aspects of magnetic nanomaterials for
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 791

Fig. 1. A schematic representation of some of the unique advantages of magnetic nanomaterials for hyperthermia-based therapy and controlled drug delivery.

hyperthermia-based therapy. While there are few review articles in Nanoparticles (NPs) offer an opportunity to create multifunction-
the literature, their focus has been primarily on either iron oxide ality with potential for innovative diagnostic and therapeutic
nanoparticle or monometallic/oxide MNP-based hyperthermia. For modalities for a number of diseases [26]. Controlled release of drugs
example, in the most recent review [20] an attempt was made to with spatiotemporal control is the key for meeting a number of
present the status and prospects of magnetite nanoparticle-based challenges in drug delivery applications [27]. Controlled release of
hyperthermia covering only theoretical and experimental investiga- drugs from NP based drug delivery systems, triggered by a number of
tions related to heat dissipation by Néel relaxation. There is a review external stimuli, has been extensively studied [28]. While the term
that covers clinical applications of MNPs for hyperthermia with focus “hyperthermia” in the literature, so far has been confined only to
on recent clinical trials conducted by MagForce Nanotechnologies AG, include the use of heat for therapy, it was found from the recent
Berlin, Germany [21]. In yet another mini review, Hergt et al. discuss literature studies that MNP-based hyperthermia can also be utilized to
how MNP properties and electromagnetic field parameters may be generate intense local heating within polymeric matrices thereby
optimized for cancer therapy based on magnetic fluid hyperthermia creating voids for the release of encapsulated drugs (Fig. 1). The use
(MFH) [22]. On similar lines, fundamental aspects related to the of magnetic fields to control drug release from large polymeric
design of MNPs for cancer-localized hyperthermia was reviewed; matrices was first reported nearly three decades back [29–31]. These
once again focusing on iron oxide-based MNPs [23]. While single investigations conceptually demonstrated the utility of magnetic field
domain MNPs absorb much more power at tolerable magnetic fields hyperthermia for controlled drug release albeit using millimeter size
and frequencies than those by multidomain particles, optimization of magnetic materials. The very first report of potential opportunity to
MNPs for MFH is not yet well established [24]. There is a strong need utilize MNPs as hyperthermic agents for controlled drug release
for the design of MNPs with enhanced magnetic properties specifi- appeared in 2005 [19]. The drug delivery system in this case was
cally tailored for hyperthermia related applications. There is also an prepared by using layer-by-layer self-assembly technique. Since then a
unmet technical challenge to design new MNPs which possess not number of investigations have been carried out to demonstrate this
only high thermal efficiency as heating elements but are also concept in a number of materials and for a number of application types.
biocompatible (nontoxic), and stable in aqueous solution. In addition, A broad range of frequencies of oscillating magnetic fields, from 50 kHz
the expectations are that they should have high capacity for to 10 MHz, were utilized to demonstrate the concept of magnetically
accumulation inside the tumor cells and with right Curie temperature modulated release of drugs using MNP encapsulated delivery systems
(TC) to provide local temperature control system [25]. [19,32]. A possible mechanism based on the creation of nanopores
792 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

within a polymeric matrix through generation of heat by switching on/ The internal (Néel) and external (Brownian) sources of friction that
off of magnetic moments was proposed. The effect was also found to be lead to a phase lag between applied magnetic field and the direction of
physically reversible upon short-term field exposure and irreversible the magnetic moments tends to generate thermal losses. By using
under long term exposure [19,33]. Similar to MNPs investigated for linear response models with known Néel and Brownian relaxation
hyperthermia treatment, majority of the investigations related to use times, one can easily predict SPL values for MNPs [38]. One
of hyperthermia for controlled release have been utilizing iron oxide predominant heating mechanism that governs heating capacity of a
NPs. In a recent review by Brazel [34], an attempt was made to analyze given MNP is based on the induction of rapid variation of magnetic
magnetothermally triggered drug delivery systems. However, the moments. In general, the SPL values increase with frequency of applied
scope of the article was limited to MNP encapsulated polymeric magnetic field and are proportional to the square of magnetic field
systems. intensity. SPL values are measured in terms of rise in temperature per
In this review, we present a case for broadening the meaning of the unit time and per gram of magnetic material, multiplied by calorific
term “hyperthermia” by including therapy as well as magnetically capacity of a sample. Fig. 2 shows that magnetization, M lags in phase
modulated controlled drug delivery through heating. We review behind applied field, H (a) and relaxation happens through two major
current literature on MNP-based hyperthermia for both thermother- pathways (b and d). One can also see that the real part, χ′ or the
apy as well as controlled drug release in order to provide a better in-phase component and the imaginary part, χ″ or the loss component
understanding on the present status as well as future perspectives. The of the susceptibility is a function of frequency, ν (c) and χ″ is
review also focuses for the first time on the application of core–shell maximum when the angular frequency ω(=2πν) = 1/τ where τ is the
type magnetic nanomaterials for hyperthermia-based therapy and relaxation time [57].
controlled drug delivery. For the purpose of this review, core–shell Generally SPL values depend on parameters such as MNPs structure
MNPs are those that have a core and a shell made up of different metals (size, shape and crystal structure), magnetic properties (magnetic
or oxides. Polymers or surfactant stabilized MNPs are not considered anisotropy and temperature dependence of magnetizations) and
truly as core–shell MNPs under this definition. Since there are a large amplitude (H) and frequency (f) of AMF [39–41]. Reduction in SPL
number of review articles covering synthesis of core–shell MNPs, values with polydispersity of MNPs is observed and this can be due to
especially those using layer-by-layer self-assembly approach [35], we decrease in the proportion of particles contributing to total heat
will only make references to their synthesis where the materials generation. The heating power of many MNPs also change with
have been synthesized specifically for the purpose of hyperthermia- surrounding environment e.g. for those particles internalized within
based treatment or controlled drug release. We will dwell on targeted the cells. In such cases, the nuclear endosomes and/or other
hyperthermia and spatiotemporal drug delivery with potential intracellular components generally hinder the movement of the
opportunities for cancer treatment. We will also discuss how the particles resulting in total heat contribution largely coming only
core–shell MFH offers a unique possibility to create materials that from Néel relaxation [42]. Therefore, for intracellular MFH, Néel
combine the potential of magnetic properties with required hyper- relaxation is the major contributor for heat release. The most effective
thermic and biological characteristics facilitating a safe and effective AMF parameters for highest reported to date for hyperthermia
treatment. Finally, we hope to bring out a message that a combination applications are a frequency of 500 kHz at the field amplitude of
of hyperthermia-based therapy and controlled drug release paradigms 10 kA/m [43].
has potential to succeed as “intelligent theranostics” for successful In the case of MNPs for hyperthermia-based controlled drug
application in personalized medicine. delivery, one can postulate two distinct types of mechanisms (Fig. 3).
In the first type, a drug molecule is attached to MNP through a linker
2. Mechanism of magnetic nanomaterials-based hyperthermia: and on application of an AMF, the drug molecule is released due to
implications for therapy and controlled drug delivery heating of the linker molecule attached to the NP's surface [44]. By
varying the power of electromagnetic field (EMF) pulses, it is possible
The absorption efficiency of any material to generate heat due to to arrive at complex release profiles to release multiple drugs in a
AMF is measured in terms of specific absorption rate (SAR) or specific series, or in combinations. This approach, represented schematically
loss power (SLP). These terms are generally used to define the in Fig. 3a, can be termed as hyperthermia-based controlled drug
transformation of magnetic energy into heat [36]. For a majority of delivery through bond breaking (DBB). In the second type, release of
applications, it is desirable to have higher temperature enhancement drugs takes place from within a polymeric matrix (nano/microparti-
rates and MNPs are proving to be far superior to micrometric particles cles/thin films) encapsulated with MNPs on application of AMF/EMF
due to their efficiency in conversion of magnetic energy into heat even (Fig. 3b) [19,29]. A possible mechanism for this type of hyperthermia-
at low concentrations [37]. based drug delivery is formation of crevices or cracks of nanometer
For MNP based hyperthermia, a general procedure involves scale within a polymeric matrix due to local heat generated by the
distribution of particles throughout the targeted tumor site, followed MNPs, thereby releasing encapsulated drugs. The second type of
by generation of heat to the tumor using an external AMF. The dynamic controlled drug release can be termed as hyperthermia-based
response of a dipole with its magnetic moment in a single direction controlled drug delivery through enhanced permeability (DEP).
due to an external AMF during the transformation of magnetic energy Depending on the nature of the polymer, hyperthermia-based drug
into heat is governed mainly by thermal fluctuations that occur in a release can be due to creation of a mechanically force openings or
particle. While there are a number of effects occurring in MNPs, the thermally responsive openings in the case of thermoresponsive poly-
heat generation mechanism can be attributed to two different mers [34]. The change in dimension of nanocrevices to a certain degree
phenomena: relaxation and hysteresis loss (Fig. 2). The relaxation is is physically reversible upon short-term field exposure. However,
of two types: Néel and Brownian relaxations. Heat generation through following long-term exposure, the nanocrevices further enlarge to
Néel relaxation is due to rapidly occurring changes in the direction of nanometer-scale cracks that propagate along the spherical shell
magnetic moments relative to crystal lattice (internal dynamics). This structure and ultimately form irreversible deformations. Having
is hindered by energy of anisotropy that tends to orient magnetic absorbed sufficient amounts of magnetic energy, the shell ruptures
domain in a given direction relative to crystal lattice. Brownian resulting in increase of both pore volume and surface area. As for the
relaxation is due to physical rotation of particles within a medium frequency dependency of permeability, the aggregates rotate or oscillate
in which they are placed (external dynamics) and is hindered by under the action of magnetic field with elastic deformation of capsule
the viscosity that tend to counter the movement of particles in the walls. At high frequencies, they will not follow variation of magnetic
medium [20]. fields and the deformation will be minimal leading to a decrease in
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 793

Fig. 2. A schematic representation of possible mechanisms for conversion of magnetic energy into heat.
Modified based on Fig. 13 from Ref. [57].

permeability. This is similar to the case of forced oscillation of a by heme oxygenase-1 to form blood hemoglobin and hence maintain
pendulum at frequencies above the resonance frequency. iron cell homeostasis by cells [52,53]. In addition, magnetite was found
to be superior to cobalt NPs with respect to its high Curie temperature,
3. Magnetic nanomaterials for hyperthermia-based therapy saturation magnetization (Ms) (90–98 emu/g, or ~450–500 emu/cm3)
and lower toxicity in preclinical tests [25,48]. Deger et al. tested a
A number of types of magnetic nanomaterials, ranging from well combination of hyperthermia with 3D conformal radiotherapy using
known and well investigated iron oxide-based nanomaterials to Co–Pd thermoseeds for the treatment of prostate cancer and the intra-
metallic NPs such as Mn, Fe, Co, Ni, Zn, Gd, Mg, and their oxides have prostatic temperatures achieved were about 42–46 °C with no side
been investigated for their hyperthermic potential. Some of the well effects [54]. There are theoretical studies predicting that hyperthermia is
known hyperthermic agents based on iron oxide are magnetite NPs dependent on particle size, size distribution, magnetic properties such
(Fe3O4) stabilized by a variety of ligands such as dextran [45], cationic as magnetic moment and magnetic anisotropy, and viscosity of fluid in
liposomes [46], polyvinyl alcohol, hydrogel [47], lauric acid [25,48] and which they are dispersed [55]. It is instructive to see how the influence
maghemite NPs (γ-Fe2O3) [47] stabilized by ligands such as dextran of these features has been validated experimentally as well. A number
[45]. Yet another category is based on ferrites such as cobalt ferrites of factors influencing the design of MNPs for optimizing their
(CoFe2O4), manganese ferrite (MnFe2O4), nickel ferrite (NiFe2O4), hyperthermic potential have been reported and these are reviewed as
lithium ferrite (Li0.5Fe2.5O4), mixed ferrites of nickel–zinc–copper described below.
(Ni0.65Zn0.35Cu0.1Fe1.9O4) and cobalt–nickel ferrite (CoxNi(1 − x)Fe2O4)
[25,48,49]. There are also ferromagnetic NPs such as Fe doped Au, 3.1. Size and size-distribution
Zn–Mn doped iron oxides (ZnxMn(1 − x)Fe3O4) and Mn–Zn–Gd doped
iron oxide (MnxZnxGdxFe(2 − x)O4) composites [50]. Very recently, The heating power of MNPs is governed by magnetic energy
extremely high heating performance of 1300–1600 W/g was reported dissipation, and therefore, is dependent on the size of MNPs. Size-
based on FeCo metallic NPs [51]. However, iron oxide-based MNPs dependent hyperthermia of highly crystalline and monodisperse iron
continue to attract attention due to their lack of toxicity, excellent oxide NPs showed that the heating rates with highest SLP of 447 W/g
biocompatibility, and in addition to the fact that they can be metabolized was achieved for 14 nm particle when particles in size range 5–15 nm
794 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

Fig. 3. Schematic representation of the two types of controlled drug delivery using magnetic nanoparticle-based hyperthermia.

were screened with a field amplitude of 24.5 kA/m and a frequency of experimental and theoretical values of SLP as a function of particle
400 kHz [56]. Fig. 4 shows the effect of size and size-distribution in size and polydispersity indices (Fig. 4(c) and (d)) [57]. On the other
magnetite nanocrystals of four different sizes. The size and size- hand, at the same field amplitude of 24.5 kA/m but at a higher
distributions, shown in Fig. 4(a), were obtained based on TEM and frequency of 700 kHz, maghemite crystals showed size-dependent
from fittings of the magnetization curves. The variation in their SPL SLP with the particle size of 16.5 nm having the highest SLP of
values on application of oscillating magnetic field of 400 kHz is 1650 W/g. In comparison, cobalt ferrite of 12 nm showed highest
shown in Fig. 4(b). It is interesting to see an agreement between SLP of 420 W/g when sizes 6–15 nm were compared for their SLPs.

Fig. 4. Size-dependent hyperthermia of MNPs.


Reproduced from Fig. 14 in Ref. [57].
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 795

Extending the size-dependent studies to larger particles (with sizes in performance with respect to tumor size reduction. Recently, a number
the range 15–50 nm), the heating efficiency shows a maximum of of in vivo studies using animal models were undertaken where MNPs
950 W/g for a 15 nm particle [47]. These results are once again in were injected into a tumor followed by AMF exposure and resulting
consistent agreement with both theoretical predictions as well as changes in the tissue and organs were investigated. These include C3H
experimentally generated size-dependent magnetization values for mouse mammary carcinoma [63], mouse EL4 T-lymphoma [11], MX11
highly crystalline and monodispersed iron oxide nanocrystals [58]. mouse sarcoma [64], human prostate cancerous and bone marrow
Overall, the size-dependent investigations reported to date clearly cells in transgenic mice [46], EL4 mouse T-lymphoma [65], T-9 rat
demonstrate that single domain iron oxide MNPs with monodispersed glioma cells [66], B16 mouse melanoma [67], MM46 mammary and
size of 14–16 nm are the most superior. However, what is surprising is skin carcinoma [68], murine B16-F10 mouse melanoma [69], cat
the finding that bacterial magnetosomes consisting of ~ 30 nm mammary tumor gland [70], Os515 hamster osteosarcoma [71], VX-7
γ-Fe2O3, unlike those synthesized in the laboratory, have the highest squamous cell carcinoma in rabbit tongue [72], DMBA induced rat
SLP value of 960 W/g (at 410 kHz and 10 kA/m) reported to date [59]. mammary carcinoma [73], and human glioma cells [74]. Most of the
Yet another recent study [60] also showed high magnetic field studies though provided positive evidence for decrease in tumor size
induction heating of aqueous dispersions of meso-2,3-dimercapto- due to hyperthermia, there is a clear lack of complete information on
succinic acid stabilized single-crystalline iron oxide NPs. In this case, host and material response. Host response corresponds to influence of
SAR increased with increasing particle size until it peaked for particles living cells on the magnetic nanomaterial and vice versa for magnetic
with an average diameter of 10.5 nm and then decreased for particles nanomaterial's response on cells [75]. Also, the effect of MNP uptake
with an average diameter of 12.1 nm. Theoretical calculations of the by healthy cells and their fate before and after the application of
effect of particle size on the heat generation were in agreement with oscillating magnetic field is not clear. A better understanding of MNPs
this trend as well. These studies also showed that there was a decrease and their interactions with tissue, and organs are of greatest
in the contribution of Brownian relaxation to the heating, leading to a importance for optimizing hyperthermia treatment in clinical setting.
decrease in the SAR with larger particles. Extending size-dependent
investigations to alloy NPs, one can predict that the heating potential
of FePt and NiPd NPs is likely to change with their size [38]. 3.6. Self-regulated hyperthermia

3.2. Carrier viscosity A highly desirable form of hyperthermia is one that is self-regulated.
Self-regulation is possible with MNPs that have Curie temperature at
The heating rate in MFH is also dependent on carrier viscosity [20]. around therapeutic temperature range. With such particles, similar to
Surprisingly, not much work is reported on the influence of viscosity. traditional hyperthermia, they are heated when AC field is applied.
In a recent publication, SLP of monodispersed particles of maghemite However, once they reach the Curie temperature, the Ms of particles
and cobalt ferrite in two different solvents, water and glycerol, was drops to zero, and heating stops. Thus, if the Curie temperature can be
investigated. With field amplitude of 24.8 kA/m, it was demonstrated fixed by a judicious selection of particle composition and size,
that SLP decreased with increase in viscosity with the effect being hyperthermia can be applied and carefully controlled; preventing
more pronounced with cobalt ferrite than with maghemite NPs [40]. from reaching excessive temperatures. Some of the obvious choice of
materials for such a self-regulated therapy includes Ni–Cu alloys,
3.3. Magnetic anisotropy Mn–Cu ferrites, Ni–Pd alloys and Co–Pd alloys. Recent studies on
polymer encapsulated Cu–Ni NPs [76], Gd-substituted Mn–Zn ferrite
Yet another factor that is important for MNP-based hyperthermia NPs [77], Fe1 − xMnxFe2O4 NPs [78], and FePt, NiPt, NiPd [79] NPs are
is to increase the anisotropy of NPs (shape or magnetocrystalline) or very promising in this direction. The Curie temperature of these alloy
increasing the applied magnetic field strength used for treatment. systems can be tuned by changing their composition especially since
Even though high-quality MNPs with much higher anisotropies, such Fe27Pt73 and Ni28Pd72 alloys have Curie temperature near 45 °C [79]. The
as Co and Co-based alloys are readily synthesized, their known Curie temperature is also expected to change with size and certainly
toxicity prevents their utility in in vivo applications [25,61]. with composition of MNPs. The methods to make FePt and NiPd NPs of
different sizes (2–9 nm) are already available [79–82]. Therefore, by
3.4. Stabilizing ligand extending the known compositional dependence of Curie temperature
(TC) for the face centered cube (fcc)-phase at high Ni concentrations,
In order to study the influence of stabilizing ligand on biocompat- one can expect metastable FeNi alloys to have low TCs in the Fe-rich
ibility and cellular uptake in vitro, Pradhan et al. used different coatings of region of the phase diagram. Thus making them suitable for self-
dextran and lauric acid on Fe3O4 NPs. In their observation, there is a regulated radio frequency (Rf) heating in cancer hyperthermia [83].
lesser biocompatibility and higher uptake of lauric acid-coated magne-
tite NPs in comparison to that of dextran-coated NPs in L929 mouse
fibroblast cells. This is likely due to different cellular interactions caused 3.7. Multi-functionality
by the coating material [53]. In a similar study by Jordan et al. with
coatings of dextran and silane on magnetite NPs, differential endocytosis Demonstrating potential opportunities that exist for combining
occurred in terms of uptake due to the coating material. The study once hyperthermia with diagnosis, a multifunctional NP (b50 nm) with a
again confirms that biocompatibility is dependent on the nature of the Fe2O3 core and labeled with a targeting agent luteinizing hormone
coating material and NP-cell interaction [62a,62b]. In addition to releasing hormone (LHRH) was designed. The multi-functionality of
providing bio-compatability, the stabilizing ligands could also alter the the nanoparticle ensured specific targeting to cancer cells of breast
magnetic properties of the nanoparticles as it was recently demonstrated and prostate in addition to having a two-photon fluorescent probe to
that there is a step-wise change in magnetism of magnetic nanoparticles aid in optical tracking [84]. Such a multifunctional NP, termed as
with step-wise change in their bio-functionalization [62c]. “nanoclinics” was used for magnetocytolysis of MCF-7 and UCI cancer
cells using a DC magnetic field in addition to providing optical imaging
3.5. Ability for tumor size reduction capability. Since the magnetocytolosys was performed using a 7 T
magnetic field NMR instrument, it is not clear what the frequencies of
While optimization of physical properties of MNPs to arrive at oscillating magnetic field are. In any case, this investigation is a step in
higher SLP values is important, it is also critical to evaluate their the right direction to bring multifunctionality to traditional MFH.
796 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

4. Magnetic nanomaterials for hyperthermia-based controlled bar = 100 μm). On further examination of the mice using a 7 T MRI
drug delivery scanner, one can see an image contrast due to the presence of NPs
(arrow). This magnetically stimulated platform for controlled drug
Drug release from the surface of NPs was previously accomplished by release demonstrates ability to remotely trigger release of a biomole-
external stimuli such as electric current, magnetic fields, temperature, cule, if necessary in a sequence, from the surface of MNPs. Yet another
light, and ultrasound and so on. In particular, the concept of using report [85] confirms similar concept by triggering release of fluorophore
external magnetic fields to achieve pulsatile release from polymer bimane amine from the surface of superparamagnetic iron oxide
composites was first demonstrated using millimeter size magnetic nanoparticles (SPIONs) in presence of oscillating magnetic fields.
particles. In this investigation, an externally controlled on-demand
insulin release was demonstrated from a magnetic composite of 4.2. Hyperthermia-based controlled drug delivery through enhanced
ethylene vinylacetate by application of low frequency oscillating permeability (DEP)
magnetic field [29]. This concept did not gain attention till recently
where modulation of permeability of polyelectrolyte microcapsule Unlike the concept of DBB, hyperthermia-based controlled drug
embedded with MNPs, fabricated using layer-by-layer self-assembly, delivery through enhanced permeability (DEP) involves release of
was first demonstrated [19]. Since then there have been a number of drug from within a polymeric nanoparticle (PNP) wherein MNPs and
reports in the literature related to application of magnetic nanomater- drug are encapsulated. DEP has been found to be reversible under
ials for hyperthermia-based controlled drug delivery. These can be certain conditions and is dependent on frequency of the oscillating
classified into two major types. magnetic field and the applied magnetic field strength. Such a release
was demonstrated from polymers, hydrogels and thermoresponsive
4.1. Hyperthermia-based controlled drug delivery through bond polymers.
breaking (DBB) Remotely controlled pulsatile drug release for a number of
different drugs as well as for different “on–off” durations of oscillating
The first successful demonstration of this concept was reported magnetic fields was demonstrated from temperature sensitive poly
using radiofrequency EMF activation of release of fluorescein-labeled (N-isopropylacrylamide) hydrogels incorporated with SPIONs [86].
18 bp single stranded DNA (a model antisense therapeutic) in a model Other hydrogel-based DEP systems are poly(N-isopropylacrylamide)
tumor near the posterior mammary fat pad of mice (Fig. 5) [44]. As (PNIPAM) microgel containing MNPs with the ability to tune
shown in the figure, first MNP bound biomolecules through a heat labile magnetic and thermoresponsive properties of individual components
linker are mixed with matrigel and injected subcutaneously near the (NPs and microgels) [87]. Extending this concept further, an approach
posterior mammary fat pad of mice, forming a model tumor (A). On to the development of artificial membranes with stimulus-responsive
application of EMF, the fluorescent biomolecules were released into opening was recently demonstrated [88]. On-demand “on–off” release
surrounding tissue (B) when compared to unexposed controls (C, scale of sodium fluorescein over multiple magnetic cycles was successfully

Fig. 5. Demonstration of the concept of DBB in vivo.


Reproduced from reference [44].
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 797

Fig. 6. Stimulus-responsive membrane triggering in vitro.


Reproduced from Ref. [88].

investigated using prototype nanocomposite membranes based on modified with pseudorotaxanes [100]. In addition to magnetic-polymer
thermosensitive, poly(N-isopropylacrylamide)-based nanogels and particles, magnetic liposomes [101,102] are also suitable as DEP-type
magnetite NPs. The concept is schematically represented in Fig. 6. The drug delivery systems.
figure shows magnetic triggering and differential flux of Extending the concept of DEP, radio-frequency magnetic-field
sodium fluorescein out of membrane-capped devices as a function of heating of NPs was used to remotely activate temperature-sensitive
time over successive on/off cycles of external magnetic field. Similarly, a cation channels within cells [103]. In this example, superparamagnetic
novel magneto-active gel based on iron oxide NPs within a temperature ferrite NPs were targeted to specific proteins on the plasma membrane
sensitive PNIPAM was used as an externally tunable flow controller of cells expressing TRPV1 (also known as Capsaicin receptor), and
inside a micro fluidic channel [89]. This opened up avenues for the heated by a radiofrequency magnetic field. High localized induction of
development of devices such as magnetic micro/nano pumps, magnetic temperature increase was noted using fluorophores as molecular
field controlled drug delivery devices and magnetic switches. Such thermometers leading to the activation of opening of TRPV1. As
controlled release was also reported from magnetic lipid NPs [90]. shown in Fig. 7, streptavidin-DyLight549 (orange)-coated superpar-
In addition to relatively fluidic polymeric gels, the concept of DEP amagnetic nanoparticles (gray) were prepared and the AP-CFP-TM
was also demonstrated in high density organic and inorganic polymeric protein was bound to the NPs through biotinylated AP domain (green
particles incorporated with MNPs. Examples using organic polymer box), which is anchored to the membrane by the TM (blue box) and CFP
particles include those made from poly-lactic acid (PLA) [91], poly (cyan box) domains. On the application of oscillating magnetic field, the
(ethylene glycol) ethyl ether methacrylate-copoly(ethylene glycol) NPs are locally heated resulting in heat (red)-induced opening of TRPV1.
methyl ether methacrylate [92], Pluronic F127 (F127) [93], poly Novelty of this approach is that it can be generalized to stimulate other
(methylmethacrylate) (PMMA) [94], poly-n-isopropylacrylamide cell types in addition to the possibility to remotely manipulate other
(PNIPAM) [95,96], poly(ethyleneimine)-modified poly(ethylene cellular machinery for novel therapeutics. It is noteworthy that use of
oxide)–poly(propylene oxide)–poly(ethylene oxide) (PEO–PPO–PEO) similar inductive heating effects were also utilized for controlled release
block copolymer [97] and nanoparticle‐assembled capsules (NACs) of drugs by switching the high frequency magnetic fields (HFMF) on and
using poly(allylamine hydrochloride), disodium phosphate, and citrate off from folic acid (FA) and β-cyclodextrin (CD)-functionalized SPIONs
bound MNPs [98]. An example where the behavior of drug release is [104]. Yet another variation of the concept was reported where micro
found to be significantly different depending on whether the applied containers with embedded superparamagnetic nanoparticles were
field varies sinusoidally or in a step-function manner with time was changed into bubble micro reactors upon exposure to AMF, which acts
reported using iron oxide core–chitosan shell microspheres [99]. In yet as a remote trigger for release of encapsulated material [105].
another example, inorganic polymer NPs such as those made from silica Finally, an important question is if there are potential opportuni-
are utilized. In this case, zinc-doped iron oxide nanocrystals were ties for translational research leading to commercialization of
encapsulated within a mesoporous silica framework that was surface- products based on the concept of hyperthermia-based controlled

Fig. 7. A schematic representation of the principles of ion channel stimulation using nanoparticle heating and local temperature sensing.
Reproduced from reference [103].
798 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

Fig. 8. A schematic representation of “active drug-elution technology”.


Reproduced from the web site http://www.biophan.com.

drug delivery. In this context, it is worth mentioning the efforts from 5. Core–shell magnetic nanomaterials for hyperthermia-based
the company, Biophan technologies Inc (http://www.biophan.com/). therapy and controlled drug delivery
The company is developing “active drug-elution technology” where
drug loaded MNPs are encapsulated in a polymer coating on a stent or 5.1. Why core–shell magnetic nanoparticles?
other medical devices from which the drugs can later be selectively
released by applying a controlled EMF at a specific frequency (Fig. 8). As the properties of nanomaterials are surface-dependent, there are
Interestingly, by fine tuning the magnetic properties of MNPs, there is a number of ways one can bring about surface changes leading to
an opportunity for using different MNPs to respond selectively to a concomitant changes in their properties. With the unraveling of their
wide range of frequencies of oscillating magnetic fields in order to chemical, physical and biological properties, nanomaterials of different
release multiple drugs temporally to treat various complications. sizes and shapes are being investigated and recent review articles and

Fig. 9. Different architectures of nanomaterials.


C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 799

book chapters cover various aspects of these nanomaterials (Fig. 9) 5.2.2. Provide oxidative stability
[106–115]. In addition to the size and shape effects, newer design of A shell around magnetic core offers oxidative stability for air
nanomaterials, most often synthesized using layer-by-layer approach, sensitive magnetic nanomaterials. This is especially important for
have also begun gaining prominence providing further opportunities to high magnetic moment particles which can provide high values of SLP
tailor properties of nanomaterials and investigate their fundamental or SPL (specific power loss). For example, body centered cubic (bcc)
behavior. Some of these new designs are core–shell, onion-type, CoFe alloys have the highest Ms (~240 emu/g) in this class of
multilayered, alloy, multimetallic and multifunctional nanomaterials materials and could be an ideal magnetic material for hyperthermia
(Fig. 9). Of these, the layer-by-layer approach for the synthesis of core– applications. However, their chemical instability makes their synthe-
shell architecture offers a great opportunity to modulate properties of sis very challenging [128]. A graphite shell around these particles led
both cores as well as shells. Not surprisingly, this approach has lead to to their increased oxidative stability without loss of their original high
the development of a variety of core–shell nanomaterials and continues magnetic moment [129].
to be the focus of scientific efforts [35].
Core–shell nanomaterials are extremely important as they can have 5.2.3. Enhanced hyperthermia
a combination of different properties and offer multifunctionality Kim et al. achieved close to 90% cancer cell destruction in vitro using
because of core and shell can have different material compositions in FeNi@Au magnetic-vortex microdiscs (MDs) on the application of only a
a single particle. They represent a novel class of hybrid materials, where few tens of hertz AMF for just 10 min [130]. This confirms that operation
composition and microstructure varies in the radial direction [116]. A of MFH at lower frequencies is possible and for effective heat generation
number of possibilities and opportunities can be envisaged using this can be achieved using core–shell type of structures. Likewise, in yet
architecture. A shell can be utilized as a protective shield for sensitive another demonstration, a gold coating of approximately 0.4 to 0.5 nm
core material, for example, in the case of air sensitive magnetic thickness around SPIONs resulted in a four- to five-fold increase in the
nanomaterials like cobalt [117,118]. The dimensions and composition amount of heat released (the highest value of 976 W/g in ethanol at
of shell can be modulated in order to influence the properties of a core 430 Hz frequency) in comparison with SPIONs on application of low
and similarly the core dimensions can be modulated to influence the frequency oscillating magnetic fields (44–430 Hz) (Fig. 10) [131]. In
properties of a shell [119]. The shell surface can also be utilized for bio- addition, the SPIONs@Au were found to be not particularly cytotoxic to
functionalization; which otherwise either difficult or not possible mammalian cells. (MCF-7 breast carcinoma cells and H9c2 cardiomyo-
on a core surface, for example, gold or silica coated ferromagnetic blasts) in in vitro studies. When similar heating experiments were
nanomaterials [117,118,120]. Having a biocompatible shell around toxic carried out using stable water suspensions of La0.75Sr0.25MnO3 cores
core material enables reduction in their toxicity, for example, coated covered by silica (conc. of Mn= 3.39 mg/ml), highest SAR of 130 W/g
quantum dots [121] or cobalt NPs [122]. Expensive nanomaterials can be Mn at 37 °C was reached for the applied amplitude and frequency of
formed as shells around inexpensive core nanomaterials. This is 8.7 kA m− 1, 480 kHz respectively [132].
particularly useful in catalysis [123]. Finally, multiple functions such as There are potential opportunities to extend these investigations
detection and treatment modalities or multiple treatment options can further in order to enhance hyperthermia through manipulation of
be incorporated in a single core–shell particle [124]. All these different magnetic properties utilizing core-shell architecture. For example, a
possibilities are reflected in enhancing the properties of core–shell tunable shell of 0.5 to 3 nm of Fe3O4 on FePt NPs on annealing resulted
nanomaterials in a number of applications and especially in the case of in enhancing the core Ms values to 1040 emu/cc [133]. Novel core–
MNPs and more so for utilization of MFH for therapy and controlled drug shell anisotropic structures such as Ni@Co nanorods with Ni nanowire
release. MagForce Nanotechnologies AG, Germany is the only company as core and Co nanotube as shell were recently synthesized
that is utilizing iron oxide NPs coated with aminosilane for hyperther- demonstrating the potential to fine tune the magnetic anisotropy to
mia treatment of tumors and the technology is at an advanced stage of further optimize hyperthermic potential [134]. Finally, here is an
clinical trials [125]. On the other hand utilization of MNP-based interesting example [135] where the heating rates for dispersion of
hyperthermia for controlled release of drugs is relatively new and to FeCo MNPs with different anisotropy values are shown in Fig. 11. For
the best of our knowledge no clinical trials have been reported. Thus, increasing anisotropy, the particle size at which the maximum heating
there is a need for designing better and more effective hyperthermic rate occurs decreases, while the peak heating rate itself is not
agents and core–shell architecture presents one such opportunity.
Similarly, application of core–shell MFH is relatively new and here is the
first attempt to review the investigations carried out to date.

5.2. Core–shell magnetic nanomaterials for hyperthermia-based therapy

Core–shell MNPs offer a number of advantages over mono metallic/


metal oxide NPs for hyperthermia-based therapy. Some of the examples
from literature demonstrating their potential advantages are discussed
below.

5.2.1. Prevent anisotropic magnetic dipolar attraction


Anisotropic magnetic dipolar attraction prevents stability of
magnetic fluids even in the absence of an external magnetic field.
This continues to be one of the main problems when dealing with
magnetic fluids in general and those particularly for MFH. One
approach to successfully prevent this attraction is by providing a
coating around magnetic particles so that they behave as isotropic
dispersions in a zero magnetic field and reversibly form anisotropic
structures in the presence of an external magnetic field [126]. This
approach was demonstrated recently in silica coated maghemite NPs; Fig. 10. Solvent dependent SPL values for SPIONs@Au.
resulting in a more stable magnetic fluid [127]. Reproduced from the reference [131].
800 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

5.3. Core–shell magnetic nanomaterials for hyperthermia-based


controlled drug delivery

The approach of hyperthermia-based controlled drug delivery using


nanomaterials is relatively new and more so with core–shell magnetic
nanomaterials. However, there are recent examples from the literature
which demonstrate unique opportunities for core–shell MNPs to play a
significant role. The very first example was reported in 2005 where
investigations using a magnetic field to modulate the permeability of
polyelectrolyte microcapsules containing core–shell MNPs were initi-
ated [19]. In these investigations, ferromagnetic gold-coated cobalt
(Co@Au) NPs (3 nm) were embedded inside the polymeric capsule
walls using layer-by-layer self-assembly technique. The final 5 μm
diameter microcapsules had wall structures consisting of 4 bilayers
of poly (sodium styrene sulfonate)/poly(allylamine hydrochloride)
(PSS/PAH), 1 layer of Co@Au, and 5 bilayers of PSS/PAH. Oscillating
Fig. 11. Variation in heating rates for FeCo MNPs with change in anisotropy constant.
magnetic fields of 100–300 Hz at field strength of 1200 Oe were applied
Reproduced from the reference [135]. resulting in distortion of the capsule wall and drastically increased its
permeability to macromolecules like FITC-labeled dextran (Fig. 12). The
capsule permeability change was estimated by taking the capsule
interior and exterior fluorescent intensity ratio using confocal laser
significantly affected. Above a certain value, increase in anisotropy scanning microscopy. Capsules with 1 layer of Co@Au NPs and 10
gives no significant change in the heating rate dependency on particle polyelectrolyte bilayers were found to be optimal for magnetically
size since Néel relaxation ceases to occur. The remaining Brownian controlling the permeability. This example not only demonstrate the
relaxation is independent of material's parameters and only depends importance of layer-by-layer self-assembly approach to fabrication of
on the particle size. Hence, one can expect to achieve high heating rate required core–shell nanomaterials for hyperthermia-based drug deliv-
at desirable sizes for core–shell MNPs. ery, but also strongly supports the unique feature this self-assembly
process provides in controlling core and shell dimensions.
Other examples from the literature are as follows. Iron core silica
5.2.4. Reduce toxicity and increase biocompatibility shell (Fe@SiO2) NPs of about 50 nm were recently synthesized by an in
With an aim to evaluate potential cellular perturbations and explore situ process, and on a short exposure to a high frequency magnetic
the relationship between biocompatibility and surface chemistry of fields (HFMF) they were able to release specific amounts of drug in
carbon coated iron NPs (Fe@CNPs), Mu et al. investigated their dynamic a burst manner [32]. The HFMF accelerated the rotation of MNPs
cellular responses, uptake, oxidative stress and effects on cellular deposited in the silica matrix; subsequently enlarging the nanostruc-
apoptosis, and cell cycle. Results indicate that biocompatibility of ture of the silica matrix to produce porous channels thereby releasing
Fe@CNPs is dependent on both cell type and NP's surface chemistry the drug easily. Learning from such magnetically responsive control-
[136]. Similarly, magnetic manganese oxide NPs when coated with a lable drug release systems, one can also design similar magnetic silica
silica shell resulted in achieving better water stability at high nanospheres for controlled burst release of therapeutic agents
concentrations and superior biocompatibility. The application of AMF especially for urgent physiological needs. Interestingly a novel core–
of 15 mT and 100 kHz for 30 min. produced cellular damage that finally shell structure where the drug is encapsulated within a silica core
lead to apoptotic cell death; even though the temperature increase in surrounded by single-crystalline iron oxide shell was also reported
the cell culture was lower than 0.5 °C [137]. [33]. This design is conceptually interesting as it can protect

Fig. 12. Scheme of the layer-by-layer self-assembly and permeability test for microcapsules embedded with Co@Au NPs under an oscillating magnetic field.
Reproduced from the reference [19].
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 801

biomolecules encapsulated within the core from damage from harsh 6.3. Eliminating or minimizing toxicity from NPs
environments. It can also eliminate uncontrollable release resulting
from natural diffusion of molecules upon delivery, for example, in a Though most of the hyperthermia studies report cell death due to
patient's body. In addition, the magnetic nanoshell displays an ultrafast heat release, there are clearly no sophisticated tools to gather precise
response and sensitivity upon exposure to HFMF. What is important is information about the fate of cells before and after MNPs based
that the high magnetic-sensitivity of the nanometer-scale shell allows a hyperthermia. This is mainly because most of the currently used
controlled burst release of drug in a quantitative manner. Yet another molecular biology assays measure only the changes occurring in short
core–shell structure, magnetic organic–inorganic nanohybrid, involving intervals of time and not much information is available about the NPs
drug-intercalated layered double hydroxides coated on a magnesium after their use. The change due to hysteresis and relaxation losses in
ferrite core, was reported for magnetically controlled drug release [138]. the biological fluids, concentration of material, frequencies applied
With ever increasing ability to fabricate fascinating types of core– and cell death are poorly explained. A better understanding between
shell NPs, the exploration of hyperthermia-based controlled drug NP's physical properties and their influence on cell's integrity needs
delivery systems using core–shell magnetic nanomaterials is poised to further optimization. What is clearly missing in the literature is the
take off. For example, the so-called “yolk–shell” structured materials confirmation that MNP based hyperthermia results in cell death only
with movable cores and porous shells [139] could exhibit unique due to hyperthermia and not due to inherent toxicity of some of the
releasing properties for drug/gene delivery because of their hierar- MNPs [75]. Since a number of magnetic nanomaterials investigated for
chical porous structures. Further, there is also a possibility to expand hyperthermia-based therapy are extremely large in comparison with
the field into the arena of implantable microchips as demonstrated those investigated for hyperthermia-based drug delivery, there is a
recently. In this particular study, drug-carrying magnetic core–shell great opportunity to design better magnetic nanomaterials with
SPION@SiO2 NPs were deposited onto an electrically conductive minimum or no toxicity for drug delivery applications.
flexible PET substrate. A flexible drug delivery chip was made using
such substrates for magnetically controlled release of anti-epileptic 6.4. Biocompatibility, catabolism and clearance issues
drugs such as ethosuximide (ESM) [96].
The unusual properties of a number of MNPs, including their
6. MNP-based hyperthermia for therapy and controlled drug multivalency and multifunctionality, pose challenge for understanding
delivery: current challenges and opportunities their pharmacokinetics because different components will have
different features that affect their distribution, clearance and catabolism.
Taking into consideration more recent experimental results, a A careful examination of each of the components as well as combination
critical examination reveals some of the major challenges and of them is required in order to arrive at reliable pharmacokinetics.
opportunities that MNPs offer for thermotherapy and controlled Such studies are crucial, especially when designing materials that have
drug delivery. These are highlighted below. sophisticated control systems, irrespective of their application either
for heat treatment or drug delivery.
6.1. Maximizing specific absorption rate (SAR)
6.5. Induce and sustain therapeutic temperatures
The most challenging task is to maximize the SAR in hyperthermia,
which allows reduction of ferrofluid dose in vivo. The dependence of In practice, it is technically challenging to induce and sustain
SAR on parameters like magnetization, size, and size-distribution of temperatures clearly above the systemic temperature of 37.5 °C in a
particles, magnetic-field strength, and frequency of the AC field were defined target volume. Perfusion counteracts the temperature rise and
previously discussed [78]. However, what is clearly needed is a perfusion rates vary widely in tumors with leaky vasculatures.
concerted effort to examine all the variables together and come to a Therefore, reaching therapeutic temperatures of 42–44 °C in the
comprehensive and conclusive understanding leading to maximiza- critical parts of tumors requires a specific heating power (SHP) in local
tion of SAR. This is also critical to ensure that maximum intracellular target regions [140]. The cooling action of flowing blood must also be
SAR is obtained at relatively lower dosages. taken into consideration, and furthermore, blood flow rates will vary
during hyperthermia treatment. Taken together, these effects invari-
6.2. Tailoring the properties of magnetic nanomaterials ably result in non-uniform temperature distributions. Adding to this
complexity is the well known fact that in some tumors, the blood flow
Numerous MNP-based alloys of Fe and Co can be readily may altogether stop completely during extended periods of hyper-
synthesized with superior magnetic properties. However, most of thermia treatment [141]. Thermal convection, caused by strong blood
these are not biocompatible and, typically, they are not suitable for in perfusion, decreases the specific energy absorption and hence reduces
vivo applications. One strategy would be to take advantage of their the temperature of the tumor. Therefore, tumors located in regions
superior magnetic properties and enhanced hyperthermia at concen- with high perfusion such as those within liver, lung, and kidney receive
trations low enough to be nontoxic. Alternatively, a core shell strategy lower heat dose, and hence results in a decrease in hyperthermia
to mitigate their toxicity needs to be explored. Yet another option efficiency. In such cases, it is not possible to maintain therapeutic
could be to take a well-known magnetic material, e.g., magnetite, temperatures for long periods of time and hence, they are not treated
which has already been approved for human use and optimize its size, by the methods of regional hyperthermia alone. In these cases, a
composition and shape-dependent magnetic characteristics. After combination of regional hyperthermia with X-ray therapy and
suitably functionalizing its surface and, following appropriate cyto- chemotherapy strengthens the anti-tumor effect and frequently
toxicity and pharmacokinetics studies, it stands a very high chance of results in desired remission. Therefore, regional hyperthermia is
being readily used in vivo. However, even though shape anisotropy is widely used in combination with X-ray therapy and chemotherapy. On
known to play an important role in magnetism, very little work has the other hand it is interesting to note that interface effects are the
been done in utilizing this approach to tailor magnetic nanomaterials reason why cancers of the brain are treated now only with application
for hyperthermia. In the case of hyperthermia-based controlled drug of regional hyperthermia, including cranial trepanation.
delivery, not many types of MNPs have been investigated to date. The Finally, the principal remaining problems with MFH are invasiveness,
field is relatively new and there are a number of opportunities to fine targeting (restricting the hyperthermia effect to a specific area of
tune the properties of MNPs in order to optimize controlled drug interest) and achieving homogenous heat distributions within the target
delivery through both DBB and DEP mechanisms. organ [4,8,54,142]. Failure to solve these problems may lead to either
802 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

insufficient treatment effects or, worse, lethal exposure to neighboring of drug release in vivo as one can correlate the in vitro characteristics of
healthy cells. drug carriers to their in vivo capabilities.

6.8. Theranostics
6.6. Monitoring temperature distribution during heating
Ability to simultaneously carry out diagnosis and therapy, termed
This is essential but remains an ongoing challenge. There is a lack
as theranostics, is now gaining popularity thanks to unique features
of systematic studies on dose and thermal response relationship
provided by nanosystems for both diagnosis and therapy. This is
especially at targeted sites. Currently, in regional hyperthermia
relevant for both hyperthermia-based treatment as well as controlled
systems, the temperature is monitored with the use of invasive
drug delivery. Diagnosis-guided therapy will result in establishing
thermometry where thermo sensors are delivered through catheters
treatment regimens in real time and thereby increasing the therapeu-
implanted surgically or hypodermically. A specific challenge is to
tic index. Even though there are already investigations reported where
develop noninvasive methods for monitoring of local temperatures.
diagnosis is combined with hyperthermia-based therapy or drug
To accomplish this, a two pronged approach that is based on modeling
delivery [44,84], it is clear that there exists a great opportunity for
studies and on designing appropriate thermosensitive materials is
this type of application in future personalized medicine.
essential. In addition to optimizing the physics of heating, further
developments in realistic heating models taking into consideration
6.9. Thermo tolerance
perfusion as well as including systematic studies with phantoms are
required. Estimates of the amount of heat that will be required and
Variation in thermo tolerance of cells is a major concern. For
knowledge of the temperature distribution within the tumor and the
example, this variation in cells ranges from those with increased heat
healthy tissue will be important. There are already some efforts in this
sensitivity to high heat resistance depending on the temperature
direction. Recently, a model was developed based on two finite
programming. Therefore, determining ideal temperature program-
concentric spherical regions; the inner sphere of cancerous tissue with
ming is a challenge. One factor that appears to be consistent in
magnetic particles, and the outer sphere containing healthy tissue
developing such ideal temperature programming is the fact that it is
[143]. Penne's bioheat transfer equation [144] describes the temper-
critical to ensure rapid initial heating. Scientific advances are needed
ature both in the diseased as well as healthy tissue as three different
towards designing heat mediators by improving material magnetiza-
functions of heat; heat generation by MNPs, the heat conducted
tion and size-dispersion. Similarly, development of new strategies to
through the tissue, and the heat removed by blood perfusion. The
differentiate intracellular and extracellular heating based on in vivo
model, applied successfully to self-regulated hyperthermia, showed
studies is needed [149]. While local hyperthermia is crucial for
that proper distribution of magnetic particles throughout the tumor is
treatment, it is the response of normal tissues that determines what
the key to minimize any damage to the surrounding healthy tissue
“dose” of heat can be applied. Based on the available data it appears
while still maintaining a therapeutic temperature within the tumor
that the dose–response curves for hyperthermia look similar to those
[145]. The model can also be used to find ways in applying the
for radiation or drug dose. However, the challenge is to understand
magnetic field to minimize damage to healthy tissues and the patient's
critical cellular targets of thermal inactivation both in cancer cells as
exposure to the field. Future computer simulations could assist doctors
well as in the surrounding healthy cells [150]. Meeting this challenge is
in determining how, when and where to place the magnetic fluids.
crucial for mapping thermo tolerance in the critical regions of a tissue.
Precise treatments would increase the likelihood of eliminating the
tumor while decreasing the side effects of heating healthy tissue.
6.10. Tolerance of magnetic field and frequency of oscillation
In terms of designing thermosensitive materials for monitoring
heat distribution, there are some recent efforts worth mentioning.
It is known that electrical conductivity of biological tissue is
For example, Herrera et al. [146] coated MNPs with a temperature-
sufficiently higher than that of AMF. This may, therefore, generate
responsive fluorescent polymer built from N-isopropylacrylamide
eddy currents and cause non-selective heating of both cancer as well
(NIPAM) and a modified acrylamide. By observing the changes in the
as normal tissue. The heat generated by such induced eddy currents
fluorescence of the polymer using a spectrofluorometer, temperature
needs to be considered in determining the conditions for optimizing
of the medium surrounding the NPs was monitored non-invasively.
the specific heating power for a given NP-application. The applied
Another approach is to use infrared thermacam, which is insensitive to
frequency is also important, as researchers have generally focused on
AC magnetic fields but sensitive to measure thermal gradients during
pulsed frequencies in the range of 50 kHz to 10 MHz to achieve
magnetic heating [147].
heating [132]. There is also a limit on the amplitude of applied field;
In the case of hyperthermia-based drug delivery it is important to
which is about 8–16 kA m− 1.
know, both from computational modeling studies as well as
Since it is well established that the required reversible profiles are
experimental determinations, the relationship between local heating
obtained only under certain magnetic field strengths and frequencies
and its influence on bond breaking in the case of DBB and permeability
[19], determining these values for each type of system is required for
through polymer in the case of DEP. These studies, hitherto never
hyperthermia-based drug delivery systems. Especially investigation of
undertaken, are critical to design controlled drug delivery systems with
kinetics of structural changes is crucial.
required release profiles.
6.11. Self-regulation of heating
6.7. Visualization
Achieving self-controlled and self regulated heating is important
In addition to monitoring the temperature distribution, ability to for minimizing some of the deleterious effects of hyperthermia. One
visualize accumulation of drug carrying NPs at the target site, the release way to achieve this self-control is by focusing on two important
of drug and its distribution is critical in designing hyperthermia-based magnetic properties of the NPs — magnetization and coercivity. These
drug delivery systems. While some progress has been made in this two can control the heating efficiency and Curie temperature so that
direction [148], there is clearly a need for focusing future efforts to they can be adjusted slightly above the therapeutic temperature
enable noninvasive assessment of accumulation of particles, drug (approx. 45 °C) in order to achieve a self-controlled heating mecha-
release and distribution. This requirement gains added importance nism. The route towards self-regulating heating mediators is open and
with possibility for extending imaging capabilities to investigate kinetics requires further improvement to permit sufficient heating. Low-Curie
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 803

temperature NPs such as iron–platinum (FePt) and nickel–palladium regression followed by rejection of melanoma cells by cured mice;
(NiPd) NPs with ideal sizes and magnetic properties would heat confirming the generation of antitumor immunity [67]. These studies
efficiently and maintain therapeutic temperatures. Another possible are supported by the observations of Ito et al. that there is
approach is to use complex magnetic oxides as core materials (for development of antitumor immunity after hyperthermia treatment
example La1 − xSrxMnO3 (LSMO) perovskites) [151], whose magnetic with MCLs in rat models as well [68]. Similar antitumor immunity was
properties can be properly tailored in various ways, in order to control also observed in addition to tumor size reduction in cat mammary
heating efficiencies with self regulation. carcinoma [70]. Therefore, there appears to be an opportunity not only
for tumor reduction due to hyperthermia but also protection from
6.12. Practical targeted hyperthermia future tumor development through the development of antitumor
immunity. In principle, this approach can be utilized for the
There is a need for development of new targeting strategies which development of in situ vaccinations leading to advancement of novel
can effectively drive the intravenously injected particles to targeted sites heat-immuno based therapies [65,68].
and complete excretion of the injected particles after the treatment. This
is relevant for both hyperthermia-treatment and drug delivery. It is 6.14. Combination of treatments
important that MNP formulations have the ability to overcome one of
the main biological barriers such as exclusion by BBB, the vascular Literature reports show that hyperthermia cancer treatment is
endothelium, the typical higher osmotic pressure, cancer lesions causing much more effective when applied in combination with other
the outward flow of any therapeutic agents, phagocytosis and clearance treatment methods like radiation, chemotherapies etc. than applying
from circulation by the reticulo-endothelial system (RES) that prevent alone. When synergistically applied with radiation, the heat released
them from reaching their targets [57,152]. It is interesting to note that first will bring changes in cell cycle causing faster denaturation of
macrophages significantly took up oligomannose-coated liposomes malignant cells through aggregation of nuclear proteins; thereby
(OMLs) when injected into the peritoneal cavity, and then gradually enhancing the sensitivity of already denatured cells to death by
accumulated in the omentum and other lymphoid tissues within 24 h radiation [63,155]. The thermal enhancement ratio (TER) is defined as
[153]. When MNPs were encased in the OMLs to achieve in vivo the ratio of radiation sensitivity at 37.5 °C to the sensitivity at an
hyperthermia at the site in a mouse i.p. metastasis model, it successfully elevated temperature [64]. Highest TER is obtained with a combination
controlled tumor development. What is truly encouraging is the treatment, since the inner portions of tumor bed are in an oxygen
accumulation of high concentrations of magnetite NPs, even up to deprived state (hypoxia) and radiation cannot easily pass through it
160 mg, in the momentum. Similarly, based on our recent results [154], during radiation therapy. A combination therapy allows denaturation
where targeting through luteinizing hormone and releasing hormone of tumor cells from inside by heat induced MNPs and outside by
(LHRH) resulted in accumulation of large concentration of magnetite radiation since the tumor beds from inside are radiation resistant and
NPs (upto 72 pg/cell) in lung metastases bode well for successful thermosensitive [67]. Further, adjuvant therapy combining MFH with
practical targeted hyperthermia and controlled drug delivery. With chemo-radiation strategies also appear to hold great promise in
potential avenues to tailor make MNPs to obtain high SAR; it is possible oncology. Even though synergy between heat and radiation dose has
to have successful targeted hyperthermia-based treatment and drug been validated by a number of preclinical studies [156,157], the time
delivery at relatively lower dosages. difference between the treatments and their sequence of application is
critical [158]. For example, best results are obtained with combined
6.13. Antitumor immunity after hyperthermia treatments through simultaneous application rather by applying heat
therapy alone. However, this may be difficult to realize in clinical
A number of literature reports suggest potential of developing practice. Overall, it can be safely concluded that higher levels of
antitumor immunity after hyperthermia based treatment. For exam- survival of experimental animals were achieved when hyperthermia
ple, Siva Sai et al. showed that ligand protected Fe/Fe3O4 inorganic was performed in combination with chemotherapy and sensitization
core–shell MNPs caused the development of anti-tumor effect on of tumor tissues [159].
murine B16-F10 melanoma in mice. Only after a three short 10-minute
AMF activation (366 kHz), an appreciable decrease in the amount of 6.15. Multi-therapeutic and multi-functional modalities
tumor size was observed [69]. Similar mice studies by Suzuki et al.,
after magnetite cationic liposomes (MCLs) injection into melanoma Potential opportunities exist for multi-therapeutic modalities-
nodules followed by AMF subjection, showed complete tumor combined hyperthermia treatment and drug delivery. For example,

Fig. 13. A schematic representation of three different types of nanoconstructs for hyperthermia-based drug delivery type DEP.
804 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

different types of nanoconstructs can be envisaged. Three examples 6.16. Drug loading and stability
are: 1) magnetic core polymer shell nanoparticles with encapsulated
drug within a polymeric shell and a targeting agent on its surface It is well established that traditional approach for controlled drug
(Fig. 13A), 2) MNP, drug and polymer nanocomposite with a targeting delivery using polymeric NPs for encapsulation of high quantities of
agent on its surface (Fig. 13B), and 3) a core containing drug with a drugs has several problems. If the particles are small, they tend to
polymer shell encapsulated with MNPs and a targeting agent on its reduce their overall stability leading to an undesired burst release effect
surface (Fig. 13C). However, there is a need to experimentally validate and reduced efficacy [160]. On the other hand, larger particles tend to
the potential of these type of constructs for hyperthermia-based have slower in vitro release profiles. However, when systemically
therapy and drug release. It is critical to optimize the influence of delivered, they may be more readily detected and cleared from
nature of polymer and MNPs on controlled drug release, as it was circulation, resulting in lack of efficacy [161]. While magnetically
recently shown that [94] the release of FITC from magnetite–PMMA controlled drug release using traditional polymeric NPs may have
particles can be induced thermally but not magnetically. On the other similar problems, a core shell structure containing drug within a core
hand there was no release of FITC from cobalt–PMMA composites and a shell composed of MNPs can overcome such as problem.
either through thermal or magnetic induction. This type of nanocon- In addition to drug loading, stability of the drug within the
structs can also meet some of the current challenges in spatiotemporal nanoparticle needs to be evaluated. The drug-nanoparticle interaction
control delivery of therapeutic agents. In addition, further fine tuning studies with focus on drug stability are critically important for both
of drug delivery is possible with potential for remotely triggered drug types of hyperthermia-based controlled drug delivery. Preliminary
release profile-controllable and switchable-between sustainable investigations reported to date have only been focused on drug
and pulsatile pattern — to precisely control under a variety of clinical molecules that are not very sensitive to the environment [44].
settings with potential opportunities to combine magnetic nanopar- However, what is required is detailed drug-excipient compatibility
ticle MRI based diagnosis with hyperthermia, multifunctional para- studies, particularly for drug molecules which are thermosensitive
digms for personalized medicine can be envisaged [154]. and prone to undergo modifications due to redox reactions.

Table 1
List of magnetic nanoparticles reported for application in hyperthermia-based therapy and controlled drug delivery.

S. no Type of magnetic nanoparticle Application Reference

1. Fe doped Au NPs Hyperthermia-based therapy [50]


2. γ-Fe2O3 Hyperthermia-based therapy [40,47]
Cobalt ferrite
3. Fe3O4 Hyperthermia-based therapy [47]
Fe3O4-poly vinyl alcohol
4. NiFe2O4 Hyperthermia-based therapy [48]
5. γ-Fe2O3 Hyperthermia-based therapy [22,47]
7. Fe3O4@chitosan Hyperthermia-based therapy [42]
8. Fe3O4@block copolymers Hyperthermia-based therapy [62]
9. Fe3O4-dextran stabilized Hyperthermia-based therapy [62]
Fe3O4@Aminosilan
10. Ferrite-Dextran stabilized Hyperthermia-based therapy [62–64]
11. Fe3O4-dextran (mono and bilayer) stabilized Hyperthermia-based therapy [45]
12. Fe3O4-lauric acid stabilized Hyperthermia-based therapy [70]
13. Fe3O4-lauric acid stabilized Hyperthermia-based therapy [25]
MnFe2O4-lauric acid stabilized
CoFe2O4-lauric acid stabilized
14. Fe@biscarboxyl-terminated poly(ethylene glycol) (cPEG) Hyperthermia-based therapy [82]
15 γ-MnxFe2 − xO3 coated Acrypol 934 polymer Hyperthermia-based therapy [142]
16. FeCo@Au Hyperthermia-based therapy [47,56,61]
17. Fe@MgO Hyperthermia-based therapy [165]
18. Fe3O4@Si Hyperthermia-based therapy [168]
19. Fe2O3@SiO2 Hyperthermia-based therapy [84]
20. FeNi@Au microdiscs Hyperthermia-based therapy [130]
21. Fe@Fe3O4 Hyperthermia-based therapy [69]
22. La0.56(CaSr)0.22MnO3@SiO2 Hyperthermia-based therapy [137]
23. Fe3O4@Au Hyperthermia-based therapy [131]
24. Magnetite cationic liposomes (MCL) Hyperthermia-based therapy [11,66–68,71,72,78]
25. Fe3O4-lauric acid stabilized Hyperthermia-based therapy [59,70]
26. Fe2O3@SiO2 bound LHRH Hyperthermia-based therapy [84]
27. SPIONs bound fluorophore bimane Hyperthermia-based Controlled drug delivery [85]
28. Porous Fe3O4/Fe/SiO2 Hyperthermia-based Controlled drug delivery [32,33]
29. Fe@SiO2 Hyperthermia-based Controlled drug delivery [93]
30. poly-(N-vinyl-2-pyrrolidone) (PVP)-modified silica core@iron oxide shell Hyperthermia-based Controlled drug delivery [33]
31. Mg–Al layered double hydroxides (LDH) coated magnesium ferrite NPs Hyperthermia-based Controlled drug delivery [138]
32. Yolk–shell type nanospheres with movable cores of Au, SiO2, Fe3O4. Hyperthermia-based Controlled drug delivery [139]
33. γ-MnxFe2 − xO3 coated Acrypol 934 polymer Hyperthermia-based therapy and controlled drug delivery [142]
34. Fe3O4@lipid membrane (MCL, magnetite cationic liposome) Hyperthermia-based therapy and controlled drug delivery [71]
35. Fe@carbon nanoparticles bound polymer Hyperthermia-based therapy and controlled drug delivery [136]
36. Co@Au@ poly(sodium styrene sulfonate)/poly(allylamine hydrochloride) Hyperthermia-based therapy and controlled drug delivery [19]
37. SPIONs@ sensitive poly (N-isopropylacrylamide) hydrogels Hyperthermia-based therapy and controlled drug delivery [86]
38. Fe@Fe3O4 loaded 4-tetracarboxy phenyl porphyrin Hyperthermia-based therapy and controlled drug delivery [69]
39. Carboplatin-Fe@C-loaded chitosan Hyperthermia-based therapy and controlled drug delivery [166]
40. Zinc doped iron oxide nanocrystals encapsulated mesoporous silica Hyperthermia-based therapy and controlled drug delivery [100]
41. MCL loaded 4-S-Cysteaminylphenol Hyperthermia-based therapy and controlled drug delivery [167]
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 805

6.17. Influence of nature and size of the shell References

This is relevant in the case of hyperthermia-based drug delivery [1] (a) S. Nagarajan, Z. Yong, Use of core/shell structured nanoparticles for
where drug molecules and MNPs are encapsulated. To date, there are no biomedical applications, Recent Patents Biomed. Eng. 1 (2008) 34–42;
(b) http://www.medicinenet.com/hyperthermia/article.html.
experimental studies that provide a clear understanding of the influence [2] G.V. Loo, X. Saelens, M.V. Gurp, M. MacFarlane, S.J. Martin, P. Vandenabeele, The
of nature of polymeric shell and its thickness in providing controlled role of mitochondrial factors in apoptosis: a Russian roulette with more than one
release profiles. While few standard computational models are available bullet, Cell Death Differ. 9 (2002) 1031–1042.
[3] L.S. Goldstein, M.W. Dewhirst, M. Repacholi, L. Kheifets, Summary, conclusions
for drug release from polymeric materials [169], what is truly needed is a and recommendations: adverse temperature levels in the human body, Int. J.
model system that takes into account multi component systems and Hyperther. 19 (2003) 373–384.
stimuli responsive polymers. [4] B. Hildebrandt, P. Wust, O. Ahlers, A. Dieing, G. Sreenivasa, T. Kerner, R. Felix, H.
Riess, The cellular and molecular basis of hyperthermia, Crit. Rev. Oncol. Hematol.
43 (2002) 33–56.
[5] (a) R. Suto, P.K. Srivastava, A mechanism for specific immunogenicity of heat
7. Conclusions shock protein — chaperoned peptides, Science 269 (1995) 1585–1588;
(b) P. Wust, B. Hildebrandt, G. Sreenivasa, B. Rau, J. Gellermann, H. Riess, R.
Felix, P.M. Schlag, Hyperthermia in combined treatment of cancer, Lancet
At the very outset, it should be pointed out that MNPs are classified Oncol. 3 (2002) 487–497.
as medical devices for regulatory purposes and as per the US-FDA [6] M.J. Santos-Marques, F. Carvalho, C. Sousa, F. Remiao, R. Vitorino, F. Amado, R.
should conform to ISO 10993 guidelines. This classification is very Ferreira, J.A. Duarte, M.L. Bastos, Cytotoxicity and cell signalling induced by
continuous mild hyperthermia in freshly isolated mouse hepatocytes, Toxicol-
important as it will enable faster translation of innovative research ogy 224 (2006) 210–218.
using nanomaterials for hyperthermia-based treatment and drug [7] G.P. Raaphorst, M.L. Freeman, W.C. Dewey, Radiosensitivity and recovery from
release. For the sake of clarity, we have summarized (Table 1) a list of radiation damage in cultured CHO cells exposed to hyperthermia at 42.5 or
45.0 °C, Radiat. Res. 79 (1979) 390–402.
different types of magnetic nanomaterials that are currently in
[8] R.W.Y. Habash, R. Bansal, D. Krewski, H. Alhafid, Thermal therapy, part 2:
development for these two applications. The table provides an hyperthermia techniques, Crit. Rev. Biomed. Eng. 34 (2006) 491–542.
overview of the possibilities for these different types of magnetic [9] (a) http://www.cancer.gov/cancertopics/factsheet/Therapy/hyperthermia;
nanomaterials. In our view and as described in this review, (b) M.H. Falk, R.D. Issels, Hyperthermia in oncology, Int. J. Hyperther. 17 (2001)
1–18.
classification of heat treatment and drug delivery under the category
[10] A. Jordan, EuroNanoForum Proceedings, 2005, pp. 76–80.
of hyperthermia is extremely important in order to unleash the [11] K. Tanaka, T. Ito, T. Kobayashi, T. Kawamura, S. Shimada, K. Matsumoto, T. Saida,
advantages of the combination. The possibilities to create core–shell H. Honda, Heat immunotherapy using magnetic nanoparticles and dendritic cells
magnetic nanomaterials and nanoshells, especially the opportunities for T-lymphoma, J. Biosci. Bioeng. 100 (2005) 112–115.
[12] R.K. Gilchrist, R. Medal, W.D. Shorey, R.C. Hanselman, J.C. Parrott, C.B. Taylor,
to utilize layer-by-layer self-assembly approaches [19,35], will lead to Selective inductive heating of lymph nodes, Ann. Surg. 146 (1957) 596–606.
innovative designs for different types of hyperthermia-based treat- [13] R.W. Rand, H.D. Snow, D.G. Elliott, G.M. Haskins, Induction heating method
ments. Such combination offers multi-therapeutic modalities in a for use in causing necrosis of neoplasm, US Patent Specification (1985), 4,
545, 368.
single treatment. As the field progresses, new paradigms such as
[14] A. Ito, H. Honda, T. Kobayashi, Cancer immnotherapy based on intracellular
magnetomechanical stimuli induced magnetic disks for treatment can hyperthermia using magnetite nanoparticles: a novel concept of “heat-controlled
also be added under this umbrella. In this approach magnetic disks are necrosis” with heat shock protein expression, Cancer Immunol. Immunother.
targeted to tumors and cancer cell destruction takes place through 55 (2006) 320–328.
[15] (a) M. Faraji, Y. Yamini, M. Rezaee, Magnetic nanoparticles: synthesis,
disruption of the cell membrane initiated by spinning them on cue stabilization, functionalization, characterization, and applications, J. Iran.
[162]. Unlike in traditional hyperthermia, biologically relevant effect Chem. Soc. 7 (2010) 1–37;
can be achieved through application of weak magnetic fields (b) G.M. Whitesides, The ‘right’ size in nanobiotechnology, Nat. Biotechnol. 21
(2003) 1161–1165.
(b100 Oe) with frequency of a few tens of Hz, applied for a duration [16] I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and
of only 10 min. Ultimately, we envision a future in which nanostruc- diagnosis, Adv. Drug Deliv. Rev. 54 (2002) 631–651.
tured materials are created with multifunctionalities with optimized [17] D. Bahadur, J. Giri, Biomaterials and magnetism, Sadhana 28 (2003) 639–656.
[18] A. Ito, M. Shinkai, H. Honda, T. Kobayashi, Medical application of functionalized
pharmacokinetics and pharmacodynamics offering most effective and magnetic nanoparticles, J. Biosci. Bioeng. 100 (2005) 1–11.
truly intelligent therapies [163]. In addition to intelligent therapies, we [19] L. Zonghuan, P.D. Malcolm, G. Zhanhu, G.O. Vladimir, C.S.S.R. Kumar, L.M. Yuri,
see a great opportunity for combining in real time diagnostic Magnetic switch of permeability for polyelectrolyte microcapsules embedded
with nanoparticles, Langmuir 21 (2005) 2042–2050.
modalities. Such a powerful combination is especially attractive for
[20] B. Jeyadevan, Present status and prospects of magnetite nanoparticles-based
hyperthermia-based treatment and drug delivery using MNPs wherein hyperthermia, J. Ceram. Soc. Jpn. 118 (2010) 391–401.
appropriate imaging tools such as MRI and fluorescence imaging can [21] B. Thiesen, A. Jordan, Clinical applications of magnetic nanoparticles for
be added to create a truly intelligent “theranostics”. Future in- hyperthermia, Int. J. Hyperther. 24 (2008) 467–474.
[22] R. Hergt, S. Dutz, R. Müller, M. Zeisberger, Magnetic particle hyperthermia:
vestigations will lead to creation of “intelligent theranostics” for not nanoparticle magnetism and materials development for cancer therapy, J. Phys.
only noninvasive assessment of pharmacokinetics and pharmacody- Condens. Matter 18 (2006) S2919–S2934.
namics of drug but also for real-time monitoring of therapeutic [23] F. Gazeau, M. Lévy, C. Wilhelm, Optimizing magnetic nanoparticle design for
nanothermotherapy, Nanomedicine-UK 3 (2008) 831–844.
responses. It is our belief that biomedical nanotechnology in general [24] S. Müller, Magnetic fluid hyperthermia therapy for malignant brain tumors—an
and hyperthermia-based treatment and drug delivery approaches ethical discussion, Nanomedicine 5 (2009) 387–393.
based on magnetic nanomaterials in particular will move pharmaceu- [25] (a) P. Pradhan, J. Giri, G. Samanta, H.D. Sarma, K.P. Mishra, J. Bellare, R.
Banerjee, D. Bahadur, Comparative evaluation of heating ability and
tical industry from ‘blockbuster drug’ model to ‘personalized medicine’ biocompatibility of different ferrite-based magnetic fluids for hyper-
[164]. thermia application, J. Biomed. Mater. Res. B Appl. Biomater. 81B (2007)
12–22;
(b) D.H. Kim, Y.T. Thai, D.E. Nikles, C.S. Brazel, Heating of aqueous dispersions
containing MnFe2O4 nanoparticles by radio-frequency magnetic field
Acknowledgment induction, IEEE Trans. Magn. 45 (2009) 64–70;
(c) O. Kaman, E. Pollert, P. Veverka, M. Veverka, E. Hadova, K. Knýzek, M.
Marysko, P. Kaspar, M. Klementova, V. Grunwaldova, S. Vasseur, R. Epherre,
We thank Louisiana Board of Regents for an equipment grant to
S. Mornet, G. Goglio, E. Duguet, Silica encapsulated manganese perovskite
purchase SQUID magnetometer (LEQSF (2008-10)-ENH-TR-07); which nanoparticles for magnetically induced hyperthermia without the risk of
was used to generate magnetic data on some of our MNPs. Funding overheating, Nanotechnology 20 (2009) 275610;
from NIH (1RO1CA142-01A1) is gratefully acknowledged. FM acknowl- (d) V.A. Atsarkin, L.V. Levkin, V.S. Posvyanskiy, O.V. Melnikov, M.N. Markelova,
O.Y. Gorbenko, A.R. Kaul, Solution to the bioheat equation for hyperthermia
edges NIH-supported INBRE program of NCRR (P20RR016456) for the with La1 − xAgyMnO3 − δ nanoparticles: the effect of temperature autostabi-
summer-2010 funding. lization, Int. J. Hyperthermia 25 (2009) 240–247;
806 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

(e) G. Kong, R.D. Braun, M.W. Dewhirst, Characterization of the effect of [50] (a) A. Wijaya, K.A. Brown, J.D. Alper, K.H. Schifferli, Magnetic field heating study
hyperthermia on nanoparticle extravasation from tumour vasculature, Cancer of Fe-doped Au nanoparticles, J. Magn. Magn. Mater. 309 (2007) 15–19;
Res. 61 (2001) 3027–3032; (b) R. Sharma, C.J. Chen, Newer nanoparticles in hyperthermia treatment and
(f) M.S. Martina, C. Wilhelm, S. Lesieur, The effect of magnetic targeting on the thermometry, J. Nanopart. Res. 11 (2009) 671–689.
uptake of magnetic-fluid-loaded liposomes by human prostatic adenocarci- [51] H. Nojima, S. Ge, Y. Katayama, S. Ueno, K. Iramina, Effect of the stimulus
noma cells, Biomaterials 29 (2008) 4137–4145. frequency and pulse number of repetitive transcranial magnetic stimulation on
[26] Nanotechnologies for the Life Sciences, Vol. 6–10, Edited by Challa Kumar, the inter-reversal time of perceptual reversal on the right superior parietal
Published by Wiley-VCH, 2006. lobule, J. Appl. Phys. 107 (2010) 09B320-09B323.
[27] J.M. Chan, L. Zhang, R. Tong, D. Ghosh, W. Gao, G. Liao, K.P. Yuet, D. Gray, J.-W. Rhee, J. [52] (a) D.L. Huber, Synthesis, properties, and applications of iron nanoparticles,
Cheng, G. Golomb, P. Libby, R. Langer, O.C. Farokhzad, Spatiotemporal controlled Small 1 (2005) 482–501;
delivery of nanoparticles to injured vasculature, PNAS 107 (2010) 2213–2218. (b) P. Gould, Nanoparticles probe biosystems, Mater. Today 7 (2004) 36–43.
[28] (a) Y. Zhao, J.L. Vivero-Escoto, I.I. Slowing, B.G. Trewyn, V.S.-Y. Lin, Capped [53] (a) P. Pradhan, J. Giri, R. Banerjee, J. Bellare, D. Bahadur, Cellular interactions of
mesoporous silica nanoparticles as stimuli-responsive controlled release lauric acid and dextran-coated magnetite nanoparticles, J. Magn. Magn.
systems for intracellular drug/gene delivery, Expert Opin. Drug Deliv. 7 Mater. 311 (2007) 282–287;
(2010) 1013–1029; (b) J.L. Corchero, A. Villaverde, Biomedical applications of distally controlled
(b) B.P. Timko, T. Dvir, D.S. Kohane, Remotely triggerable drug delivery systems, magnetic nanoparticles, Trends Biotechnol. 27 (2009) 468–476.
Adv. Mater. (2010), doi:10.1002/adma.201002072. [54] S. Deger, D. Boehmer, I. Türk, J. Roigas, V. Budach, S.A. Loening, Interstitial
[29] J. Kost, J. Wolfrum, R. Langer, Magnetically enhanced insulin release in diabetic hyperthermia using self-regulating thermoseeds combined with conformal
rats, J. Biomed. Mater. Res. 21 (1987) 1367–1373. radiation therapy, Eur. Urol. 42 (2002) 147–153.
[30] E.R. Edelman, J. Kost, H. Bobeck, R. Langer, Regulation of drug release from polymer [55] C.L. Ondeck, A.H. Habib, P. Ohodnicki, K. Miller, C.A. Sawyer, P. Chaudhary, M.E.
matrices by oscillating magnetic fields, J. Biomed. Mater. Res. 19 (1985) 67–83. McHenry, Theory of magnetic fluid heating with an alternating magnetic field
[31] J. Kost, R. Noecker, E. Kunica, R. Langer, Magnetically controlled release systems: with temperature dependent materials properties for self-regulated heating,
effect of polymer composition, J. Biomed. Mater. Res. 19 (1985) 935–940. J. Appl. Phys. 105 (2009) 07B324 (3pages).
[32] S.H. Hu, T.Y. Liu, H.Y. Huang, D.M. Liu, S.Y. Chen, Magnetic-sensitive silica [56] M.G. Weimuller, M. Zeisberger, K.M. Krishnan, Size-dependant heating rates of
nanospheres for controlled drug release, Langmuir 24 (2008) 239–244. iron oxide nanoparticles for magnetic fluid hyperthermia, J. Magn. Magn. Mater.
[33] S.H. Hu, S.Y. Chen, D.M. Liu, C.S. Hsiao, Core/single-crystal-shell nanospheres for 321 (2009) 1947–1950.
controlled drug release via a magnetically triggered rupturing mechanism, Adv. [57] K.M. Krishnan, Biomedical nanomagnetics: a spin through possibilities in
Mater. 20 (2008) 2690–2695. imaging, diagnostics, and therapy, IEEE Trans. Magn. 46 (2010) 2523–2558.
[34] S.C. Brazel, Magnetothermally-responsive nanomaterials: combining magnetic [58] (a) Y.W. Jun, Y.M. Huh, J.S. Choi, J.H. Lee, H.T. Song, S. Kim, S. Yoon, K.S. Kim, J.S.
nanostructures and thermally-sensitive polymers for triggered drug release, Shin, J.S. Suh, J. Cheon, Nanoscale size effect of magnetic nanocrystals and
Pharm. Res. 26 (2009) 644–656. their utilization for cancer diagnosis via magnetic resonance imaging, J. Am.
[35] (a) W. Schärtl, Current directions in core–shell nanoparticle design, Nanoscale 2 Chem. Soc. 127 (2005) 5732–5733;
(2010) 829–843; (b) S. Purushotham, R.V. Ramanujan, Modeling the performance of magnetic
(b) M.M. De Villiers, Y. Lvov, Nanoshells for drug delivery, in: Challa S.S.R. nanoparticles in multimodal cancer therapy, J. Appl. Phys. 107 (2010) 114701.
Kumar (Ed.), (Nanomaterials for Medical Diagnosis and Therapy) of the [59] R. Hergt, R. Hiergeist, M. Zeisberger, D. Schüler, U. Heyen, I. Hilger, W.A. Kaiser,
Book Series on Nanotechnologies for Life Sciences, vol. 10, Wiley VCH, 2007. Magnetic properties of bacterial magnetosomes as potential diagnostic and
[36] B.A. Bornstein, P.S. Zouranjian, J.L. Hansen, S.M. Fraser, L.A. Gelwan, B.A. Teicher, therapeutic tools, J. Magn. Magn. Mater. 293 (2005) 80–86.
G.K. Svensson, Local hyperthermia, radiation therapy, and chemotherapy in [60] Kim D-H, T.Y. Thai, D.E. Nikles, C.S. Brazel, Heating of aqueous dispersions
patients with local–regional recurrence of breast carcinoma, J. Radiat. Oncol. Biol. containing MnFe2O4 nanoparticles by radio-frequency magnetic field induction,
Phys. 25 (1993) 79–85. IEEE Trans. Magn. 45 (2009) 64–70.
[37] A. Jordan, P. Wust, H. Fahling, W. John, A. Hinz, R. Felix, Inductive heating of [61] G.W. Marcela, Z. Matthias, M.K. Kannan, Size-dependant heating rates of iron
ferrimagnetic particles and magnetic fluids: physical evaluation of their oxide nanoparticles for magnetic fluid hyperthermia, J. Magn. Magn. Mater. 321
potential for hyperthermia, Int. J. Hyperther. 9 (1993) 51–68. (2009) 1947–1950.
[38] R.E. Rosensweig, Heating magnetic fluid with alternating magnetic field, J. Magn. [62] (a) A. Jordan, R. Scholz, P. Wust, H. Schirra, T. Schiestel, H. Schmidt, R. Felix,
Magn. Mater. 252 (2002) 370–374. Endocytosis of dextran and silan-coated magnetite nanoparticles and the
[39] R. Hergt, W. Andrä, C.G. d'Ambly, I. Hilger, W.A. Kaiser, U. Richter, H.G. Schmidt, effect of intracellular hyperthermia on human mammary carcinoma cells in
Physical limits of hyperthermia using fine magnetite particles, IEEE Trans. Magn. vitro, J. Magn. Magn. Mater. 185 (1999) 185–196;
34 (1998) 3745–3754. (b) A. Aqil, S. Vasseur, E. Duguet, C. Passirani, J.P. Benoît, R. Jérôme, C. Jérôme,
[40] (a) J.P. Fortin, C. Wilhelm, J. Servais, C. Menager, J.C. Bacri, F. Gazeau, Size-sorted Magnetic nanoparticles coated by temperature responsive copolymers for
anionic iron oxide nanomagnets as colloidal mediators for magnetic hyperthermia, J. Mater. Chem. 18 (2008) 3352–3360;
hyperthermia, J. Am. Chem. Soc. 129 (2007) 2628–2635; (c) C.S.S.R. Kumar, F. Mohammad, Magnetic Gold Nanoshells: Step-wise
(b) J.P. Fortin, F. Gazeau, C. Wilhelm, Internal heating of living cells with Changing of Magnetism through Step-wise Biofunctionalization, J. Phys.
magnetic nanomediators, Eur. Biophys. J. 37 (2008) 223–228. Chem. Lett. 1 (200) (2010) 3141–3146.
[41] (a) Q.A. Panhurst, J. Connolly, S.K. Jones, J. Dobson, Applications of magnetic [63] A. Jordan, R. Scholz, P. Wust, H. Fahling, R. Felix, Magnetic fluid hyperthermia
nanoparticles in biomedicine, J. Phys. D Appl. Phys. 36 (2003) R167–R181; (MFH): cancer treatment with AC magnetic field induced excitation of
(b) A.K. Gupta, M. Gupta, Synthesis and surface engineering of iron oxide biocompatible superparamagnetic nanoparticles, J. Magn. Magn. Mater. 201
nanoparticles for biomedical applications, Biomaterials 26 (2005) 3995–4021. (1999) 413–419.
[42] (a) D.L. Zhao, X.X. Wang, X.W. Zeng, Q.S. Xia, J.T. Tang, Preparation and inductive [64] N.A. Brusentsov, L.V. Nikitin, T.N. Brusentsova, A.A. Kuznetsov, F.S. Bayburtskiy, L.I.
heating property of Fe3O4–chitosan composite nanoparticles in an AC magnetic Shumakov, N.Y. Jurchenko, Magnetic fluid hyperthermia of the mouse experimen-
field for localized hyperthermia, J. Alloys Compd. 477 (2009) 739–743; tal tumor, J. Magn. Magn. Mater. 252 (2002) 378–380.
(b) C. Riviere, C. Wilhelm, F. Cousin, V. Dupuis, F. Gazeau, R. Perzynski, Internal [65] K. Tanaka, A. Ito, K. Kobayashi, T. Kawamura, S. Shimada, K. Matsumoto, T. Saida,
structure of magnetic endosomes, Eur. Phys. J. E Soft Matter 22 (2007) 1–10. H. Honda, Intratumoral injection of immature dendritic cells enhances antitumor
[43] R. Hergt, S. Dutz, Magnetic particle hyperthermia—biophysical limitations of a effect of hyperthermia using magnetic nanoparticles, Int. J. Cancer 116 (2005)
visionary tumour therapy, J. Magn. Magn. Mater. 311 (2007) 187–192. 624–633.
[44] A.M. Derfus, G.V. Maltzahn, T.J. Harris, T. Duza, K.S. Vecchio, E. Ruoslahti, S.N. [66] M. Yanase, M. Shinkai, H. Honda, T. Wakabayashi, J. Yoshida, T. Kobayashi,
Bhatia, Remotely triggered release from magnetic nanoparticles, Adv. Mater. 19 Intracellular hyperthermia for cancer using magnetite cationic liposomes: an in
(2007) 3932–3936. vivo study, Jpn. J. Cancer Res. 89 (1998) 463–469.
[45] C.L. Dennis, A.J. Jackson, J.A. Borchers, R. Ivkov, A.R. Foreman, P.J. Hoopes, R. [67] M. Suzuki, M. Shinkai, H. Honda, T. Kobayashi, Anti-cancer effect and immune
Strawbridge, Z. Pierce, E. Goerntiz, J.W. Lau, C. Gruettner, The influence of induction by hyperthermia of malignant melanoma using magnetite cationic
magnetic and physiological behaviour on the effectiveness of iron oxide liposomes, Melanoma Res. 13 (2003) 129–135.
nanoparticles for hyperthermia, J. Phys. D Appl. Phys. 41 (2008) 134020. [68] (a) A. Ito, M. Shinkai, H. Honda, K. Yoshikawa, S. Saga, T. Wakabayashi, J.
[46] N. Kawai, M. Futakuchi, T. Yoshida, A. Ito, S. Sato, T. Naiki, H. Honda, T. Shirai, K. Yoshida, T. Kobayashi, Heat shock protein 70 expression induces antitumor
Kohri, Effect of heat therapy using magnetic nanoparticles conjugated with immunity during intracellular hyperthermia using magnetite nanoparticles,
cationic liposomes on prostate tumor in bone, Prostate 68 (2008) 784–792. Cancer Immunol. Immun. 52 (2003) 80–88;
[47] (a) M. Suto, Y. Hirota, H. Mamiya, A. Fujita, R. Kasuya, K. Tohji, B. Jeyadevan, (b) A. Ito, Y. Nakahara, M. Fujioka, T. Kobayashi, T. Takeda, I. Nakashima, H.
Heat dissipation mechanism of magnetite nanoparticles in magnetic fluid Honda, Complete regression of hereditary melanoma in a mouse model by
hyperthermia, J. Magn. Magn. Mater. 321 (2009) 1493–1496; repeated hyperthermia using magnetite cationic liposomes, Jpn. J. Hy-
(b) M. Lévy, C. Wilhelm, J.-M. Siaugue, O. Horner, J.-C. Bacri, F. Gazeau, perthermic Oncol. 11 (2005) 139–149.
Magnetically induced hyperthermia: size-dependent heating power of [69] B. Sivasai, R. Rajashekar, W. Hongwang, S.N. Thilani, D. RajKumar, P. Marla, K.O.
γ-Fe2O3 nanoparticles, J. Phys. Condens. Matter 20 (2008) 204133. Franklin, W. Brandon, L. Xiaoxuan, K.B. Olga, T. Masaaki, C. Viktor, B.H. Stefan,
[48] S. Bae, S.W. Lee, Y. Takemura, E. Yamashita, J. Kunisaki, S. Zurn, C.S. Kim, T.L. Deryl, A/C magnetic hyperthermia of melanoma mediated by iron(0)/iron
Dependence of frequency and magnetic field on self-heating characteristics of oxide core/shell magnetic nanoparticles: a mouse study, BMC Cancer 10
NiFe2O4 nanoparticles for hyperthermia, IEEE Trans. Magn. 42 (2006) 3566–3568. (2010) 1–9.
[49] D.H. Kim, S.H. Lee, K.N. Kim, K.N. Kim, K.M. Kim, I.B. Shim, Y.K. Lee, Temperature [70] M. Sincai, D. Ganga, M. Ganga, D. Argherie, D. Bica, Antitumor effect of magnetite
change of various ferrite particles with alternating magnetic field for nanoparticles in cat mammary adenocarcinoma, J. Magn. Magn. Mater. 293
hyperthermic application, J. Magn. Magn. Mater. 293 (2005) 320–327. (2005) 438–441.
C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808 807

[71] F. Matsuoka, M. Shinkai, H. Honda, T. Kubo, T. Sugita, T. Kobyashi, Hyperthermia [100] C.R. Thomas, D.P. Ferris, J.H. Lee, E. Choi, M.H. Choo, E.S. Kim, J.S. Shin, J.F.
using magnetite cationic liposomes for hamster osteosarcoma, BioMagn. Res. Stoddart, J. Cheon, J.I. Zink, Noninvasive remote-controlled release of drug
Technol. 2 (2004) 1–6. molecules in vitro using magnetic actuation with silica-encapsulated iron oxide,
[72] H. Matsuno, I. Tohnai, K. Mitsudo, K. Hayashi, M. Ito, M. Shinkai, T. Kobayashi, J. J. Am. Chem. Soc. 132 (2010) 10623–10625.
Yoshida, M. Ueda, Interstitial hyperthermia using magnetite cationic liposomes [101] S. Nappini, F.B. Bombelli, M. Bonini, B. Nordèn, P. Baglioni, Magnetoliposomes for
inhibit to tumor growth of VX-7 transplanted tumor in rabbit tongue, Jpn. J. controlled drug release in the presence of low-frequency magnetic field, Soft
Hyperthermic Oncol. 17 (2001) 141–150. Matter 6 (2010) 154–162.
[73] J. Motoyama, N. Yamashita, T. Morino, M. Tanaka, T. Kobayashi, H. Honda, [102] L.A. Tai, P.J. Tsai, Y.C. Wang, Y.J. Wang, L.W. Lo, C.S. Yang, Thermosensitive
Hyperthermic treatment of DMBA-induced rat mammary cancer using magnetic liposomes entrapping iron oxide nanoparticles for controllable drug release,
nanoparticles, Biomagn. Res. Technol. 6 (2008) 1–6. Nanotechnology 20 (2009) 135101/1–135101/9.
[74] B. Le, M. Shinkai, T. Kitade, H. Honda, J. Yoshida, T. Wakabayashi, T. Kobyashi, [103] H. Huang, S. Delikanli, H. Zeng, D.M. Ferkey, A. Pralle, Remote control of ion
Preparation of tumor-specific magnetoliposomes and their application for channels and neurons through magnetic-field heating of nanoparticles, Nat.
hyperthermia, J. Chem. Eng. Jpn. 34 (2001) 66–72. Nanotechnol. 5 (2010) 602–606.
[75] J. Black, Biological Performance of Materials: Fundamentals of Biocompatibility, [104] K. Hayashi, K. Ono, H. Suzuki, M. Sawada, M. Moriya, W. Sakamoto, T. Yogo, High-
3rd ed. Marcel Dekker, Inc., New York, 1999. frequency, magnetic-field-responsive drug release from magnetic nanoparticle/
[76] J. Chatterjee, M. Bettge, Y. Haik, C.J. Chen, Synthesis and characterization of organic hybrid based on hyperthermic effect, ACS Appl. Mater. Interfaces 2 (2010)
polymer encapsulated Cu–Ni magnetic nanoparticles for hyperthermia applica- 1903–1911.
tions, J. Magn. Magn. Mater. 293 (2005) 303–309. [105] F. Yang, P. Chen, W. He, N. Gu, X. Zhang, K. Fang, Y. Zhang, J. Sun, J. Tong, Bubble
[77] T.N. Brusentsova, N.A. Brusentsov, V.D. Kuznetsov, V.N. Nikiforov, Synthesis and microreactors triggered by an alternating magnetic field as diagnostic and
investigation of magnetic properties of Gd-substituted Mn–Zn ferrite nanoparticles therapeutic delivery devices, Small 6 (2010) 1300–1305.
as a potential low-TC agent for magnetic fluid hyperthermia, J. Magn. Magn. Mater. [106] E. Roduner, Nanoscopic Materials: Size Dependent Phenomena, RSC Publishing,
293 (2005) 298–302. Cambridge, UK, 2006.
[78] J. Giri, P. Pradhan, T. Sriharsha, D. Bahadur, Preparation and investigation of [107] M.C. Daniel, D. Astruc, Gold nanoparticles: Assembly, supramolecular chemistry,
potentiality of different soft ferrites for hyperthermia applications, J. Appl. Phys. quantum-size-related properties, and applications toward biology, catalysis, and
97 (2005) 10Q916. nanotechnology, Chem. Rev. 104 (2004) 293–346.
[79] H.G. Bagaria, J.L. Phillips, D.E. Nikles, D.T. Johnson, Self-regulated magnetic fluid [108] Y.W. Jun, J.W. Seo, J. Cheon, Nanoscaling laws of magnetic nanoparticles and
hyperthermia, AIChE Annual Proceedings, , 2005, pp. 14336–14340. their applicabilities in biomedical sciences, Acc. Chem. Res. 41 (2008) 179–189.
[80] S. Sun, C.B. Murray, D. Weller, L. Folks, A. Moser, Monodisperse FePt nanoparticles [109] C. Burda, X. Chen, R. Narayanan, M.A. El-Sayed, Chemistry and properties of
and ferromagnetic FePt nanocrystal superlattices, Science 287 (2000) 1989–1992. nanocrystals of different shapes, Chem. Rev. 105 (2005) 1025–1102.
[81] M. Chen, J.P. Liu, S. Sun, One-step synthesis of FePt nanoparticles with tunable [110] S. Saini, S. Goundla, B. Bagchi, Distance and orientation dependence of excitation
size, J. Am. Chem. Soc. 126 (2004) 8394–8395. energy transfer: from molecular systems to metal nanoparticles, J. Phys. Chem. B
[82] S.U. Son, Y. Jang, J. Park, H.B. Na, H.M. Park, H.J. Yun, J. Lee, T. Hyeon, Designed 113 (2009) 1817–1832.
synthesis of atom-economical Pd/Ni bimetallic nanoparticle-based catalysts for [111] B.L.V. Prasad, C.M. Sorensen, K.J. Klabunde, Gold nanoparticle superlattices,
sonogashira coupling reactions, J. Am. Chem. Soc. 126 (2004) 5026–5027. Chem. Soc. Rev. 37 (2008) 1871–1883.
[83] K.L. McNerny, Y. Kim, D.E. Laughlin, M.E. McHenry, Chemical synthesis of [112] R. Narayanan, C. Tabor, M.A. El-Sayed, Can the observed changes in the size or
monodisperse γ-Fe–Ni magnetic nanoparticles with tunable Curie temperatures shape of a colloidal nanocatalyst reveal the nanocatalysis mechanism type:
for self-regulated hyperthermia, J. Appl. Phys. 107 (2010) 09A312. homogeneous or heterogeneous? Top. Catal. 48 (2008) 60–74.
[84] (a) C.G. Hadjipanayis, M.J. Bonder, S. Balakrishnan, X. Wang, H. Mao, G.C. [113] M. Grzelczak, J. Perez-Juste, P. Mulvaney, L.M. Liz-Marzan, ChemInform Abstract:
Hadjipanayis, Metallic iron nanoparticles for MRI contrast enhancement and Shape control in gold nanoparticle synthesis. ChemInform, Chem. Soc. Rev. 37
local hyperthermia, Small 4 (2008) 1925–1929; (2008) 1783–1791.
(b) E.J. Bergey, L. Levy, X. Wang, L.J. Krebs, M. Lal, K.-S. Kim, S. Pakatchi, C. [114] C.L. Nehl, J.H. Hafner, Shape-dependent plasmon resonances of gold nanopar-
Liebow, P.N. Prasad, DC magnetic field induced magnetocytolysis of cancer ticles, J. Mater. Chem. 18 (2008) 2415–2419.
cells targeted by LH–RH magnetic nanoparticles in vitro, Biomed. Micro- [115] I. Lisiecki, Size, shape, and structural control of metallic nanocrystals, J. Phys.
devices 4 (2002) 293–299. Chem. B 109 (2005) 12231–12244.
[85] S.L. McGill, C.L. Cuylear, N.L. Adolphi, M. Osiński, H.D. Smyth, Magnetically [116] Z. Liu, D. Elbert, C.L. Chien, P.C. Searson, FIB/TEM characterization of the
responsive nanoparticles for drug delivery applications using low magnetic field composition and structure of core/shell Cu–Ni nanowires, Nano Lett. 8 (2008)
strengths, IEEE Trans. Nanobiosci. 8 (2009) 33–42. 2166–2170.
[86] N.S. Satarkar, J.Z. Hilt, Magnetic hydrogel nanocomposite for remote controlled [117] (a) L. Wang, H.-Y. Park, S.I. Lim, M.J. Schadt, D. Mott, J. Luo, X. Wang, C.-J. Zhong,
pulsatile drug release, J. Control. Release 130 (2008) 246–251. Core@shell nanomaterials: gold-coated magnetic oxide nanoparticles,
[87] J.E. Wong, A.K. Gaharwar, D.M. Schulte, D. Bahadur, W. Richtering, Layer-by- J. Mater. Chem. 18 (2008) 2629–2635;
layer assembly of a magnetic nanoparticle shell on a thermoresponsive microgel (b) A.H. Latham, M.E. Williams, Controlling transport and chemical function-
core, J. Magn. Magn. Mater. 311 (2007) 219–223. ality of magnetic nanoparticles, Acc. Chem. Res. 41 (2008) 411–420.
[88] T. Hoare, J. Santamaria, G.F. Goya, S. Irusta, D. Lin, S. Lau, R. Padera, R. Langer, D.S. [118] B. Silke, B. Helmut, N. Matoussevitch, D. Eckhard, H. Modrow, N. Palina, M.
Kohane, A magnetically triggered composite membrane for on-demand drug Frerichs, V. Kempter, M.-F. Wolfgang, A. Heinemann, M. Kammel, A. Wiedenmann,
delivery, Nano Lett. 9 (2009) 3651–3657. L. Pop, S. Odenbach, E. Uhlmann, N. Bayat, J. Hesselbach, J.M. Guldbakke, Air-stable
[89] S. Ghosh, A. Neogi, C. Yang, T. Cai, S.G. Mitra, D. Diercks, Z. Hu, Thermoresponsive Co-, Fe-, and Fe/Co-nanoparticles and ferrofluids, Z. Phys. Chem. 220 (2006) 3–40.
hydrogel microvalve based on magnetic nanoheaters for microfluidics, Mater. [119] (a) J. Zhai, M. Huang, S. Dong, Electrochemical designing of Au/Pt core shell
Res. Soc. Symp. Proc. 1095 (2008) pp. 1095-EE03-20. nanoparticles as nanostructured catalyst with tunable activity for oxygen
[90] M.H. Hsu, Y.C. Su, Iron-oxide embedded solid lipid nanoparticles for magnetically reduction, Electroanalysis 19 (2007) 506–509;
controlled heating and drug delivery, Biomed. Microdevices 10 (2008) 785–793. (b) J. Hormes, H. Modrow, H. Bonnemann, C.S.S.R. Kumar, The influence of
[91] H. Yin, S. Yu, P.S. Casey, G.M. Chow, Synthesis and properties of poly(D,L-lactide) various coatings on the electronic, magnetic,and geometric properties of
drug carrier with maghemite nanoparticles, Mater. Sci. Eng. C 30 (2010) 618–623. cobalt nanoparticles, J. Appl. Phys. Lett. 97 (2005) pp. 10R102-10R106;
[92] S. Ghosh, S.G. Mitra, T. Cai, D.R. Diercks, N.C. Mills, D.L. Hynds, Alternating (c) C.M. Sweeney, W. Hasan, C.L. Nehl, T.W. Odom, Optical properties of anisotropic
magnetic field controlled, multifunctional nano-reservoirs: intracellular uptake core–shell pyramidal particles, J. Phys. Chem. A 113 (2009) 4265–4268;
and improved biocompatibility, Nanoscale Res. Lett. 5 (2010) 195–204. (d) P. Reiss, M. Protiere, L. Li, Core/shell semiconductor nanocrystals, Small 5
[93] T.Y. Liu, S.H. Hu, K.H. Liu, R.S. Shaiu, D.M. Liu, S.Y. Chen, Instantaneous drug (2009) 154–168.
delivery of magnetic/thermally sensitive nanospheres by a high-frequency [120] (a) J.P. Lellouche, in: Challa S.S.R. Kumar (Ed.), Biofunctionalization of
magnetic field, Langmuir 24 (2008) 13306–13311. Nanomaterials, Wiley-VCH, 2005, pp. 299–329;
[94] M.C. Urbina, S. Zinoveva, T. Miller, C.M. Sabliov, W.T. Monroe, C.S.S.R. Kumar, (b) S.K. Han, R.S. Kim, J.H. Lee, G. Tae, S.H. Cho, S.H. Yuk, in: Challa S.S.R. Kumar
Investigation of magnetic nanoparticle–polymer composites for multiple- (Ed.), Nanomaterials for Medical Diagnosis and Therapy, Wiley-VCH, 2007,
controlled drug delivery, J. Phys. Chem. C 112 (2008) 11102–11108. pp. 143–188;
[95] S. Purushotham, P.E.J. Chang, H. Rumpel, I.H.C. Kee, R.T.H. Ng, P.K.H. Chow, C.K. (c) Y. Yamaguchi, R. Igarashi, in: Challa S.S.R. Kumar (Ed.), Nanomaterials for
Tan, R.V. Ramanujan, Thermoresponsive core–shell magnetic nanoparticles for Medical Diagnosis and Therapy, Wiley-VCH, 2007, pp. 310–341;
combined modalities of cancer therapy, Nanotechnology 20 (2009) 305101 (d) M. De Villiers, Y. Lvov, in: Challa S.S.R. Kumar (Ed.), Nanomaterials for
(11pages). Medical Diagnosis and Therapy, Wiley-VCH, 2007, pp. 527–556;
[96] R. Regmi, S.R. Bhattarai, C. Sudakar, A.S. Wani, R. Cunningham, P.P. Vaishnava, R. (e) X. He, X. Lin, K. Wang, L. Chen, P. Wu, Y. Yuan, W. Tan, in: H.S. Nalwa (Ed.),
Naik, D. Oupicky, G. Lawes, Hyperthermia controlled rapid drug release from Encyclopedia of Nanoscience and Nanotechnology, 1, American Scientific
thermosensitive magnetic microgels, J. Mater. Chem. 20 (2010) 6158–6163. Publishers, 2004, pp. 235–253;
[97] S. Chen, Y. Li, C. Guo, J. Wang, J. Ma, X. Liang, L.R. Yang, H.Z. Liu, Temperature- (f) Challa S.S.R. Kumar (Ed.), Mixed Metal Nanomaterials, Wiley-VCH, 2008.
responsive magnetite/PEO–PPO–PEO block copolymer nanoparticles for con- [121] T. Zhang, J.L. Stilwell, D. Gerion, L. Ding, O. Elboudwarej, P.A. Cooke, J.W. Gray,
trolled drug targeting delivery, Langmuir 23 (2007) 12669–12676. A.P. Alivisatos, F.F. Chen, Cellular effect of high doses of silica-coated quantum
[98] K. Sheng, L. Wang, P. Read, J. Larner, W. Yang, Geometrically targeted radiation dot profiled with high throughput gene expression analysis and high content
enhancer using semiconductive nanoparticles, Med. Phys. 37 (2010) 3172 (1page). cellomics measurements, Nano Lett. 6 (2006) 800–808.
[99] X. Gong, S. Peng, W. Wen, P. Sheng, W. Li, Design and fabrication of magnetically [122] N. Matoussevitch, A. Gorschinski, W. Habicht, J. Bolle, E. Dinjus, H. Boennemann,
functionalized core/shell microspheres for smart drug delivery, Adv. Funct. Mater. S. Behrens, Surface modification of metallic Co nanoparticles, J. Magn. Magn.
19 (2009) 292–297. Mater. 311 (2007) 92–96.
808 C.S.S.R. Kumar, F. Mohammad / Advanced Drug Delivery Reviews 63 (2011) 789–808

[123] (a) X.-B. Zhang, J.-M. Yan, S. Han, H. Shioyama, Q. Xu, Magnetically recyclable [146] A.P. Herrera, M. Rodríguez, M. Torres-Lugo, C. Rinaldi, Multifunctional magnetite
Fe@Pt core–shell nanoparticles and their use as electrocatalysts for nanoparticles coated with fluorescent thermo-responsice polymeric shells,
ammonia borane oxidation: the role of crystallinity of the core, J. Am. J. Mater. Chem. 18 (2008) 855–858.
Chem. Soc. 131 (2009) 2778–2779; [147] D.H. Kim, D.E. Nikles, D.T. Johnson, C.S. Brazel, Heat generation of aqueously
(b) S.H. Joo, J.Y. Park, C.-K. Tsung, Y. Yamada, P. Yang, G.A. Somorjai, Thermally dispersed CoFe2O4 nanoparticles as heating agents for magnetically activated
stable Pt/mesoporous silica core–shell nanocatalysts for high-temperature drug delivery and hyperthermia, J. Magn. Magn. Mater. 320 (2008) 2390–2396.
reactions, Nat. Mater. 8 (2009) 126–131. [148] T. Lammers, F. Kiessling, W.E. Hennink, G. Storm, Nanotheranostics and image-
[124] (a) G.F. Schneider, V. Subr, K. Ulbrich, G. Decher, Multifunctional cytotoxic guided drug delivery: current concepts and future directions, Mol. Pharm. (2010),
stealth nanoparticles. A model approach with potential for cancer therapy, doi:10.1021/mp100228v.
Nano Lett. 9 (2009) 636–642; [149] R.T. Gordon, J.R. Hines, D. Gordon, Intracellular hyperthermia a biophysical
(b) S. Sengupta, D. Eavarone, I. Capila, G. Zhao, N. Watson, T. Kiziltepe, R. approach to cancer treatment via intracellular temperature and biophysical
Sasisekharan, Temporal targeting of tumour cells and neovasculature with a alterations, Med. Hypothesis 5 (1979) 83–102.
nanoscale delivery system, Nature 436 (2005) 568–572. [150] A. Jordan, R. Scholz, J. Schüler, P. Wust, R. Felix, Arrhenius analysis of the thermal
[125] A. Jordan, Size matters. How the tiny world of nanotechnology is having a big response of human colonic adenocarcinoma cells in vitro using the multi-target,
impact on the pharma industry, Next Generation Pharma, 2007 Available from:, single-hit and the linear-quadratic model, Int. J. Hyperther. 13 (1997) 83–88.
http://www.ngpharma.com/article/Issue-4/Drug-Discovery-AND-Development/ [151] E. Pollert, P. Veverka, M. Veverka, O. Kaman, Závˇeta K., S. Vasseur, R. Epherre, G.
Size-matters/. Goglio, E. Duguet, Search of new core materials for magnetic fluid hyperthermia:
[126] J.B. Hayter, R. Pynn, Structure factor of a magnetically saturated ferrofluid, Phys. preliminary chemical and physical issues, Prog. Solid State Chem. 37 (2009) 1–14.
Rev. Lett. 49 (1982) 1103–1106. [152] R.K. Jain, The next frontier of molecular medicine: delivery of therapeutics, Nat.
[127] P. Tartaj, T. González-Carreño, C.J. Serna, Single-step nanoengineering of silica Med. 4 (1998) 655–657.
coated maghemite hollow spheres with tunable magnetic properties, Adv. [153] I. Yuzuru, N. Toru, B. Le, I.S. Kabata, O. Norifumi, K. Takeshi, S. Yoshitaka, K. Naoya,
Mater. 13 (2001) 1620–1624. N. Hayao, A carbohydrate recognition-based drug delivery and controlled release
[128] C. Wang, S. Peng, L.M. Lacroix, S. Sun, Synthesis of high magnetic moment CoFe system using intraperitoneal macrophages as a cellular vehicle, Cancer Res. 66
nanoparticles via interfacial diffusion in core/shell structured Co/Fe nanoparticles, (2006) 8740–8748.
Nano Res. 2 (2009) 380–385. [154] (a) T. Branca, Z.I. Cleveland, C. Leuschner, C. Kumar, B. Fubara, R.R. Maronpot, W.
[129] W.S. Seo, J.H. Lee, X. Sun, Y. Suzuki, D. Mann, Z. Liu, M. Terashima, P.C. Yang, M.V. Warren, B. Driehuys, Hyperpolarized 3He MRI to detect lung metastases
McConnell, D.G. Nishimura, H. Dai, FeCo/graphitic-shell nanocrystals as targeted by magnetic nanoparticles, Proc. Natl. Acad. Sci. 107 (2010)
advanced magnetic-resonance-imaging and near-infrared agents, Nat. Mater. 5 3693–3697;
(2006) 971–976. (b) C. Leuschner, C.S.S.R. Kumar, J. Zhou, W.O. Soboyejo, W. Hansel, J. Hormes,
[130] D.H. Kim, E.A. Rozhkova, I.V. Ulasov, S.D. Bader, T. Rajh, M.S. Lesniak, V. Novosad, LHRH-conjugated magnetic iron oxide nanoparticles for detection of breast
Biofunctionalized magnetic-vortex microdiscs for targeted cancer-cell destruc- cancer metastases, Breast Cancer Research and Treatment 99 (2) (2006)
tion, Nat. Mater. 9 (2009) 165–171. 163–176.
[131] F. Mohammad, G. Balaji, A. Weber, R.M. Uppu, C.S.S.R. Kumar, Influence of gold [155] B.H. Park, B.S. Koo, Y.K. Kim, M.K. Kim, The induction of hyperthermia in rabbit
nanoshell on hyperthermia of superparamagnetic iron oxide nanoparticles, liver by means of duplex stainless steel thermoseeds, Korean J. Radiol. 3 (2002)
J. Phys. Chem. C 114 (2010) 19194–19201. 98–104.
[132] E. Pollert, O. Kaman, P. Veverka, M. Veverka, M. Marysko, K. Záveta, M. Kacenka, I. [156] J.M.C. Bull, An update on the anticancer effects of a combination of
Lukes, P. Jendelová, P. Kaspar, M. Burian, V. Herynek, Core–shell La1 − xSrxMnO3 chemotherapy and hyperthermia, Cancer Res. 44 (1984) 4853s–4856s.
nanoparticles as colloidal mediators for magnetic fluid hyperthermia, Philos. [157] O. Dahl, Interaction of hyperthermia and chemotherapy, Recent Results Cancer
Trans. R. Soc. A 368 (2010) 4389–4405. Res. 107 (1988) 157–169.
[133] H. Zeng, J. Li, Z.L. Wang, J.P. Liu, S. Sun, Bimagnetic core/shell FePt/Fe3O4 [158] J. Overgaard, H.D. Suit, Time–temperature relationship in hyperthermic treatment
nanoparticles, Nano Lett. 4 (2004) 187–190. of malignant and normal tissue in vivo, Cancer Res. 39 (1979) 3248–3253.
[134] T.N. Narayanan, M.M. Shaijumon, P.M. Ajayan, M.R. Anantharaman, Synthesis of [159] V.N. Nikiforov, Magnetic induction hyperthermia, Russ. Phys. J. 50 (2007)
high coercivity core–shell nanorods based on nickel and cobalt and their 913–924.
magnetic properties, Nanoscale Res. Lett. 5 (2010) 164–168. [160] K.S. Soppimath, T.M. Aminabhavi, A.R. Kulkarni, W.E. Rudzinski, Biodegradable,
[135] A.H. Habib, C.L. Ondeck, P. Chaudhary, M.R. Bockstaller, M.E. McHenry, Evaluation of polymeric nanoparticles as drug delivery devices, J. Control. Release 70 (2001)
iron–cobalt/ferrite core–shell nanoparticles for cancer thermotherapy, J. Appl. Phys. 1–20.
103 (2008) 07A307. [161] F. Alexis, E. Pridgen, L.K. Molnar, O.C. Farokhzad, Factors affecting the clearance
[136] Q. Mu, L. Yang, J.C. Davis, R. Vankayala, K.C. Hwang, J. Zhao, B. Yan, Biocompatibility and biodistribution of polymeric nanoparticles, Mol. Pharm. 5 (2008) 505–515.
of polymer grafted core/shell iron/carbon nanoparticles, Biomaterials 31 (2010) [162] H.G. Bagaria, D.T. Johnson, D.E. Nikles, AIChE Annual Proceedings, , 2004.
5083–5090. [163] (a) C.A. Haberzettl, Nanomedicine: destination or journey? Nanotechnology 13
[137] A. Villanueva, P. de la Presa, J.M. Alonso, T. Rueda, A. Martinez, P. Crespo, M.P. (2002) R9–R13;
Morales, M.A. Gonzalez-Fernandez, J. Valdes, G. Rivero, Hyperthermia HeLa cell (b) G.M. Whitesides, The once and future nanomachine, Sci. Am. 285 (2001)
treatment with silica-coated manganese oxide nanoparticles, J. Phys. Chem. C 114 78–83.
(2010) 1976–1981. [164] C.S.S.R. Kumar, Nanotechnology tools in pharmaceutical R&D, Mater. Today 12
[138] H. Zhang, D. Pan, K. Zou, J. He, X. Duan, A novel core–shell structured magnetic (2010) 24–30.
organic–inorganic nanohybrid involving drug-intercalated layered double [165] (a) A. Chalkidou, K. Simeonidis, M. Angelakeris, T. Samaras, C. Martinez-Boubeta,
hydroxides coated on a magnesium ferrite core for magnetically controlled L. Balcells, K. Papazisis, C. Dendrinou-Samara, O. Kalogirou, In vitro
drug release, J. Mater. Chem. 19 (2009) 3069–3077. application of Fe/MgO nanoparticles as magnetically mediated hyperthermia
[139] J. Liu, S. Qiao, S.B. Hartono, G. Lu, Monodisperse yolk–shell nanoparticles with a agents for cancer treatment, J. Magn. Magn. Mater. 323 (2011) 775–780;
hierarchical porous structure for delivery vehicles and nanoreactors, Angew. (b) C. Martinez-Boubeta, L. Balcells, R. Cristofol, C. Sanfeliu, E. Rodriguez, R.
Chem. Int. Ed. 49 (2010) 4981–4985. Weissleder, S. Lope-Piedrafita, K. Simeonidis, M. Angelakeris, F. Sandiu-
[140] W. Tilly, P. Wust, B. Rau, C. Harder, J. Gellermann, P. Schlag, V. Budach, R. Felix, menge, Self-assembled multifunctional Fe/MgO nanospheres for magnetic
Temperature data and specific absorption rates in pelvic tumours: predictive resonance imaging and hyperthermia, Nanomedicine 6 (2010) 362–370.
factors and correlations, Int. J. Hyperther. 17 (2001) 172–188. [166] F. Li, W. Yan, Y. Guo, H. Qi, H. Zhou, Preparation of carboplatin-Fe@C-loaded
[141] P. Vaupel, F. Kallinowski, P. Okunieff, Blood flow, oxygen and nutrient supply, chitosan nanoparticles and study on hyperthermia combined with pharmaco-
metabolic and microenvironment of human tumors: a review, Cancer Res. 49 (1989) therapy for liver cancer, Int. J. Hyperther. 25 (2009) 383–391.
6449–6465. [167] A. Ito, M. Fujioka, T. Yoshida, K. Wakamatsu, S. Ito, T. Yamashita, K. Jimbow, H.
[142] K.N. Prasad, K. Rathinasamy, D. Panda, D. Bahadur, Mechanism of cell death Honda, 4-S-cysteaminylphenol-loaded magnetite cationic liposomes for combi-
induced by magnetic hyperthermia with nanoparticles of γ-MnxFe2 − xO3 nation therapy of hyperthermia with chemotherapy against malignant melano-
synthesized by a single step process, J. Mater. Chem. 17 (2007) 5042–5051. ma, Cancer Sci. 98 (2007) 424–430.
[143] H.G. Bagaria, D.T. Johnson, Transient solution to the bioheat equation and [168] M.A. Gonzalez-Fernandez, T.E. Torres, M. Andrés-Vergés, R. Costo, P. de la Presa, C.J.
optimization for magnetic fluid hyperthermia treatment, Int. J. Hyperther. 21 Serna, M.P. Morales, C. Marquina, M.R. Ibarra, G.F. Goya, Magnetic nanoparticles for
(2005) 57–75. power absorption: optimizing size, shape and magnetic properties, J. Solid State
[144] E.H. Wissler, Pennes' 1948 paper revisited, J. Appl. Physiol. 85 (1998) 35–41. Chem. 182 (2009) 2779–2784.
[145] J.L. Phillips, H.G. Bagaria, D.T. Johnson, Simulations of the heat generation in [169] V. P. Torchilin, Multifunctional pharmaceutical nanocarriers: promises and
magnetic fluid hyperthermia cancer treatment, AIChE annual conference, Nov. problems, in Polymeric Nanomaterials, Edited by Challa Kumar, published by
2004, Austin, TX. Wiley-VCH, 2011.

You might also like