Вы находитесь на странице: 1из 35

REVIEW

Magnetic Nanoparticles www.advhealthmat.de

Advances in Magnetic Nanoparticles for


Biomedical Applications
Vanessa Fernandes Cardoso, António Francesko, Clarisse Ribeiro, Manuel Bañobre-López,
Pedro Martins,* and Senentxu Lanceros-Mendez

1. Introduction
Magnetic nanoparticles (NPs) are emerging as an important class of biomed-
ical functional nanomaterials in areas such as hyperthermia, drug release, Nanoparticles (NPs) are particles having at
tissue engineering, theranostic, and lab-on-a-chip, due to their exclusive least one dimension in the nanometer size
range up to ≈100 nm.[1] Such nanoentities
chemical and physical properties. Although some works can be found
are categorized by high surface-area-to-
reviewing the main application of magnetic NPs in the area of biomedical volume ratio and are therefore particularly
engineering, recent and intense progress on magnetic nanoparticle research, strong, versatile, and reactive, when com-
from synthesis to surface functionalization strategies, demands for a work pared to the bulk state. These specific
that includes, summarizes, and debates current directions and ongoing properties offer new and interesting possi-
bilities of optimizing optical, mechanical,
advancements in this research field. Thus, the present work addresses the
and magnetic properties of NPs, among
structure, synthesis, properties, and the incorporation of magnetic NPs in others.[1,2]
nanocomposites, highlighting the most relevant effects of the synthesis on Particularly, magnetic NPs are of large
the magnetic and structural properties of the magnetic NPs and how these interest, having successfully demonstrated
effects limit their utilization in the biomedical area. Furthermore, this review their utility in a widespread range of appli-
next focuses on the application of magnetic NPs on the biomedical field. cations, namely magnetic fluids, magnetic
energy storage, catalysis, environmental
Finally, a discussion of the main challenges and an outlook of the future
remediation, magnetic inks, and magnetic
developments in the use of magnetic NPs for advanced biomedical applica- resonance imaging (MRI) (Figure 1).[3–7]
tions are critically provided. A large interest on those inorganic
nanomaterials with metal-based configu-
ration emerges once they can be simply
Dr. V. F. Cardoso, Dr. A. Francesko, Dr. C. Ribeiro, Dr. P. Martins manipulated using alternating current magnetic field and later
Centro de Física engaged in such numerous applications.[1] Progress in nano-
Universidade do Minho
technology and nanoparticle research field allows us to syn-
4710-057 Braga, Portugal
E-mail: pmartins@fisica.uminho.pt thesize magnetic NPs with precise morphology and to suitably
Dr. V. F. Cardoso modify particle surfaces, manipulating their characteristics for
MEMS—Microelectromechanical Systems Research Unit precise applications. For example, green chemistry methods
Universidade do Minho are already being used to synthesize magnetic dextran-coated-
4800-058 Guimarães, Portugal gadolinium nanoparticles at low temperature (up to 80 °C) for
Dr. C. Ribeiro biomedical applications on the whole body level, sparkling the
CEB—Centre of Biological Engineering
University of Minho kidneys, liver, spleen, tumor, and tumor angiogenesis without
Campus de Gualtar promoting toxicity (Figure 1).[6] Other extensive studies have
4710-057 Braga, Portugal been carried out, and protocols have been developed aiming the
Dr. M. Bañobre-López optimization of magnetic nanoparticle characteristics such as
INL—International Iberian Nanotechnology Laboratory composition, surface charge, shape, size and size distribution,
4715 Braga, Portugal
and magnetic properties.[8]
Prof. S. Lanceros-Mendez
With the latest evolution and demands of nano-biotech-
BCMaterials
Parque Científico y Tecnológico de Bizkaia nology, magnetic NPs are attracting increasing attention due to
48160 Derio, Spain their potential to improve conventional therapeutic procedures
Prof. S. Lanceros-Mendez and traditional clinical diagnostic, as well as to introduce novel
IKERBASQUE approaches in biomedicine and tissue engineering (TE).[2,9]
Basque Foundation for Science Magnetic NPs are typically classified into pure metals, metal
48013 Bilbao, Spain
oxides, and magnetic nanocomposites. The most popular mag-
The ORCID identification number(s) for the author(s) of this article
netic NPs in the biomedical field are Co, Fe, Ni, Ti, iron oxide,
can be found under https://doi.org/10.1002/adhm.201700845.
and some ferrites (BaFe12O19 and CoFe2O4). Among them, iron
DOI: 10.1002/adhm.201700845 oxide NPs (typically Fe2O3 or Fe3O4) are the most used due to

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (1 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

their lower toxicity.[9] The key issues affecting the biocompati-


bility and toxicity of such materials are the characteristics of the Pedro Martins graduated in
component that is magnetically reactive, such as cobalt, iron, Physics and Chemistry in
nickel, samarium–cobalt, or neodymium–iron–boron, the size 2006 and received the Ph.D.
of the NPs, and their coating and core.[2] Knowing that mag- degree in Physics in 2012,
netic NPs can reveal novel phenomena, such as super-paramag- from the University of Minho,
netic behavior, high magnetic field irreversibility, and additional Braga, Portugal. He is now
anisotropic contributions,[2] the specific biomedical application a PostDoc researcher in the
is determined by the distinct magnetic properties. Such proper- University of Minho, Braga,
ties are, in turn, ruled by the type of magnetic nanoparticle, the Portugal and his work is
synthesis procedure, the interaction between NPs, and the focused on polymer-based
nano­­particles’ size, shape, and distribution. In this way, an magnetoelectric materials
appropriate synthesis procedure must be selected to achieve and electroactive polymers
specific and precise performance.[1] for advanced technological applications. He collaborates
From the different magnetic NPs that have been devel- with the Basque Country University, Spain; Wollongong
oped, the ones that exhibit fast change of magnetic state with University, Australia; and Cambridge University, UK, among
the application of an external magnetic field and no rema- others.
nence at room temperature (super-paramagnetism) are usually
desired.[5,10] Thus, these NPs must combine high magnetic sus-
Senentxu Lanceros-Mendez
ceptibility and loss of magnetization after removal of the mag-
was born in Bilbao, Spain.
netic field.[11]
He graduated in physics at
The appropriate functionalization of the magnetic NPs for
the University of the Basque
biomedical applications is ruled not only by their intrinsic
Country, Leioa, Spain. He
magnetic properties (magnetic saturation (Ms) and Curie
obtained his Ph.D. degree at
temperature (Tc)) but also by their biophysical properties (col-
the Institute of Physics of the
loidal stability, nontoxicity, specific absorption rate (SAR), and
Julius-Maximilians-Universität
biocompatibility) under pH conditions similar to the physio-
Würzburg, Germany. He
logical ones. To avoid magnetic NPs’ agglomeration, considera-
was a Research Scholar at
tions on the nanoparticle surface chemistry must be taken into
Montana State University,
account and should be stabilized by covering them with dif-
Bozeman, MT, USA and a
ferent types of materials, including inorganic and organic coat-
visiting scientist at The Pennsylvania State University, USA
ings.[1] Additionally, demands from the biomedicine require
and University of Potsdam, among others. He is Associate
magnetic NPs that are water stable at pH ≈ 7 and with salinity
Professor at the Physics Department of the University of
values close to the physiological ones. Such colloidal stability
Minho, Portugal (on leave), and from 2012 to 2014 he
will be dependent on NPs’ dimensions, surface chemistry, and
was also Associate Researcher at the INL—International
charge.[12] Other limitations or requirements strongly rely on
Iberian Nanotechnology Laboratory. Since 2016, he has
the use of the NPs, whether for in vitro or in vivo applications.
been Ikerbasque Professor at the BCMaterials, Basque
In the case of in vitro applications, the limitations are not as
Center for Materials, Applications, and Nanostructures,
restricted as in the in vivo case. For in vivo applications, the
Derio, Spain. His work is focused on the area of smart and
magnetic structures need to be covered with a biocompatible
functional materials for sensors and actuators, energy, and
polymer (if the nanoparticle is not biocompatible) after or
biomedical applications.
during the synthesis procedure to avoid toxicity, decrease the
risk of blood capillary obstruction, and prevent nanoparticle
aggregation.[5,9]
Due to their super-paramagnetism, some magnetic
NPs can be precisely collected in a desired location by an bridging sites for adjusting Trastuzumab, respectively, that
applied magnetic field, which is essential for target drug and represented a key paradigm development in cancer treatment
gene delivery,[9,13,14] allowing new therapeutic approaches and nanocarrier design in a preclinical application environ-
of merging multistage short-term magnetic control and ment (Figure 2).[14]
directing with enhanced mediated-ligand targeting in Furthermore, magnetic NPs are being developed for hyper-
recently developed nanodelivery systems.[14] A good example thermia and heat-activated drug release as a result of their
of such attractive capacity of super-paramagnetic NPs is the heating ability in high-frequency magnetic fields.[9,15]
synthesis of Trastuzumab-functionalized DOX-encapsulated Large efforts are being spent on the improvement of hyper-
magnetic nanocapsules (T-DOX-MNCs) through a simple thermia techniques for clinical uses. Advances in magnetic nano­­
double emulsion process reported by Chiang et al. The particle research contributed to the fast and, sometimes, dis-
authors have used synthesized antibody-targeted super-par- ruptive development of magnetic hyperthermia (MH), making
amagnetic iron oxide nanoparticles and poly(vinyl alcohol) this technique a promising tool for cancer treatment due to
(PVA)/poly (methacrylic acid), which include the capacity the possibility of targeting cancerous tissues. This approach
of magnetically targeting tumor localization and adequate leads to lower side effects than traditional radiotherapy and

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (2 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 1.  Scheme of the synthesis of gadolinium-based NPs and their magnetic resonance imaging performance at the whole body level, tumor, and
tumor angiogenesis. Reproduced with permission.[6]

chemotherapy. Similarly, controlled drug release trough magnetic to be not only able to be used as agents for near-infrared (NIR)
NPs has an essential role in the future of personalized medi- fluorescence imaging but also for MRI. After in vivo studies,
cine; once recent/ongoing clinical tests revealed substantial the composite multifunctional nanoparticles were capable of
reduction of such side effects (nausea/vomiting, fatigue, con- ablating tumor xenograft effectively with NIR laser exposure
stipation, and fever/chills).[16,17] The outstanding safety profiles and can be used in the future for cancer treatment studies.
and appropriate magnetic properties of super-paramagnetic Additionally and closely connected to the theranostics con-
iron oxide nanoparticles make them very good platforms for cept, lab-on-a-chip (LOC) technologies are assumed as one of
the design of cancer treatments that have negligible side effects the most relevant potential tools for in vitro medical analyses.
and with the capacity to target cancers that cannot be obtained Nevertheless, the integration of truly multifaceted functions in
by other types of nanostructures.[17] a very narrow space still remains challenging. NPs are an inno-
Due to the synergic effect of magnetic NPs on cell’s differ- vative option, allowing noncontact manipulation of physical,
entiation, proliferation and adhesion, one of the most popular chemical, and biological samples.[21]
areas of investigation in the biomedical field is the development Despite some reviews that can be found on the applica-
of functional tissue engineering systems that can be triggered tion of magnetic NPs in the biomedical area, recent progress
by an external magnetic field.[18] on magnetic NPs, from synthesis to surface functional strate-
Regarding the use of magnetic NPs for theranostic, NPs gies and applications, demands for a comprehensive work that
have the ability of providing in vivo real-time information, includes, summarizes, and discusses current directions and
drug administration, optimal dosage selection, and therapeutic ongoing advances in this interesting and fast-growing research
response monitoring, though there are still some missing field.
improvements in order to ensure successful clinical applica- In this way, the first part of this review deals with the
tions. As the capabilities and multifunctionality of magnetic structure, synthesis, and properties of magnetic NPs, as
NPs for theranostic continue to growth, it is expected that this well as their incorporation into nanocomposite systems. A
research field will play a key role in the new era of personal- particular emphasis will be given to the most pertinent syn-
ized medical care by allowing more efficient diagnostic and thesis effects on the structural and magnetic properties of the
therapeutic strategies.[19] In addition to this promising future, magnetic NPs and how they limit their use in the biomedical
encouraging results are already started to appear as the quan- area. The second part will focus on the biofunctionalization
titative analysis performed by Peng et al.[20] which proves the and technological application of such magnetic NPs in hyper-
ability of multifunctional iron oxide nanoparticles modified thermia, drug release, tissue engineering, theranostic, and
with YGRKKRRQRRR (TAT) peptide to obtain higher accumu- lab-on-a-chip.
lation in the nuclei, when compared to iron oxide nanoparti- Finally, a brief summary and some future perspectives and
cles without modification (Figure 3). Furthermore, the TAT- trends in the use of magnetic NPs for advanced biomedical
modified multifunctional iron oxide nanoparticles were found applications will be outlined.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (3 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 2.  Schematic representation of the functionality and synthesis of dual-targeting T-DOX- multifunctional magnetic nanocarriers and the influence
of modulating the trastuzumab density and respective use on intratumoral distribution. Reproduced with permission.[14]

2. Magnetic NPs
In particular, MNPs are a class of nanomaterials with the
2.1. General Structure potential to revolutionize the biomedical field due to their
multi­functional properties such as small size, high operational
NPs are submicrometer moieties made of organic or inorganic surface areas, low sedimentation rates, and optimized tissular
materials and with unique characteristics compared to their diffusion, and because they can be manipulated under an
bulk counterparts. Besides their intrinsic nanoscale physical applied magnetic field. Such MNPs have been prepared using
phenomena, their often exhibit exceptional optical, electrical, different compositions and phases, involving pure metals such
magnetic, and chemical properties, attracting vast attention in as Fe, Co, Ti, and Ni; metal oxides such as Fe3O4 and γ-Fe2O3;
many biotechnological areas,[22–24] such as magnetic resonance ferrites such as BaFe12O19, SrFe12O19, and MFe2O4 (M cor-
imaging,[25,26] drug delivery systems,[27,28] hyperthermia[29,30] responds to Cu, Ni, Mn, Mg, among others); and metal alloys
and labeling and separation of cells[31,32] and proteins,[33,34] such as CoPt and FePt.[9,43] Highly magnetic-responsive mate-
among other biological entities.[35,36] Different kinds of NPs, rials, such as Co and Ni, are toxic and are liable to oxidation,
such as quantum dots,[37] carbon nanotubes,[38] gold NPs,[39] which impedes their use in most of the biomedical applications.
silica NPs,[40] polymeric NPs,[41] and magnetic NPs (MNPs)[42] On the other hand, iron oxide NPs, namely magnetite (Fe3O4)
have been designed and synthesized. and maghemite (γ-Fe2O3), are the most extensively studied and

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (4 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 3.  Schematic illustration of iron oxide NPs-YGRKKRRQRRR-peptide- covalently conjugating trans-ferrin (IONP-TAT-Tf) synthesis and nuclear
targeted approach aiming operational cancer therapy. Reproduced with permission.[20]

applied ones in the biomedical field for their biocompatibility, 2.2. Magnetic Considerations
super-paramagnetic behavior, and chemical stability at room
temperature.[43] Magnetite shows a cubic symmetry with the The property of super-paramagnetism is beneficial over fer-
spinel structure (Figure 4a). The oxygen atoms are arranged in romagnetism for several biomedical applications, as super-
a face-centered cubic (fcc) lattice, where the Fe2+ ions occupy paramagnetic NPs show a high magnetic moment and work
half of the octahedral sites and Fe3+are split evenly across the as a giant paramagnetic atom with a fast response to external
remaining octahedral sites and tetrahedral sites. Maghemite magnetic fields with insignificant coercivity and remanence at
is the fully oxidized form of magnetite and shows an inverse room temperature.[46,47] Nevertheless, ferromagnetic NPs can
spinel structure with some cation deficiency (Figure 4b). Thus, be super-paramagnetic when they show single domains, typi-
the structures of maghemite and magnetite are similar but cally with sizes below 20 nm.[48] However, an inevitable issue
with vacancies, where Fe3+ ions are still occupying octahedral associated with particles in this size range is their intrinsic
and tetrahedral sites, but the vacancies of maghemite have a instability and trend to aggregate in order to minimize the high
preference for the octahedral sites. Specifically, one out of every surface energy resulting from their large surface-to-volume
six octahedral sites in magnetite is vacant in maghemite struc- ratio.[49] Moreover, naked iron oxide NPs are chemically highly
ture.[44] Magnetite is sensitive to oxidation and usually trans- active and easily oxidized in air, resulting, usually, in a loss of
forms to maghemite in the presence of oxygen. The oxidation magnetism and dispersity. Thus, for most of the applications,
happens frequently from the migration of the positive ions proper surface modifications or coating—using, for example,
through the lattice framework. Under such an oxidation pro- surfactants, polymers, silica, or carbon—are required to retain
cedure, the cationic vacancies will be formed to maintain the the stability of the MNPs, especially in water at pH ≈ 7 and
charge.[45] physiological environment.[4] In almost all cases, the used shell
not only stabilizes and protects the MNPs from biodegradation
but can also confer biocompatibility or high surface area for fur-
ther functionalization with other NPs or ligands, depending on
the desired application, giving rise to complex coated and mul-
tiple functional surface engineered MNPs.[50] They can bind to
proteins, antibodies, enzymes, nucleotides, or drugs enabling
some exciting new approaches for bioseparation, biodetection,
and targeted drug delivery, and can be absorbed to an organ,
tissue, or tumor using an external magnetic field.[51,52] In addi-
tion, they can also respond resonantly to alternating magnetic
fields (AMFs) and function as a heater, offering a promising
therapeutic for use in hyperthermia.

2.3. Main Synthesis Methods


Figure 4.  Crystalline structure and crystallographic data of the a) mag-
netite and b) maghemite (the black spheres are Fe2+, the green spheres Several synthesis methods have been developed over the
are Fe3+, and the red spheres are O2−). Adapted with permission.[44] years to obtain MNPs with controllable size, size distribu-
Copyright 2015, IOP Science. tion, shape, surface chemistry, magnetic characteristics, and

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (5 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

reproducibility.[43] In fact, from a fundamental scientific point Hydrothermal processes are used to synthesize MNPs in
of view, the synthesis of uniform-sized NPs with controllable aqueous media in reactors or autoclaves at high temperatures
size, shape, and surface is very important in many biotechno- (higher than 200 °C) and high pressure (higher than 2000 psi),
logical fields and is generally challenging. Moreover, they must resulting in a rapid nucleation and fast growth of the newly
be made of a nontoxic and non-immunogenic material and formed MNPs, which therefore lead to the formation of small-
when it comes to in vivo applications, MNPs should be small sized MNPs.[67,68] Hydrolysis and oxidation is one main route
enough to be stable in the biological circulation and to transit adopted for forming MNPs under hydrothermal synthesis, this
via the capillary systems of tissues.[53] A short review of some being the neutralization of mixed metal hydroxides other highly
common procedures used for the synthesis of MNPs is sum- used route.[11] After the completion of the reaction, the auto-
marized in the following sections. claves are cooled down to room temperature, and the MNPs are
The coprecipitation technique is perhaps the mostly adopted, washed with several solvents to eliminate impurity agents.[53,69]
simplest, and most efficient chemical pathway to synthesize However, Since the reaction undergoes in an automatic way,
MNPs with an average diameter below 50 nm from aqueous it is challenging to understand the hydrothermal processes
salt solutions.[54] Technically, the synthesis procedure is car- through their mechanistic principles, which, in turn, is diffi-
ried out by a stoichiometric mixture of ferric and ferrous salts cult for the development of a universal theoretical framework,
in aqueous solution, followed by the addition of a base, e.g., remarkable progresses have been achieved in the synthesis of
NH4OH or NaOH to form MNPs, following a chemical reaction: MNPs by hydrothermal techniques in order to better control
M2+ + 2Fe3+ + 8OH− → MFe2O4 + 4H2O, where M represents the MNP phases and morphologies by properly studying and
Fe2+, Co2+, Cu2+, Zn2+, Mn2+, or Ni2+.[55] After the pioneering optimizing relevant experimental conditions such as tempera-
work developed by Massart in 1981,[56] numerous investigations ture, reaction time, reactants concentration, stoichiometry, and
have been performed not just to study and optimize the effects nature of the solvent, among others.[70] Moreover, adaptation
of the synthesis parameters such as the nature and concentra- of the hydrothermal processes has been carried out to increase
tion of salts, pH and ionic strength of the medium, reaction the kinetics of crystallization by means of localized microwave
temperature, presence of oxygen, injection flux rates, but also heating, resulting in a more convenient and fast reaction time
adapting the procedure itself [57–59] in order to control and tailor in comparison with conventional hydrothermal methods.[71,72]
the size, shape, morphology, magnetic behavior and composi- Microemulsion is an alternative method used to synthesize
tion of the MNPs.[11,60,61] In addition, surfactants or polymers shape and size-controlled MNPs based on an isotropic and
have been introduced in the reaction in order to control and thermodynamically stable systems composed by two immis-
stabilize the surface of the MNPs to avoid NPs aggregation and cible phases: a polar phase—usually water—and a nonpolar
to increase biocompatibility.[60,62] Despite the successful produc- phase—commonly oil—in the presence of a surfactant.[73] The
tion of MNPs at high process yield and high magnetization, surfactant molecules, which commonly include cetyltrimeth-
MNPs with broad size distributions and irregular morphologies ylammonium bromide, bis(2-ethylhexyl) sulfosuccinate, poly-
are usually obtained by coprecipitation.[45] vinylpyrrolidone (PVP), produce a monolayer at the water–oil
High-temperature thermal decomposition appears as an interface, with the hydrophobic tails of the surfactants dis-
interesting alternative procedure to obtain monodispersed solved in the oil phase and the hydrophilic head groups in the
NPs with high level of size control, shape, and high crystal- aqueous phase.[44] In such a system, where the polar aqueous
linity. In this approach, organometallic precursors, such as phase could contain metal salts or/and other compounds, the
[Mn+(acetylacetonate)n] (M is Fe, Mn, Co, Ni, or Cr; and n corre­ oil phase may be composed by a complex mixture of olefins
sponds to 2 or 3; Mx(N-nitrosophenylhydroxylamine)x) or car- and hydrocarbons.[44] The two basic types of microemulsions
boxyls (such as Fe(CO)5 or Fe(Cup)3) are decomposed inside are based on oil droplets dispersed in a persistent water phase
a nonpolar boiling solvent with the presence of a stabilizer (o/w) and water droplets dispersed in a persistent oil phase
such as oleic acid, 1-octodecene, hexadecylamine, under high- (w/o).[74] Thus, the nanodroplets, employed as nanoreactors,
temperature conditions.[44,63,64] The stabilizer has the ability create an appropriate environment for controlled nucleation
to slow down the nucleation process, which may inhibit the and growing of MNPs.[65,75] The walls of the nanodroplets
growth and favor the formation of MNPs with a spherical made by the surfactant molecules act as cages for the growing
shape and a size of <30 nm. Decomposition of precursors course, reducing the average size of the particles during the
with cationic metal centers leads directly to metal oxide NPs, collision and aggregation processes. Therefore, the size and
while organometallic precursors in which metal has zero dynamic formation of the MNPs can be easily controlled by the
valence in their composition initially lead to the formation droplet size, initial concentration of reactants, and the nature
of metal NPs, but when followed by oxidation they can lead of surfactants.[76,77]
to high-quality monodispersed metal oxides.[65] Three main In addition to these popular methods, MNPs have been suc-
factors control the size and shape of the obtained MNPs, cessfully synthesized by means of other techniques including
which include the decomposition temperature, the precursor/ chemical vapor deposition,[78,79] combustion,[80,81] carbon
capping agent ratio, and the duration of the reaction after arc,[82,83] electrochemical synthesis,[84,85] laser pyrolysis tech-
reaching the boiling point.[44,66] Despite the referred advan- niques,[86,87] microbial synthesis[88,89] and in lab-on-a-chip sys-
tages, thermal decomposition usually leads to complicated tems,[90,91] among others. Representative published reviews
processes at relatively high temperatures and to the produc- concerning existing synthesis routes to obtain MNPs along
tion of organic-soluble MNPs that may limit their applica- with their physicochemical properties, toxicity, and biocompat-
bility in the biomedical field. ibility can be found in refs. [11,44,45,55]. The magnetization and

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (6 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

coercive field of different representative magnetic nanoparticles charged iron oxide NPs form aggregates easily and are more
used in the biomedical field can be consulted in ref. [92]. likely to interact with usually negatively charged cell surfaces
and facilitate nanoparticle uptake by cells. Repeated systemic
administration of high-concentration positively charged NPs
2.4. Biofunctionalization may lead to enhanced nonspecific uptake into cells, causing
toxicity in normal tissues.[110,111] Neutral and negatively charged
Besides the need to produce NPs with required size, shapes, NPs, on the other hand, are recommendable for the production
and monodispersity, for biomedical applications, it is of para- of targeted nanoparticle-imaging probes and drug carriers for
mount importance that NPs are also biocompatible. Coating of in vivo applications, especially in cases where long circulation
NPs is a strategy to prevent or reduce toxicity and undesired times through blood are required.[112] Finally, various proteins,
toxic side effects.[93] Nevertheless, biofunctionalization of mag- such as human serum albumin,[103] have also been tested for
netic NPs is not only directed to improve the biocompatibility nanoparticle coating to study cell labeling. Proteins are excellent
with cells, but also to modify the cell interaction processes, choice once, besides stabilization of magnetic NPs, they offer
pharmacokinetics, and biodistribution of the NPs.[94,95] For availability for a wide variety of functional groups, for further
instance, the residence time of circulating iron oxide NPs can functionalization routes with drugs or other active agents. In
be doubled by simple functionalization with “stealth” polymer, addition, proteins being natural polymers are easily cleared from
polyethylene glycol (PEG), while at the same time, the particle the body and dissociate in cells allowing agglomeration of mag-
accumulation in different organs can be reduced.[96] The (bio) netic NPs inside the target to enhanced MRI contrast effect.[113]
functional layers that shell magnetic nanoparticle cores aim at Toxicity remains the major concern for nanosystems in
(i) protect the therapeutic and targeting agents during the circu- biomedical applications, although in vivo studies on animals
lation in the body, (ii) improve their delivery efficiency, and (iii) repeatedly show minimal damage in the case of MNPs. In vitro
avoid undesired accumulation in important organs. and in vivo studies raised initial concerns after discovering that
Surface of MNPs can be engineered to feature multifunc- many NPs, including the magnetic ones, induce formation of
tionality combining, e.g., prevention, diagnosis, and treatment reactive oxygen species (ROS) after internalization into cells,
of diseases. Intrinsic properties of MNPs allow for external which results in the alteration of the levels of expression of
control and targeting the desired cells/tissues via the magnetic cell proliferative responsive genes.[114] Biocompatible coatings
core, which, at the same time might be also explored as an MRI reduce to different extents the toxicity following administra-
contrast agent. On the other hand, a biocompatible coating pro- tions, as shown by in vivo studies on mice and monitoring the
vides a therapeutic effect.[97] If a surface charge is imparted to no long-term toxicity in the major organs. Different response of
these nanostructures, it may further contribute to colloidal sta- organs to exposure to NPs is due to their different biodistribu-
bility, circulation time, cellular uptake and/or nontoxicity. Most tion following administration. It has been also shown that non-
unmodified MNPs show low stability and easily form aggre- targeted super-paramagnetic iron oxide NPs accumulate mostly
gates, which negatively affect the reactive surface area, bio- in liver (86%) and spleen (6.2%), interestingly already within
availability, and toxicity.[98] Surface modifications are directed 1 h following intravenous injection. For a half-life of 3–4 d, it
to increase stability and provide functional groups for biocon- is necessary to be cleared out from these organs, whereas the
jugation; however, this may also lead to a decrease in magnetic incorporation of nanoparticles into hemoglobin of erythrocytes
response due to the coating thickness.[99] The most studied raises more concerns.[115] Furthermore, studies of targeted mag-
surface coatings are polymer based, including dextran and its netic NPs show a lack of apparent systemic toxicity even after
derivatives,[100,101] PEG and PEG copolymers,[102] proteins,[103] long-term subjection to administration (up to 3 months).[116]
and antifouling polymers.[104] Dextran was the first extensively These investigations encouraged safety, biodistribution, and
investigated biopolymer to coat MRI-contrast MNPs, studied, pharmacokinetic studies of approved magnetic NPs in human
among others, in experimental animal models and in humans. patients. Super-paramagnetic iron oxide nanoparticle-based fer-
Similar to PEG, dextran enables long-circulating properties for umoxytol, an Food and Drug Administration (FDA)-approved
systemic delivery of iron oxide NPs with suitable accumulation agent for treatment of iron deficiency anemia,[117,118] was clini-
rates in tumors for MRI detection.[105] Long circulation is espe- cally tested in over 2000 patients for MRI detection of lymph
cially useful for nontargeted detection nanosystems, as proved node metastasis. The studies revealed safe doses for adminis-
for dextran-coated super-paramagnetic iron oxide NPs used as tration (2.9 mg Fe per kg in humans), while it was previously
MRI contrasts for the detection of liver tumors or lymph node demonstrated that NPs with 30 nm in size were uptaken by
metastases in humans.[106–108] Anti-biofouling surface modifica- the macrophages in the spleen, liver, and lymph nodes.[119]
tions have been shown to best reduce nonspecific interactions Magnetic-iron-based NPs thus will likely show lower safety con-
of magnetic NPs with serum proteins and macrophage uptake cerns than other nanomaterials for the translational develop-
during systemic delivery. Specially designed low-fouling PEG- or ment of new theranostic agents for image-guided and targeted
polyethylene glycol (PEO)-copolymeric systems, besides biocom- cancer therapeutic approaches.[120]
patibility and tumor-targeting ability, provide exceptionally long- The FDA- or Europe-approved MNP-based formulations are
circulated times in vivo with significantly reduce nonspecific paving the way for other similar formulations and different
binding to serum proteins and uptake by macrophages in the biomedical applications. Careful formulation and considera-
liver and spleen.[104,109] Studies show that nanoparticle surface tion of biofunctional components to the shell of the magnetic
charge plays also an important role in interaction with a wide cores allow for identification of the most suitable solutions
variety of biological molecules during circulation. Positively based on the expected performance of the multifunctional

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (7 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

NPs.[121] Targeted accumulation at the site of interest with cytoprotection,[129,130] and imaging purposes.[126] The current
minimal losses during circulation is necessary to achieve pre- state-of-the-art indicates that although the process of metal–
cise diagnosis of diseases and eventually real-time monitoring phenolic coordination is thoroughly studied, the engineering
of the treatment efficacy, the cornerstone of the personalized of functional materials using this coordination and simultane-
therapy concept, while using toxicity-free concentrations. The ously addressing different biofunctional requirements is still in
multifunctional NPs should be further designed as carriers its infancy. As the properties of the assembled supramolecular
to protect the therapeutics against cleavage in the body, i.e., networks largely depend on the coordinated metal species,
premature degradation during the circulation.[122] However, the next challenge is to master the coordination in order to
advances in the multifunctionality concept are the key to clin- tailor materials with desired functional properties.[131] How-
ical and market product approval. Overall, substantial research ever, developing bulk materials with combined mechanical and
efforts are being carried out to integrate nanoparticle systems functional properties remains a significant challenge. Excellent
with multiple functionalities, resulting in relevant advances in trials are being carried out with the development of a class of
the field. However, obstacles such as nonspecific distribution metallogels formed by direct gelation of tannic acid polyphenol
and inadequate accumulation of actives are still important chal- and different metal ions. These metallogels combine self-
lenges to overcome. healing properties with transparency, while various materials
can be added by in situ cogelation to increase the robustness
and flexibility required in a variety of chemical, biomedical, and
2.5. Nanocomposites environmental applications.[132]
Perhaps the most straightforward approach to fabricate
There is a significant scientific and technological interest on bulk materials with the required characteristics is the direct
developing nanocomposite materials based on the coordination assembling of metal NPs with polyphenols, as both structural
of magnetic inorganic components and organic compounds. and functional components. In the case of MNPs, their fea-
The implementation of inorganic components into the bulk tures could be explored directly within the material. In addi-
organic matrix without affecting their intrinsic properties can tion to the intrinsic magnetic properties, these NPs can serve
provide high-performance novel materials for biomedical pur- as centers of multiple coordination and hence exhibit localized
poses. Magnetic NPs typically suffer of low processability in increased functionality (Figure 5). Nevertheless, the excellent
terms of shaping and adapting to a particular application. performance of magnetic NPs in nanomaterial and biomedical
Many organic compounds found in nature, such as polymers applications often relies on achieving the attachment of suit-
and polyphenols, are more flexible components extracted or able polyphenolic ligands to the nanoparticle surface both in
produced at a low cost. They allow easy processing and can be sufficient numbers and with proper orientation.[133] The nature
shaped into a wide variety of materials using traditional and of ligand binding does not only solely depend on the presence
advanced processing methods. The integration of inorganic of suitable functional groups, but also on relative positioning of
NPs into an organic matrix would thus open new possibilities these groups within the ligand and dynamics of ligand exchange
for exploiting functionalities of both in a combined composite on the nanoparticle surface. This is probably the reason why
material. the literature is still scarce on direct incorporation of magnetic
Techniques for materials assembled by supramolecular NPs into polymer-based coordinated materials, and this review
coordination chemistry between polyphenols, found in many will briefly highlight the most representative findings in fabri-
plants and different metal species (e.g., metal ions), became cation of surface coatings and hydrogel nanocomposites.
the matter of interest for numerous applications. On its basis, A chitosan–polyphenol conjugate was synthesized by Zvarec
this assembly is inspired from nature’s organic/inorganic com- et al.[134] for the coordination of super-paramagnetic iron oxide
posites with high mechanical strength found in squid beaks NPs in an organic/inorganic bio-nanocomposite. This hybrid
and mussel thread coatings. The recent interest led to the material mimics the interfacial chemistries of squid beaks
development of thin films with advanced and dynamic proper- and mussel thread coating. The MNPs offered a significant
ties. Importantly, this supramolecular self-assembly occurs in improvement in the functionality of the biomaterial conjugates,
one step and can serve for material development or coating whereas inorganic/organic interface was strengthened by coor-
of various interfaces, including a range of planar, biological, dination bonding, resulting in a nanocomposite that able to
organic, and inorganic particle templates. Material develop- support challenging physiological pH conditions and exhibits
ment is started by the polyphenol adsorption and controlled by great thermal stability at high temperatures. The presence
pH-dependent and multivalent coordination bonding.[123] More- of super-paramagnetic nanoparticles also made composites
over, this self-assembly is achieved in aqueous medium within stimuli-responsive.[134]
minutes; thus, the technique is low cost, scalable and combined Another study that demonstrated a universal strategy for the
with pH responsiveness of materials and negligible cytotoxicity, encapsulation of inorganic NPs, including iron oxide ones, into
and makes these materials very useful for technological bio- distinct polymer phases was reported by Yabu et al.[135] These
medical applications. NPs were independently encapsulated in two distinct mussel-
Self-assembly via polyphenol coordination of a variety of inspired amphiphilic copolymers. After inorganic–organic com-
metal species indeed offers the possibility of fabricating supra- posite, Janus particles were obtained through solvent evapo-
molecular materials customized for desired properties,[124,125] ration from the dispersion containing the nanoparticle and
including stimuli-responsiveness. Such supramolecular the polymer. These hybrid particulates were found to rotate
materials are currently investigated for drug delivery,[126–128] extremely quickly in response to external magnetic fields. Such

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (8 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 5.  Strategy for coordination of magnetic NPs with biopolymer–polyphenol conjugates. Reproduced with permission.[134] Copyright 2013, RSC.

features can be potentially useful for many biomedical appli- mortality worldwide. According to the World Health Organiza-
cations, including hyperthermia for cancer treatment and tar- tion,[63] despite increasing national expenditures for cancer care
geting tumor sites. (at least, in developed countries), the incidence of this ageing-
A study that revealed insights into interactions between related disease is expected to rise by about 70% over the next
polymer molecules and inorganic NPs in nanocomposite two decades. Therefore, innovative and targeted policies are of
mechanics and interfacial interaction dynamics was recently urgent demand to tackle this threatening problem worldwide,
published.[136] Iron oxide NPs were incorporated into a cat- which is emphasized by the incessant increase of aged people.
echol-modified polymer (PEG and PEGCOOH) network to In this scenario, nanotechnology is a powerful tool with the
obtain hydrogels cross-linked via metal coordination bonds expectation of revolutionizing the entire healthcare scene in the
at the nanoparticle surfaces (Figure 6). The supramolecular future, especially in the fight against cancer. Currently, chemo-
cross-linking of the gels is thus achieved to provide solid-like, therapy, radiotherapy, and surgery are the three clinically avail-
yet reversible, hydrogel mechanics. This work highlighted the able therapies in cancer treatment. Alone or in combination
relation between macroscale mechanics of nanocomposite constitutes the existent toolbox to treat cancer patients in the
materials and nanoscale dynamics at the polymer–nanopar- clinics. However, all of them have serious drawbacks mainly
ticle interface. It was also demonstrated how magnetic NPs related to the lack of specificity and harmful secondary effects.
can be incorporated into functional bulk materials (e.g., cross- As an adjuvant alternative, thermotherapy has been tradition-
linked hydrogel networks). The foundation for future design ally used to kill tumor cells in two different ways: on the one
of stimuli-responsive smart materials with remotely controlled hand by hyperthermia, in which the temperature of a local
mechanical and functional properties was also addressed. region or the whole body is increased up to 41–45 °C by ultra-
sounds, microwaves, or radiofrequency radiation;[138] on the
other hand, by thermal ablation, in which a temperature above
3. Representative Biomedical Applications 45 °C is applied to the diseased area of interest that destroys the
tissue.[139] Both methods are based on the more thermosensi-
3.1. Hyperthermia tive character of the cancer cells compared to the healthy ones,
due to its high glycolytic activity and consequent lower pH.[140]
3.1.1. Introduction and Traditional Approach However, these conventional technologies resulted to be very
aggressive and suffer from low penetration depths and limited
According to the “World Population Ageing 2015”[137] report by targeting. Actually, targeted therapies remain a challenge in
the United Nations, every country in the world is facing growth cancer treatment and are the focus of nanotechnology in order
both in the number and in the proportion of older persons to improve the therapy efficiency and prognosis of disease.
in their already aged population. The most optimistic demo- An important step toward targeted and higher specific cancer
graphic trend estimations agree that population ageing—the therapy has been given with MNP-mediated hyperthermia
increasing percentage of older persons in the population—will (MH) which is already being used, for example, on the treat-
become one of the most relevant social transformations of our ment of mice’s tumors (Figure 7).[141]
century, with implications for nearly all sectors of society. As an MH takes the advantage of the magnetic properties of magnetic
example, between 2015 and 2030, the number of people in the NPs in a fluid to transform the electromagnetic energy (exter-
world aged 60 years or over is projected to grow by 56%. One nally applied as an oscillating magnetic field) into heat.[142–145]
of the most sensitive sectors that will mostly suffer this popula- MH progressively consolidated itself as a promising alterna-
tion ageing will be undoubtedly the healthcare. Linked to this, tive to fight cancer and currently constitutes one of the most
cancer is currently one of the leading causes of morbidity and intense research areas of nanotechnology in the biomedical

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (9 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 6.  a) Structures of polymers four-armed-PEG and mPEG-COOH. b) Schematic illustration of the preparation procedures of Fe3O4 nanoparticle
cross-linked hydrogel. Representative photos of samples at each stage, from left to right: Fe3O4 nanoparticle dry powder, stabilized Fe3O4 nanoparticle
in aqueous dispersion before gel assembly, the self-standing solid hydrogel obtained after assembly with four-armed-PEG, and magnetic attraction of
the resulting gel. Reproduced with permission.[136] Copyright 2016, ACS.

field.[146] The localization of MNPs in the tumor area allows for main drawback of the current methodologies used nowadays,
a localized heating that selectively kills the tumor cells while such as chemotherapy and radiotherapy, which show harmful
keeping healthy the normal cells. This approach overcomes the secondary effects derived from a lack of treatment specificity.[147]

Figure 7.  a) Tumor growth curves of ferromagnetic Fe1−xMnxO nanostructures magnetic hyperthermia. b) Mice weight in two groups (test and control).
c) Day 50 photographs of representative mice from test and control groups. Reproduced with permission.[141]

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (10 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Different physical mechanisms are involved in the absorp- where C is the specific heat capacity of the medium (assumed
tion and transformation of the applied oscillating magnetic equal to that of water, Cwater = 4185 J L−1 K−1), m is the con-
energy into heat by the MNPs, which depend on the chemical centration of the transition metal of interest (g L−1) of the
nature of the NPs and their physicochemical and colloidal prop- magnetic material in solution, and ΔT/Δt is the slope of the ini-
erties, as well as on experimental parameters such as the fre- tial linear section of the temperature versus time curve. Consid-
quency and intensity of the magnetic field.[148–153] For example, erable efforts have been devoted to maximize the SAR of a wide
in metallic NPs, the main mechanism by which the NPs dis- range of NP compositions, since this would imply a lower dose
sipate the applied electromagnetic energy is by the generation of NPs to be administered to the patient with the subsequent
of eddy currents as a consequence of the law of induction, lower risk experiencing harmful side effects directly derived
which largely depends on the electric resistivity of the material. from the treatment in itself and from toxicity issues related to
In the case of MNPs (essentially metal transition oxides), the the administered heat mediators. Although different nanoma-
main mechanism of heat generation is dominated by hysteresis terial compositions have been explored which show relatively
losses, which are strongly dependent on the particle size. Above high SAR values, iron oxide NPs are so far the most accepted
a certain diameter, the creation of magnetic multidomains is nanostructures as heat mediators in magnetic hyperthermia at
energetically favorable in order to reduce the magnetocrystal- both preclinical and clinical practice levels. They are biocompat-
line energy. These NPs are characterized by ferromagnetic ible and nontoxic, and show relatively high saturation magneti-
behaviors with open hysteresis loops that dominate the energy zation and zero residual magnetization in the absence of a mag-
dissipation process by reversal magnetization. However, as the netic field, and are easy to be surface-functionalized to improve
particle size decreases, the multidomain magnetic structure is colloidal stability and therapeutic purposes.[157,158] However,
no longer energetically supported, and the NPs become single SAR values in iron oxides are far from optimal, and typically
domain, characterized by super-paramagnetic magnetic behav- vary from a few units of W g−1 up to several hundreds,[159–161]
iors and closed hysteresis loops. Super-paramagnetic NPs are which remains a major issue for their clinical use. Efforts have
the most demanded NPs in researched biomedical applications, been devoted in the last years to optimize the heating efficiency
since magnetic interactions between NPs are minimized once of iron oxide NPs. Intrinsic structural and magnetic properties
the magnetic field has been removed. In this particular case of have been deeply studied and correlated to the hyperthermia
magnetic oxides with particle sizes below the single-to-multi- performance of iron oxides.[162–164] On the one hand, key struc-
domain limit, there are two mechanisms involved by which a tural parameters such as high crystallinity and narrow particle
fluid of NPs efficiently transforms the electromagnetic energy size distribution revealed to favor the energy losses.[165–167] On
into heat: (i) Neél relaxation, based on the alignment of the the other hand, magnetic parameters such as saturation mag-
magnetic moment of the particle with the applied magnetic netization and magnetic anisotropy were found to largely affect
field direction and their reorientation to the equilibrium posi- the heating properties of the nanomaterials under exposure of
tion under no magnetic field; (ii) Brownian relaxation, which is an AMF.[142,153,168,169] Moreover, solvent properties such as vis-
based on the friction between the nanoparticle surface and the cosity and specific heat capacity, as well as the colloidal prop-
solvent as a consequence of the torque forces generated in the erties of the NP dispersions, i.e., nanoparticle concentration,
process of reversible orientation of the magnetic moment under resulted to be key in the optimization of the heating proper-
the magnetic field followed by the reorientation of the equilib- ties of the fluids.[170–172] The effect of magnetic dipolar inter-
rium position once the magnetic field has been removed, which actions has also been the focus of intense research in the last
provokes the subsequent rotation of the nanoparticle in the years.[161,173] With the aim of better understanding the effect
fluid.[148,154,155] Both Brownian and Neél fluctuations are catego- of the magnetic NPs’ interaction in the heating properties of
rized by different magnetic relaxation times, which will regu- the nanomaterials, Grillo et al. developed a magnetic nanocap-
late the heating output of the NPs for specific frequency and sule with which evidenced the key role of the magnetic dipolar
intensity of the alternating magnetic field applied. They take interactions in the optimization of the SAR.[174] Additional theo-
place in parallel, and the heating is driven by the one having retical and experimental studies have been performed which
the shorter characteristic time.[156] SAR or specific loss power showed nanoparticle aggregation and arrangement phenomena
represents normalized parameters that allow us to measure and to govern the heating properties of magnetic fluids.[168,175–179]
quantify the efficiency of a given material to transform the elec- However, general conclusions about the general trend of the
tromagnetic energy into heat at specific values of frequency and nanoparticle aggregation remain unclear. The variation of one
amplitude of the alternating magnetic field. or several of these factors is responsible for the wide ranged
The well-known Rosensweig equation[156] predicts the heat SAR values observed among different laboratories, in addition
loss of a magnetic hyperthermia test executed on a ferrofluid to the different results shown when the SAR is calculated by
will be dependent on (a) the magnetic field frequency and using different approaches.[180,181] Also in the presence of dif-
strength, and (b) the physical properties of the ferrofluid such ferent surfactant coatings, the nanoparticles were found to
as magnetic and hydrodynamic radius of the NPs and solvent largely affect the structural, magnetic, and hyperthermia prop-
viscosity, particle size distribution, and the magnetic anisotropy erties of iron oxide NPs.[182] Chitosan, dextran, polyacrylic acid,
energy constant of the magnetic core. Experimentally, the SAR PEG, PVP, folic acid, and cysteine, among many others, were
(W g−1) is calculated according to the following equation some of the functional coating agents used for hyperthermia
applications of iron oxide NPs.[157,170,183–188] Recently, synthesis
C ∆T procedures have been studied in depth,[189] and efficient NP
SAR = (1)
m ∆t designs and coatings have been developed with the aim of

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (11 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

increasing the SAR of iron oxides as much as possible. Iron a given material to be intrinsically used for magnetic hyper-
oxide NPs with high shape and magnetic anisotropies, such thermia applications. In particular, the last significant advances
as nanowires (NWs),[190] nanocubes,[164] nanoflowers,[191] nano- to increase the SAR of iron oxide NPs were mainly based on
flakes,[192] and nano-octopods,[152,169] as well as multicore[161,176] the fabrication of nanostructures with high magnetic anisot-
and core–shell nanostructures joining different material com- ropy (e.g., magnetite cubes,[164,204] rods,[205] and octopods[169])
positions and magnetic behaviors,[193,194] have showed the and core–shell NPs that exploit the exchange and surface ani-
highest SAR values reported so far. In spite of a lack of con- sotropy appearing when soft and hard magnetic materials are
sensus regarding the SAR trend as a function of some experi- join together in one single nanostructure.[193,206–208] However,
mental parameters, i.e., nanoparticle concentration in the the challenge remains since high heating power in suspension
fluid and AMF magnitude, the maximum SAR values reached solutions may not transform into efficient heating in the cel-
already the 1–2 kW g−1. In an attempt to search new materials lular environment,[209] where a drastic decrease in the SAR of
with higher hyperthermia efficiency under an AMF field, other colloidal nanoparticle suspensions was observed when they got
nanomaterial compositions have been explored, Fe NPs, Co-, into endosomes as a result of the cell internalization mecha-
Mn-, Ni- and Zn-ferrites (pure or doped), bimetallic FePt, FeNi, nisms. Soukup et al.[210] demonstrated by in situ measure-
FeCo, iron carbides such as FexCy, Fe@FexC, and combinations ments of magnetization relaxation that cellular internalization
of Fe with Au and Ag have been some of the materials investi- can disable Brownian relaxation in live cells, which has impor-
gated for hyperthermia performance.[142,148,153,162,187,194–200] Even tant repercussions for designing suitable NPs for intracellular
magnetic nanomaterials with particle sizes above the single-to- hyperthermia applications.
multidomain transition exhibiting ferromagnetic behavior have In the case of targeting, many small organic molecules and
been investigated.[188,201,202] However, too low SAR values and biomolecules such as peptides and proteins have been used as
serious toxicity concerns pointed out iron oxides as the mag- targeting moieties of functionalized iron oxide NPs for selective
netic NPs of excellence for hyperthermia applications. accumulation of the nanoheaters in the tumor,[184–186,211] but
The main application of magnetic fluid hyperthermia this remains a challenge for translational research, especially,
appears in the biomedical field in anticancer therapy.[146,203] when the intended administration route of the heat probes is
The localized character of the temperature increase generated systemic. On the other hand, the accurate control of the tem-
by the heat mediators was quickly conceived as an opportu- perature in the treated area is considered a major drawback
nity to selectively kill tumor cells and diminish the harmful of magnetic fluid hyperthermia. Numerous efforts have been
side effects derived from more conventional and aggressive devoted to this issue in recent years, and important advances
cancer treatments such as chemo- and radiotherapy. Many have been already achieved. In an initial stage, the research
studies have been designed to explore this methodology both was focused on demonstrating the higher temperature around
in vitro and in vivo that resulted in promising results; how- the NPs when an AMF was applied. Bañobre-López et al.[212]
ever, the limited efficacy observed in clinical trials seems to and Rodrigues et al.[212] were pioneer in demonstrating the
determine its slow translation to the clinics. Magnetic hyper- local temperature increase around magnetic heat mediators
thermia is already included in the portfolio of treatments of and their superior bactericide effect on food spoilage micro-
the national healthcare systems of some European countries organisms in both planktonic and biofilm living states when
such as Germany and the Netherlands, and it appears as one subjected to magnetic fluid hyperthermia, compared to the
of the recommended treatments in the USA National Compre- situation in which the heat is traditionally and macroscopically
hensive Cancer Network guidelines for several types of cancer applied. This superior effect of magnetic hyperthermia com-
diseases. NanoTherm treatment from MagForce is one of the pared to other conventional methods of macroscopic heating
best known examples where magnetic hyperthermia is being has been extended later on to eukaryotic and tumor cells.[213,214]
applied at clinical level for the treatment of glioblastoma with The localized thermal effect of magnetic hyperthermia was
successful results. Clinical trials have been extended to other also correlated with the gene expression of enhanced green
type of tumors, such as pancreatic, prostate, or esophageal. fluorescent protein (EGFP) in human lung adenocarcinoma
Magnetic hyperthermia can be administered as a stand-alone cells, expression that was found to be linked to the intracel-
therapy or in combination with radiotherapy and chemo- lular temperature increase[215] due to the cell stress induced
therapy. In this way, magnetic fluid hyperthermia showed the to the cell even when this temperature increase was not mac-
most successful results, but it is worth mentioning that the roscopically observed. Parallel efforts were made with the
most successful results point to an increase of the life expec- aim of accurately measuring the temperature value locally
tancy up to about 8 months after diagnosis of primary tumor reached around a magnetic nanoheater. Thus, Riedinger
in patients suffering glioblastoma.[96] This fact evidences the et al.[216] designed a thermoresponsive linker that allowed us
promising but, so far limited, therapeutic efficacy of magnetic to infer the temperature variation a few nanometers away from
hyperthermia to fight against cancer. Current limitations of the nanoparticle surface. And more recently, the development
magnetic hyperthermia are associated with low SAR of heat of different designs incorporating magnetic and upconver-
mediators, lack of tumor targeting, and impossibility to accu- sion NPs was exploited to provide a temperature measurement
rately control the temperature in the treated area, which have through the temperature-dependent fluorescent properties,
hindered the large implementation of the technique into the but the lack of validation of the nanoprobes in vitro, the dif-
clinics. Fine-tuning of NPs’ size, shape, and interparticle mag- ficulty of the data interpretation due to the background fluores-
netic interaction, which determine the effective magnetic field cent emission of living cells, together with the low penetration
of anisotropy, are crucial to optimize the heating efficiency of depth applicability and the toxicity issues associated with the

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (12 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

lanthanide elements, still make the accurate


measurement of the local temperature on
treated tumor site a major challenge in this
field.[217,218]
As a result of all the efforts devoted to
the optimization of MNPs to overcome the
aforementioned current limitations of MH,
numerous super-paramagnetic nanostruc-
tured systems have been established to
improve the effectiveness of heat therapy as a
treatment of cancer, mainly focused on intra-
cellular actuation. Doping of iron oxides with
transition metals[198] and coating agents with
different ligands such as antibodies, peptides,
small molecules, and carbohydrates[219,220]
have been used to increase the cell inter-
nalization of MNPs and the efficiency of
intracellular hyperthermia.[204] An extensive
literature reports on the use of super-para-
magnetic NPs in MH and the heating mech-
anisms involved, as well as on challenges and
Figure 8.  Scheme of application and mechanism of remote controlled drug delivery trough
promises of the technology.[16,221]
nanocomposite sol–gel materials. a) Injection of the gel. b) Application of the magnetic field
Beyond the local thermal therapy in which that causes heat and releases the trapped drug. c) Removal of the applied magnetic field that
a generated temperature increase can pro- leads to the temperature decrease (until the initial body temperature), entrapping the residual
duce on tumor site by the thermal activa- drug. d) Application of a magnetic field heats to upper transition, releasing residual drug.
tion of MNPs under a remote AMF (due to Reproduced with permission.[223]
more temperature sensitivity of cancer cells
compared to normal cells), the energy produced by the MNPs magnetic hyperthermia has provided innovative solutions is
in the presence of an AMF can be utilized in other therapeutic the development of biocompatible magnetic scaffolds for tissue
approaches, such as drug delivery, nanoscale energy delivery, regeneration. The incorporation of MNPs into natural and syn-
and combinational treatments. thetic matrices mimicking body tissues provided the final struc-
tures with heating properties when exposed to AMFs, which
were used either to deliver loaded drugs and grow factors or to
3.1.2. Hyperthermia through Drug Delivery induce magnetic gradients and mechanical effects that resulted
in an enhancement of the angiogenesis and cell proliferation in
Therapeutic capacity of magnetic hyperthermia has been the implanted regions. Magnetic hydroxyapatite (HA), gelatine,
greatly enhanced when exploited as an external thermal stimuli polycaprolactone, and silk are some of the natural materials uti-
generator in drug delivery. Here, we will highlight those macro- lized as hosts of MNPs for hyperthermia-induced bone tissue
and nanoscale systems containing MNPs in which a remotely engineering.[227–229]
applied AMF induces a thermal stimulus that is used to control On the other hand, a field of intense activity of nanotech-
the loading and delivery of therapeutic compounds previously nology, nowadays, in the fight against cancer is the develop-
incorporated into them. At the macroscale, a few recent exam- ment of organic–inorganic hybrid nanocomposites for diag-
ples of novel materials exist in which a remote AMF triggers nosis and therapy applications. Regarding therapy, magnetic
thermal transitions and morphological/structural changes that micro/nanohybrid self-assemblies have been found promising
result in the modulation of drug release. For example, ther- in hyperthermia-induced drug delivery, as they combine the
moresponsive magnetic hydrogels were developed by Campbell encapsulating properties and modified drug release together
et al.[222] who showed a fourfold enhancement of drug release. with the physicochemical properties of the inorganic coun-
Other magnetic thermoresponsive nanocomposites based terpart. Thus, a wide variety of magnetic organic–inorganic
on the combination of certain polymers, nanogels, or lipids, drug delivery hybrids have been developed which showed
together with iron oxide NPs, were developed in which thera- a modified cargo release under thermal stimuli induced by
peutic compounds (or drug models), such as lysozyme, sodium an AMF.[148,217,230–235] For example, Kim et al.[232] developed
fluorescein, ibuprofen (IBU), and nifedepin, were released by stimuli-responsive magnetic nanomicelles as multifunctional
the triggered thermal effect of a remote AMF (Figure 8).[223–226] heat and cargo delivery vehicles. Béalle et al.[236] also devel-
In most of the above examples, the burst of the drug release oped ultramagnetic liposomes with hyperthermia capability for
responds to structural phase transitions caused in the materials hyperthermia-induced drug delivery, and Katagiri et al.[237] opti-
as a consequence of the local heating induced by exposure to an mized hybrid liposomes constituted of thermosensitive block
AMF. In many cases, these phase transitions are even revers- copolymers, iron oxide MNPs, and phospholipids as ON–OFF
ible, and the thermoresponsive materials undergo several cycles switchable drug release activated by an AMF. In addition to
of responsive drug release. Other field of great activity in which magnetoliposomes, numerous magnetic hybrid architectures

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (13 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

have been synthesized which are able to transform an applied thermoreversible Diels–Alder and 1,3-dipolar cycloaddition. PEG
electromagnetic energy into a triggered thermal stimuli for con- was chosen as a hydrophilic polymer to enhance colloidal
trolled drug delivery. Polymeric micelles (PMs), polymersomes, dispersability in water and maleimide-bearing rhodamine
polymeric nanocapsules, solid lipid NPs, among others, are B was employed as a model system for a drug. The authors
some of the hybrid nanoformulations which are able to simul- showed that rhodamine B was released from the NPs after
taneously incorporate a significant amount of MNPs and bio- the application of AFM due to retro-Diels–Alder reaction. In
active compounds, enabling an AMF-triggered cargo delivery. the same line, Saint-Cricq et al.[248] encapsulated a drug in a
Thus, Hu et al.[238] utilized a double emulsion technique to mesoporous shell surrounding an SPIO nanoparticle. The pro-
develop composite nanocapsules based on a block copolymer posed drug delivery mechanism is based on the break of an
of poly(allyl alcohol) and polystyrene that were able to integrate azo-containing PEG polymer, that after degradation expose the
both hydrophobic and hydrophilic components along with porous, and consequently, releases the drug. The temperature
oleic-acid-coated MNPs, whereas Qu et al.[239] encapsulated both of the solution did not increase significantly; therefore, this
camptothecin and hydrophobic MNPs into polymeric assem- work highlights the local generation of very high temperatures
blies of an amphiphilic polymer that was thermosensitive. In in the vicinity of SPIOs due to MH. All the aforementioned
both systems, the cargo release significantly increased when examples of MNP-mediated drug delivery, both with covalent
exposed to an AMF. Also, Almeida et al.[240] incorporated iron and noncovalent drug conjugation approaches (see the review
oxide NPs and onco-A in triglyceride-based solid lipid NPs that from Ulbrich et al.[17] for further details), showed high promise
showed hyperthermia properties under an AMF. More recently, for cancer therapy. However, more studies are needed about
Carregal-Romero et al.[234] proposed a system of microcapsules their interaction with other biomolecules, cells, and tissues.
prepared by layer-by-layer methodology in which the core was As was already mentioned, MH can be applied as a stand-
loaded with dextran cascade blue, as a drug model, and mag- alone methodology or in combination with radiotherapy or
netic nanocubes were placed at the shell of the structure. The chemotherapy. Nowadays, even when combined, all these meth-
authors showed that after irradiation with AMF, the outer shell odologies are provided separately. In the last years, important
was partially disrupted and the cargo was partially released. efforts have been made to combine MH and chemotherapy by
Other recent example was reported by Griffete et al.[241] In this using a single nanostructured nanoplatform, which will be able
study, magnetic NPs were used as individual cores with mole­ to provide thermal cell killing and localized delivery of a chem-
cularly imprinted polymers and doxo loaded by hydrogen bonds otherapeutic drug. Kumar and Mohammad[148] reviewed the
in the binding sites. After AFM treatment, a significant amount magnetic nanomaterials used for hyperthermia-based therapy
of the doxo was released, and in vitro studies with cancer cells and controlled drug delivery. Since then, important results have
confirmed a reduction in the viability after AFM treatment. been achieved related to the design of both magnetic cores
and nanocomposites aiming at achieving a joint thermo- and
chemo-therapy.[239,249–253] For example, Qu et al.[239] developed a
3.1.3. Alternative Approaches magnetoresponsive drug-loaded nanocarrier for efficient thermo-
chemotherapy achieved by combining highly efficient magnetic
In parallel to the use of magnetic nanocomposites for AMF- hyperthermia with magnetothermally facilitated drug release,
mediated drug delivery applications, alternative methods have which resulted in a synergy therapeutic effect. The operational
been explored based on the direct conjugation of a drug model magnetothermal response leads to a high improvement of tumor
to the surface of MNPs.[242] Thus, a decade ago Derfus et al.[243] cell killing by an operating procedure involving heat shock pro-
conjugated a DNA strand to a dextran-coated iron oxide MNPs tein overexpression and MH-facilitated cellular uptake. Last
and bound to this surface a fluorophore drug model. The trends regarding the use of MH in drug delivery applications
thermal activation of the MNPs under an AMF was enough involve the conjugation of imaging and hyperthermia-derived
to melt the DNA strands and release the cargo model. More therapy in the same material architecture. Thus, Sanson et al.[250]
recently, Nair et al.[244] validated the remote controlled drug developed doxorubicin (DOX)-loaded magnetic polymersomes
release of an anti-human immunodeficiency virus (anti-HIV) as theranostic nanocarriers that combine contrast enhancement
component that was tethered to a magnetoelectric core–shell MRI capability and dual thermo- and chemotherapy, whereas
structure, constituted of a cobalt ferrite core and a BaTiO3 Hayashi et al.[254] also developed theranostic NPs potentially suit-
shell, whereas very recently Meikle et al.[245] surface-conjugated able for MRI-guided thermo-chemotherapy by controlling the
super-paramagnetic NPs with thermoresponsive poly(epsilon– particle clustering. On the other hand, Thorat et al.[252] devel-
lysine) dendrons tethered with carboxybetaine for the mild oped self-regulating La0.7Sr0.3MnO3 nanoheaters functionalized
hyperthermia-controlled delivery of vascular endothelial growth with an oleic acid–PEG PM structure, and loaded it with DOX,
factor (VEGF). Other recent designs involve the organic func- which showed cancer cell killing up to 90% over apoptosis and
tionalization of iron oxide NPs with drug models through late necrosis pathway in MCF7 cells. In this line, Huang et al.[233]
linkers with thermosensitive bonds (i.e., Diels–Alder reaction— have recently provided an overview on the last targeted and
maleimide and furan moieties—intramolecular lactamization, image-guided approaches of MNPs in drug delivery, empha-
and azo release[246]), which cleave the active ones once an AMF sizing the high importance of biomarker pointing for a more
is applied.[116,117] As an example, N′Guyen et al.[247] reported personalized therapy and the single contrast-enhancing proper-
super-paramagnetic iron oxides (SPIOs) stabilized with phos- ties of theranostic MNPs in cancer therapy.
phonic ligands bearing alkyne and furan moieties. The nano- Beyond the uses of MH in drug delivery and its enhanced
conjugate offered the possibilities of functionalization through therapeutic effect against cancer when it is in combination with

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (14 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

chemotherapy, MH has also been utilized in combination with and personalized therapy, these MNPs-mediated thermo-chemo-
photodynamic therapy. A. Espinosa et al.[255] designed a smart therapy approaches have been also exploited as theranostic nano-
nanoplatform based on dually loaded hybrid liposomes with structures, which additionally show image (i.e. MRI)-guided drug
photosensitizers (in the bilayer) and MNPs (in the aqueous delivery capabilities and allow for monitoring of patient response
core). After excitation with a laser, the generation of singlet to treatment. Despite the significant advances achieved, impor-
oxygen in combination with the heat induced by MH resulted tant drawbacks related to the heating properties of MNPs, their
in complete cancer cell death in vitro and in solid tumor abla- selective accumulation at tumor site, or cells and the monitoring
tion in an in vivo rodent model. In the same way, Kolosnjaj-tabi of the on-site temperature in vivo still remain. However, the
et al.[256] developed multicore iron oxide NPs coated with a gold increasing number of in vivo assays and the promising results
branched shell with optimized magnetic and plasmonic prop- obtained augur an increasing translational research with a great
erties, which allowed for a bimodal thermotherapy (magnetic impact on the treatment of several diseases.
hyperthermia plus photothermal therapy) validated in vivo.
In addition to combinational treatment approaches involving
MH and chemotherapy, radiotherapy, and photodynamic therapy, 3.2. Drug Release
magnetic hyperthermia can also be applied in combination with
gene therapy.[242] Besides the development of responsive carriers Traditional application of medicines is impaired by limited
for the delivery of therapeutic molecules mediated by an AFM, effectiveness, poor distribution in the body, and low selec-
magnetic hyperthermia has been found to significantly enhance tivity, the therapeutic delivery of drugs on the target placed
the efficiency of nucleic acid delivery. Thus, Jiang et al.[235] at the right time and with the right drug quantity being the
developed lipoid-coated iron oxide NPs for improved DNA and main problems in the treatment of numerous health disorders.
small-interfering RNA (SiRNA) delivery induced by an AMF. Those problems and disadvantages can be outdated by the use
Combined thermal therapy via MNP-mediated hyperthermia of controlled drug delivery using MNPs.[263] As a result, the so-
and gene therapy was found to provoke an enhanced toxicity called magnetic “smart” and “stimuli-responsive” systems are
compared to gene treatment alone. As examples, Walther and being developed and optimized to deliver medicines at specific
Stein[257] and Yin et al.[258] conjugated an amphipathic tail- sites or at precise times and rates within the body, according to
anchoring peptide and lethal-7a microRNA (miRNA) to MNPs, a magnetic stimulus that is externally applied.[264] In this way,
respectively. In both cases, combined MH and gene therapy magnetically controlled drug delivery is one of the most prom-
showed additive effects than either treatment alone. Moreover, ising and popular approaches for delivering the medicine to the
MH has also been utilized as an immunotherapeutic treat- specified site, meeting two drug delivery milestones: reducing
ment[259] and as an ROS generator[260,261] with promising results. the drugs’ side effects by accurately targeting to the desired
Apart from biomedical applications, MNPs have also been incor- tissue/organ and the controlled release of the medicine to avoid
porated into different polymeric matrices that show interesting traditional underdoing cycle/overdosing problems.[263,265]
mechanical behaviors (i.e., shape memory effects) when sub- From all types of magnetic materials, namely ferrimagnetic
jected to local heating under an applied AMF.[262] materials, paramagnetic, super-paramagnetic, antiferromag-
As conclusion, the translation of MNP-mediated hyper- netic, ferromagnetic, and diamagnetic, the super-paramagnetic
thermia to the clinics has been hindered by the current limi- ones are the most interesting ones to use in drug delivery due to
tations found in in vivo experiments, mainly derived from the their ability to develop a magnetization in an external magnetic
lack of specificity and lower heating performance once the field, losing their magnetic moment in the absence of an applied
MNPs get inside the cells. In this sense, increasing efforts have external magnetic field.[263] In addition to that real-time tracking,
been devoted to increase the specific absorption rate of MNPs using established imaging techniques, surfaces of MNPs can be
by studying different morphologies, structures, and designs. coated with various polymers or surfactants that, in turn, are able
The energy transfer at the nanoscale together with the intracel- to interact with or bind pharmaceutically active materials.[266]
lular targeting at tumor site, avoiding nonspecific accumula- The most used MNPs on drug delivery systems are magnetite
tion of MNPs in other tissues, seems to be the most promising (Fe3O4), maghemite (γ-Fe2O3), and cobalt ferrite (CoFe2O4).
approache to improve the efficiency of nanoscale thermal therapy Such MNPs doped with particular drugs have some application
applications. Furthermore, combinational treatments combining problems such as stability and nontoxicity in the physiological,
AMF-mediated hyperthermia with chemotherapy, radiotherapy, not being easy to join high biocompatibility, magnetic satura-
gene therapy, immunotherapy, and photodynamic therapy have tion and interactive functions on the magnetic nanoparticle
shown enhanced toxicity and tumor growth inhibition compared surface. As a solution, the polymer coating not only allows the
to either stand-alone treatment. Besides, magnetic hyperthermia increase of the hydrophobicity of the nanoparticle, but also
has been explored not only as a direct thermal therapy actuator, offers a diversity of surface functional groups to bind drug
but also as a thermal stimuli provider. In this regard, important mole­­cules, increase stability, and inhibit agglomeration.[263]
advances have been achieved in the field of drug delivery. Novel Precise targeting approach depends on two aspects: first,
architectures and nanoparticle designs have been synthesized the magnetic NPs show magnetic attraction and second, the
that allow for a combined thermal (magnetic)- and chemotherapy coupling of high-affinity-modified ligands on the surface of
by using dual-loaded magnetic platforms that able to encapsulate the MNP.[267]
both MNPs and bioactives (e.g., drug, RNA, and DNA). Thus, the After magnetic guidance, drug release can be initialized
thermal energy generated is additionally used to induce the drug not only by external stimuli such as a confined temperature
delivery. In a step forward towards the treatment monitoring increase but also by internal ones such as a change in pH.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (15 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Diverse approaches have been reported to attach a drug to received two treatment sequences; three patients received one
magnetic NPs; the medicine molecule can be loaded in stimuli- sequence; and two patients received three sequences of mag-
responsive hydrogel/polymer frameworks,[268] encapsulated in netically controlled drug targeting that consisted of the infusion
the polymer interspace of magnetic nanostructures,[269] linked of epirubicin in increasing doses (5–100 mg m−2) inserted into
in magnetoliposomes,[270] or trapped to the activated surface of a magnetic fluid. The tumors were treated with magnetic fields
MNPs.[266,271] Furthermore, drugs can be linked to the MNPs by for 1–2 h. In 2 of 14 patients, the same dose of epirubicin, not
covalent connections that are susceptible to degradation as part inserted in the magnetic fluid, was systemically administrated
of the tissue metabolism.[266] 3 weeks after drug targeting, allowing intraindividual com-
Any overview on magnetic drug delivery must start with the parisons. It was reported that the magnetic material could be
merited credit of Paul Ehrlich (1854–1915), a German doctor effectively directed to the tumors in about 50% of the patients
who worked in the fields of hematology, immunology, and anti- without an increase of toxicity. It was concluded that improve-
microbial chemotherapy and proposed that if any molecule ments were necessary to make this procedure more effective
could discriminatorily target a disease-causing entity, then a and independent of patient origin or disease, leading to an
drug for that entity could be accurately delivered sideways with increase in the scientific activity of this research area.
the mediator of selectivity.[272] Such a “magic bullet” could be Furthermore, a pilot study has used slices of tissue samples
created to exterminate the selected organism exclusively. Later, of VX2 squamous carcinoma cells that were measured by mag-
Paul Ehrlich won the 1908 Nobel Prize in Medicine for con- netic relaxation, and the quantification of iron magnetic NPs on
tributions to the immunology research field and since then, the hind limb of New Zeeland white rabbits was determined by
various strategies have been proposed to magnetically con- using the original ferrofluid suspension as a reference. From
trolled drug delivery, the first work concerning the magneti- such distribution of the iron particles within the tissue sample,
cally assisted drug delivery being developed by Widder et al. in the tumor was reconstructed, as highlighted by the higher vas-
1978,[273] reporting a carrier system for the delivery of chemo- cularization of regions with higher magnetite content than the
therapeutic molecules to desired places. It was discovered that remaining tumor tissue. This study additionally revealed that the
the carrier, albumin/Fe3O4 microspheres can be in vivo accu- microscopic histological methods and the integral method mag-
mulated at a desired place with an external magnetic field. Adri- netorelaxometry can compete each other in an efficient way.[280]
amycin delivery was supported by the presence of a significant Trying to comprehensively understand the performance of mag-
content of the medicine at the site of the magnetic carrier local- netic NPs in the blood stream, it was reported[281] that there are
ization. This delivery system allowed us to concentrate in the three types of in vivo behaviors to magnetizable NPs (boundary-
desired site the same Adriamycin dose that was usually accom- layer formation, magnetic dominated, and velocity dominated)
plished by administration of 100-times higher dose of the drug. exclusively identified by three essential nondimensional num-
In the 1980s, several reports were found developing this bers (the Renkin, the mass Péclet, and the magnetic Richardson
strategy to deliver distinct drugs using magnetic microspheres numbers). When skin cells were added to this scenario, a new
and microcapsules, namely mitomycin C (FM-MMC-mc) anti- interface was developed where magnetic NPs can build up,
cancer drug,[274] the anthracycline antibiotic with antineoplastic adding, in this way, a qualitatively new behavior that requires a
activity, doxorubicin hydrochloride,[275] and doxorubicin, a fourth nondimensional number to map out its new effect.
chemotherapy medication used to treat cancer.[276] More recently, smart material-based approaches allow us to
In the following decades, Häfeli et al. developed magnetic increase the efficiency of the traditional drug release magnetic
poly(lactic acid) (PLA) microspheres that include both mag- nanoparticle-based method, ensuring that a specific drug is
netite and the β-emitter 90Y in vivo treatment of tumor cells.[277] delivered with the right dosage, at the right time and in the right
Despite those promising approaches, some tissues in the place[282] using the so-called smart and stimuli-responsive method.
human body are difficult to reach as drugs need to pass tight To achieve such smart drug delivery, smart polymer systems such
tissues; only structures in the nanometer-sized range are able as pH-responsive polymers, temperature-responsive polymers,
to pass.[278] In this way, magnetic NPs were first used in animal and temperature/pH-dual-responsive polymers are being used
models by Lübbe et al.[279] This work is divided into two parts: due to their biocompatible and fast responsive properties, nontox-
first, several contents of a magnetic fluid was tested in rats and icity, biodegradability, effective payload, and release capability.[283]
immunosuppressed nude mice with regard to tolerance and The introduction of an external stimuli-sensitive element to the
efficacy studies; second, the same issues were evaluated after active system that may change or trigger the drug release mecha-
administration of the ferrofluid with chemically bound epiru- nism is of great interest.[284] In this novel methodology, one of the
bicin (4′-epldoxorublcin). It was discovered that the ferrofluid most promising strategies consists in using magnetic NPs that
did not cause major laboratory abnormalities and there was no are stimulated by means of an external magnetic field in poly-
LD50 and no intolerances with the epirubicin-bound ferrofluid. meric composites.[285] In this context, it was reported[264] that the
Both forms of treatment led to complete tumor responses in an fabrication, characterization, and study of the release kinetics of
experimental human kidney as well as in a xenotransplanted IBU on a poly(l-lactic acid) (PLLA) membrane with inclusions of
colon carcinoma model. This pioneer work proved that mag- a zeolite (Faujasite) and magnetostrictive Terfenol-D particles. It
netic nanomaterials are safe agents, which can be used in dif- was observed that the applied magnetic field modifies the release
ferent ways for confined forms of disease treatment in combi- kinetics of the polymeric membrane, leading to an increase of the
nation with high-energy magnetic fields. After these preclinical release rate by more than 30%. Additionally, the increase of the
experiences, a clinical study was carried out with 14 patients magnetic field intensity from 100 to 200 mT promotes an increase
having advanced solid tumors.[279] Nine of these patients in the percentage of IBU released from 67% to 75%.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (16 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 9.  a) Schematic image of the biphasic ferrogel implant in mouse hindlimb. b,c) Cross-sectional images of biphasic ferrogels. Reproduced with
permission.[286]

A new biphasic ferrogel, containing iron oxide NPs (2–13 wt%), doxorubicin and super-paramagnetic iron oxide NPs in their
capable of large deformations and the triggering of drug composition, are capable of inducing cytotoxicity and increasing
release, was presented.[286] In this way, biphasic ferrogels intracellular drug release. A new approach that opened new
were subcutaneously, surgically, implanted in the hindlimb of horizons in the improvement of smart delivery materials and
6-week-old mice (Figure 9). systems capable of releasing drugs upon demand and opti-
Biphasic ferrogels demonstrated optimized mechanical mizing the treatment control was presented in ref. [288]. It was
properties, enhanced porosity, and increased biocompatibility found that internalized hybrid polymer-based materials filled
as a result of their low iron oxide content. With their capability with γ-Fe2O3 super-paramagnetic iron oxide NPs and doxoru-
to on-demand deliver drugs and cells, it is predictable that bicin, when exposed to a high-frequency (750 kHz) magnetic
such magnetic gels will gain wide values in the fields of drug field (14 mT), were capable of increasing cytotoxicity to 18%
delivery, regenerative medicine, and tissue engineering. and activate intracellular drug release. Nevertheless, in clinical
By employing magnetic nanocomposites of temperature- tests the field intensity was decreased to 3–14 kA m−1 and the
responsive hydrogels composed of N-isopropylacrylamide and field frequency was decreased to 100 kHz with the objective
Fe3O4 NPs, the use of magnetic fields to trigger drug release on to contain patient discomfort, which depends on the treated
demand has been discussed.[287] The application of the magnetic region of the body.[289,290]
field results in the heating of the nanocomposites leading to a A multifunctional magnetic mesoporous Fe3O4/silica nano-
faster squeezing and collapse of large quantities of embedded particle with both a successive dual-targeting (both active and
drug. When submitted to an AC magnetic field with a high magnetic, Figure 10) ability and a controllably switched drug
frequency, internalized hybrid polymer-based materials, with release options was presented in ref. [291].

Figure 10. Schematic representation of doxorubicin (DOX)-loaded cross-linked magnetic mesoporous silica NPs MMSN-S2- poly(ethyleneimine)
(PEI)-PEG- phenylboronic acid (PBA) reservoirs being consecutively targeted to the tumor site through the magnetic targeting enhanced permeability
and retention effect and NanoEL, and the SA-positive HepG2 cells by the active-targeting, succeeding a fast drug release by GSH stimulus in the
cytoplasm. Reproduced with permission.[291]

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (17 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

In vivo essays revealed that this magnetic nanoparticle can be and biological outputs, being able to produce structures more
initially concentrated close to the tumor site under the applica- similar in size to the natural extracellular matrix.[294,295]
tion of a magnetic field. After that, it can enter into tumor cells MNP-based approaches and scaffold production methods
by (poly(ethyleneglycol))-3-aminophenylboronic acid-mediated have been developed with the main objective to control cell
endocytosis. In the end of the process, the chemically cross- function. Cell functions are mainly controlled by bioactive
linked network on the surface of this magnetic structure is dis- and growth factors and mechanic transduction pathways (for
ordered as a response to the high concentration of glutathione example, by mechanical stress) that can influence and con-
in cytoplasm, opening the mesopores and quickly releasing the trol the cell behavior by the conversion of mechanical stress
drugs, consequently, prompting apoptosis in cell. into intracellular biochemical cues.[296] The first use of MNPs
Apart from the above-mentioned studies, there are some for intracellular labeling was reported in 1993; however, only
limitations in the drug release based on MNPs, such as poor in 2000, they were applied to stem and progenitor cells.[297] It
tissue penetration and cell damage, which should be addressed should be noted that for the mechanic transduction studies, the
in the future. Additionally, as a suggestion for future investi- most commonly used magnetic particles are iron oxide parti-
gations, a new type of smart MNPs that are able to consecu- cles because they are more easy to synthesize by coprecipita-
tively release drugs and genes at precise time intervals and at tion from iron salts.[298] This approach allows the remote con-
pre-established sites is needed. One way to meet this challenge trol of the mechanic transduction pathway with spatial and/or
is to take advantage of physiological stimuli that are usual on temporal precision in order to modulate the cell behavior,[296,298]
cancer cells and tumors such as lower pH to activate the drug such as by cell stretching, micropost manipulation,[299,300] local-
release.[292] Additionally, the possibility of associating control- ized stimulus at the single-cell level,[301] or cell receptors’ acti-
lable drug release with tumor targeting allows the design of vation.[298] These techniques that can apply mechanical forces
therapies with desirable pharmacodynamics and pharmacoki- directly to the cell or to the mechanoresponsive receptors of
netics behaviors that will trigger the use of MNPs in clinical individual cells are important for a wide variety of tissue engi-
personalized medicine.[17] neering applications, because they do not require the scaffold
deformation.[302] For example, micropost arrays composed of
cobalt nanowires and an array of elastomeric posts were used
3.3. Tissue Engineering as sensors for cellular traction forces to study the cell response
to the different applied forces. It was found that, when the
Over the last decades, the use of MNPs has been increasingly force was applied, these microposts enhanced the focal adhe-
utilized in regenerative medicine and tissue engineering due sion recruitment and size at the local of the stimuli and not at
to the novel approaches that they offer and to their promising the nearby nonmagnetic posts.[299] In order to apply a confined
potential. The magnetic particles have directly been incorpo- stimulation at the single-cell level, patterned MNP-dosed cells
rated into the cells or used to interact with cell membranes in microengineered magnetic substrates have been used.[301]
but they have also been combined with biomaterials in order Compared to the receptors’ activation, magnetic fields and
to obtain new functionalities in the structural framework (scaf- MNPs, all have been employed to control specific cell signaling
fold) that has been employed to support cells, namely for the pathways by targeting specific cell surface receptors or sign-
control of the cellular responses (Figure 11). aling proteins inside cells.[302,303]
Several different methods exist to produce scaffolds for In these days, tissue engineering is based on the cell cul-
tissue engineering. Most common are jetting methodologies ture onto 3D scaffolds in order to mimic their primitive
such as aerodynamically assisted jetting/ threading, pressure- function. However, the construction of a scaffold or func-
assisted/driven jetting/spinning, laser-guided writing, inkjet tional organ that can mimic both complexity and precision
printing, electrospray, and electrospinning.[294] Printing, elec- of the organ in vivo is difficult. This can be overcome by the
trospray, and electrospinning are currently undergoing a wide- application of a mechanical stimulation based on a biomag-
spread revival of scientific investigation of chemical, physical, netic approach (Figure 11c). Different strategies based on

Figure 11.  Magnetic actuation strategies for tissue engineering. a) Production of a cell sheet with magnetically prelabeled cells. Cell expansion of
magnetically labeled cells in a low adhesive cell culture Petri dish placed on top of a magnet yields an easily detachable cell sheet. b) Cell seeding
and invasion in a 3D porous scaffold with magnetically prelabeled cells. Placing a magnet underneath the tissue-engineered construct promotes cell
migration into the porous scaffold and homogeneous dispersion. c) Magnetically actuated scaffolds. The application of a magnetic field will potentially
induce a structural deformation in the scaffold and cells. Adapted with permission.[293] Copyright 2015, Elsevier.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (18 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 12.  Representative images of the different effects on the cell behavior—mechanical and mechanoelectric—that can be applied through magne-
toelectric scaffolds. Reproduced with permission.[304] Copyright 2016, Elsevier.

biomagnetic biomaterial were developed, such as films,[304–306] Other magnetic nanocomposite scaffolds made of magnetic
hydrogels,[307,308] fibers,[295,309,310] and 3D scaffolds.[311,312] The particles and different kinds of polymers have been devel-
combination of magnetic particles with polymeric biomaterials oped to study in vitro cell responses and in vivo tissue compat-
has demonstrated large potential for disease treatment and ibility, such as poly(caprolactone),[325,326] silk fibroin/chitosan,[327]
tissue repair, namely bone,[313,314] muscle,[315] nerve[316] and alginate,[328] poly(vinylidene fluoride) (PVDF),[304,329,330] PLLA,[331]
cardiac tissue regeneration[317] with external magnetic stimula- chitosan,[327,332] among others. Iron oxide was incorporated into a
tion. In this way, the magnetically actuated scaffolds allow the 3D-aligned structure of polycaprolactone fibers and starch. Then,
remote delivery of mechanical cell stimulation by the response it was used to study the influence of an applied magnetic field on
of the incorporated magnetic particles. For bone tissue engi- the adipose stem cells’ differentiation, cultured on theses devel-
neering, different kinds of magnetic scaffolds have been devel- oped magnetic scaffolds.[326] The obtained results, on the dif-
oped, and their evaluation, in vitro and in vivo, was performed. ferent magnetic scaffolds, combined with magnetic stimuli have
Magnetic scaffold composed of MNPs and HA were developed shown their large potential for tissue engineering.
and used to study their influence on the cell behavior with and As a novel approach, magnetic particles have been used to
without the application of an external magnetic field. Com- obtain a magnetoelectric scaffolds,[295,333,334] where these scaf-
posite scaffold, composed of PLA nanofibers, hydroxyapatite folds, remotely triggered by an external magnetic field, can
NPs, and paramagnetic γ-Fe2O3 NPs, was prepared, and the provide electrical and mechanical stimulation to the cells.
scaffold responds to the external static magnetic field. These Thus, it has been shown that magnetoelectric Terfenol-D/
are implanted in rabbit defects and the results have shown poly(vinylidene fluoride-co-trifluoroethylene) multiferroic com-
that NPs incorporated in the nanofibers enhanced bone posites can enhance the cell proliferation of MC3T3-E1 pre-
tissue formation and remodeling.[318] Other scaffolds with osteoblast.[304] In this way, magnetoelectric cell stimulation
hydroxyapatite and magnetic particles have been used and represents an innovative and appropriate approach that allows
have demonstrated that they can not only modulate cell adhe- magnetic, mechanical, and electrical stimuli (Figure 12).
sion, proliferation, and differentiation, but also enhance bone However, the use of artificial scaffolds in in vivo environ-
healing, showing promising results to be used for magnetic ments can become a problematic issue; once after their trans-
controlling in orthopedic applications based on tissue engi- plantation, they can limit the cell migration into and from
neering.[9,319–321] Additionally, a high content of MNPs on the the scaffold, and their degradation can induce an inflamma-
magnetic scaffolds leads to a more noteworthy stimulation. tion.[296,335] In this way, another approach includes the construc-
In the particular case of MNP-based bone scaffolds, the high tion of 3D tissues without the use of scaffolds.
scientific interest results from the better capacity of magnetic MCLs were extensively studied for tissue engineering appli-
bone scaffolds to promote bone healing,[9] as proven by Hei- cations once they provide the possibility of reconstructing a 3D
dari et al.,[322] which applied chitosan/nanohydroxyapatite/ tissue without scaffolds, allowing also cell patterning.[336–338]
Cu–Zn alloy nanoparticle composite scaffolds on bone tissue Basically, MLCs present a positive charge at the liposomal sur-
engineering. These scaffolds demonstrated improved anti- face that can interact with the negative charge of the cell sur-
bacterial effects and exhibited high potential for tissue engi- face, being able to be used as carriers introducing magnetite
neering in clinic environments. Another successful approach particles into the target cells, obtaining cells labeled with mag-
on magnetic-based tissue engineering was reported by Peri- netite particles. So, in this way, these cells can be manipulated
yathambi et al.[323] By using goat blood as the starting mate- by the use of a magnetic field (such as magnets or magnetic
rial, the authors prepared new magnetic fibrin and performed bioreactors) for the construction in vitro of 3D tissues, mul-
Saos-2 cells’ viability assay and quantified alkaline phosphatase tilayered cell sheet-like structures and tubular structures, as
levels to evaluate the magnetic fibrin nanoparticles. The MNPs well as to isolate target cells in culture systems for the isola-
exhibited good biocompatibility allowing it to be used in bone tion and recovering of target cells.[293,339] This technique is
tissue engineering. Furthermore, adipose-derived regenerative identified as magnetic-force-based tissue engineering (Mag-
cell sheets were also produced with magnetite Fe3O4 MNPs TE, Figure 11a)[337] and has been used for different kinds of
aiming to treat myocardial infarction. Previously, Ishii et al.[324] TE applications, such as bone,[337,340] muscle,[341–344] skin,[345]
have investigated the therapeutic potential of such MNPs. For vascular,[346–348] cardiac,[349,350] and retinal[351] applications.
that, these NPs were first combined with liposomes to create The definition of Mag-TE was proposed in 2008 by Mironov
magnetic cationic liposomes (MCLs) with a positive surface et al. as “the bioinspired fabrication of more complex tissue
charge and later cultured on adipose-derived regenerative is performed using cells, cellular monolayers, and cell aggre-
cells. The special cell sheets were found to be a promising gates labeled with MNPs.”[348,352] MCLs have been used for the
and worthy regenerative strategy for the treatment of ischemic construction of 3D artificial skeletal muscle by the labeling of
heart disease.[9,323] myoblast cells applying a magnetic force.[341] This approach has

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (19 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

received increasing attention for vascular tissue engineering in order to improve drug targeting to pathological sites.[356]
once it allows the construction of 3D vascular tubes.[346,348] Deep insight into the drug delivery process is a prerequisite for
MCLs have also been employed to enrich and proliferate mes- predicting the therapeutic outcome of the used treatment. In
enchymal stem cells in vitro for clinical application.[353] Bone this way, the studies directed to real-time imaging of therapy
marrow stromal cells were used to construct a cell sheet to be delivery may enable distinguishing between the carrier mate-
transplanted into a bone defect, enhancing the formation of a rials suitable for long circulation and systemic administration
new bone.[354] of drugs, as well as those that more effectively accumulate in
Other strategy is related to cell seeding in a 3D porous targeted tissues, e.g., tumors.[358]
scaffold for cell proliferation and differentiation to be later Not only it is important to predict the accumulation of
implanted in the injury local. However, this approach presents therapy at the target site, but it is also necessary to know if
some unsolved challenges due to the depth and structure of the the used drug is efficiently released. Many literature studies
3D scaffolds.[293] To overcome these problems, labeling cells are reporting only in vitro release kinetics in proof-of-concept
with magnetic particles can be used to be seeded into the scaf- studies for developing novel therapeutics and carrier materials.
fold surface, as with the application of magnetic forces they can However, visualization and analysis of the release patterns
be driven to invade the pores of the scaffold (Figure 11b). This under physiologically relevant in vivo conditions is far more
technique is named Mag-seeding and allows cell distribution complicated. The post-therapy analysis, for example, includes
with great precision and resolution. cell lysis and release of drugs into suitable analysis buffers, the
Finally, despite MNP-based scaffolds with the “core–shell” procedures that can lead to destabilizing carriers and drugs,
structure, which have unique biological, physical, and chemical resulting in ambiguous analysis outcomes. Enabling real-time
properties such as nontoxicity and biocompatibility that make in vivo visualization of the drug release would overcome this
them good candidates for tissue engineering applications, fur- shortcoming. Therefore, theranostic nanoformulations also
ther investigations into their diagnostic and therapeutic appli- aim at simultaneous loading of carriers with drugs and imaging
cations are necessary before considerations.[9] agents for real-time monitoring of drug release. Of particular
usefulness for such applications are contrast agents for MRI,
which emit different signals depending on the interaction
3.4. Theragnosis with surrounding environment, i.e., inside carriers and when
released at the targeted site. Noninvasive visualization of drug
In the last two decades, there is a great interest for conceptu- release could be achieved through generation of an increased
alizing and developing nanotechnology systems that are able MR signal in case of release of contrast agents versus the signal
to diagnose and treat diseases such as cancer. Different from of nonreleased agents. One of the systems that enabled non-
traditional molecular contrast agents and therapeutics, the new invasive visualization of drug release via increased MR signal
concept of cancer nanomedicine aims at integrated designs to is based on temperature-sensitive carriers that, at temperatures
explore multiple functions in one system, including targeting below the transition temperature, emitted signals that are com-
the site of interest, sensitive imaging, and therapy, the so-called parable to nonsensitive carriers, but the signal substantially
all-in-one system.[355] Multifunctional nanotechnology systems increased upon heating to temperatures exceeding the transi-
are considered the next-generation materials in medicine that tion ones.[359,360] Such a system also proved suitable for moni-
would eventually enable premature detection of disease, simul- toring of doxorubicin release.[361]
taneous treatment, and monitoring of the response to the treat- A dual MR-contrast technique for monitoring drug release
ment, and targeted therapy with low toxicity. The theranostics from theranostic nanoformulations was introduced that relies
paradigm was originally seen as a treatment strategy that joins on synthetic polymer-based carriers loaded with 5-fluorouracil
a targeted therapy with a diagnostic test. Nowadays, the thera- (5-FU) as anticancer drug and two contrast agents: gadolinium–
nostic nanomedicine evolved into integrated nanotherapeutic diethylenetriaminepentacetate (Gd-DTPA) and super-paramag-
systems that are able to diagnose, deliver targeted drug therapy, netic iron oxide NPs.[362] Immediately upon intravenous admin-
and monitor the reaction to therapy, all at the same time. istration, a strong T2 contrast was found in the tumor area as a
Lammers et al. highlighted the paramount importance of result of the presence of iron oxide NPs, which remained unal-
imaging function within the nanotheranostic platforms for tered for 2.5 h. In contrast, T1 contrast that corresponds to the
three-level monitoring of therapy: (i) drug delivery, (ii) drug release of Gd-DTPA from these NPs was spotted only after 30
release, and (iii) drug efficacy.[356] The optimization of this min upon administration and lasted less than T2 contrast (less
three-level imaging and combination with the therapeutic func- than 2.15 h). Assuming that 5-FU is released from iron oxide
tion within a single nanoformulation aspire to provide real- NPs with the same kinetics as is Gd-DTPA, this system thus
time feedback on drug kinetics, drug localization on target site, allowed for simultaneous screening of both therapy delivery
and off-target accumulation (i.e., in healthy organs). Possibility and drug release. Despite all the advances in monitoring drug
of monitoring real-time drug release and distribution opens release, in vivo screening is by far the most difficult task for
avenues for predicting the effectiveness of the therapy and opti- drug imaging, as it requires developing systems with increased
mizing treatment outcomes—steps toward performing a real complexity.
personalized medicine that takes into account individual vari- Real-time imaging of drug efficacy requires somewhat less
ability of response to treatments.[357] complexity in the theranostics development. The theranostic
Imaging of drug delivery refers to allowing easy, noninvasive, nanoformulations featuring this property are based on pre-
and real-time pharmacokinetic and biodistributional analyses dicting and monitoring therapeutic responses for personalized

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (20 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

therapy concepts. Also, imaging of drug efficacy is tightly the first (bio)polymer extensively investigated in experimental
related to drug delivery, since both require precise informa- animal models and in humans as an MRI contrast combined
tion with regard to target site accumulation. Nevertheless, the with magnetic NPs. While these NPs provide high contrast
rational predictions of therapeutic efficacy are only possible in imaging, the heating efficiency of magnetic hyperthermia
clinical trials. In a simplified concept, a trial should focus on is often low to effectively treat most advanced tumors using
imaging a theranostic nanoformulation for prescreening the toxicity-free concentrations. For providing only one function-
patients in order to identify who have tumors permeable to ality, some of these formulations have been discontinued or
the used therapy and who have not, thereby predicting which their production is abandoned by the manufacturers. Instead,
patients are likely to respond to the therapy. Good tumor accu- new trend is the synthesis of hybrid inorganic magnetic nano-
mulation of a nanotheranostic would typically means a good constructs or combining magnetic NPs with traditional or
response to a therapy. A continuous subjection of patients to advanced therapeutics to achieve synergistic antitumor effects
imaging during receiving the therapy would allow visualiza- via a variety of mechanisms.
tion of the therapy efficacy in real-time (e.g., tumor shrinking), An increase of MR contrast effects can be even achieved by
which is important information for deciding if the therapy the substitution of one of the Fe ions in iron oxide NPs with
should be continued or not, and whether or not to adjust drug magnetic or nonmagnetic atoms replace the Fe ions.[366] For
doses. This is the main goal of theranostic nanomedicines for example, the substitution of Fe ion with Zn2+ synthesizes Zn-
tailor-made therapy on individual patients.[356] doped ferrite NPs of ≈15 nm with increased magnetization.[367]
Recently, magnetic NPs (e.g. iron oxide) have replaced tradi- An optimum Zn-to-Fe doping ratio to enhance the contrast
tional gadolinium contrast agents that are widely available as effect was found at Zn2+ of 0.4, which is six times higher than
commercial formulations, but commonly featuring poor body that achieved by commercially available iron oxide formulation
clearance. Iron oxide NPs’ expansion as contrast agents is owed Feridex. In another approach, the T1 and T2 contrast effects
to their high magnetic moment and very high resolution in were merged in a dual-mode contrast agent for providing
noninvasive monitoring of their distribution and accumulation more accurate MRI and were better distinguished between
in soft tissues. Furthermore, magnetic NPs, if used in suitable the normal and diseased areas.[58,368] Core–shell-type inorganic
concentrations, offer additional hyperthermia effect and also NPs were fabricated with the fine layer of Gd-based T1 contrast
the possibility to be used as carriers themselves for entrapment agent in the shell for direct contrast effects and the super-para-
of various drugs.[363,364] They can also be externally controlled magnetic T2 contrast material (MnFe2O4) of 15 nm in the core,
(via magnetic field) for targeted delivery of these NPs, which separated by SiO2. Using this hybrid structure, ambiguity of the
may, in addition, reduce toxicity risks (used in small concen- single mode contrast agent from different artifacts is effectively
trations) and offer enhanced treatment specificity.[365] Thus, eliminated, while self-confirmation of images is allowed and
multifunctionality makes the magnetic NPs extremely attractive leads to larger diagnostic accuracy.
for developing theranostic nanoformulations for personalized Doxorubicin, as an FDA-approved agent, is by far the most
nanomedicine. frequent anticancer drug loaded onto magnetic NPs, regardless
Iron oxide NPs are unique in featuring all the above-men- of the study positioning: from proof-of-concept to clinical trials.
tioned functionalities and are perhaps the most promising A wide variety of cancers and tumors, including breast,[369]
nanoplatforms for clinical applications, especially regarding lung,[370] and colon,[371] were investigated after subjection to
treatment of cancers (Figure 13). As a consequence, several doxorubicin-loaded magnetic NPs, synthesized and function-
formulations based on iron oxide NPs received FDA/ European alized by many different methods. Doxorubicin is followed by
approval for cancer imaging or are currently and the clinical other anticancer drugs such as paclitaxel,[372] methotrexate,[373]
trial stages for therapy approval (Table 1). The approved formu- and epirubicin.[374] Despite achieving the great advances, espe-
lations are dominated by dextran-doped NPs, since dextran was cially when combined with targeted functions, and the fact

Figure 13.  Schematics of multifunctionality within theranostic magnetic NPs for tumor targeting, imaging (diagnosis) and therapy.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (21 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Table 1.  Some of the magnetic NPs clinically approved or in the phase of clinical trials.

Commercial name Formulation Application Status


Feridex Iron oxide coated with dextran MRI FDA approval, discontinued in 2008
Combidex Ultrasmall iron oxide coated MRI Approval in Europe, withdrawal in 2007
with low Mw dextran
Resovist Iron oxide NPs coated with MRI Approval in Europe, production abandoned in
carboxyldextran 2009
Gastromark Silicone-coated iron oxide NPs MRI FDA approval, discontinued in 2012
Feraheme Iron oxide coated with polyglucose Initially, iron-replacement therapy; FDA approval
sorbitol carboxymethylether Currently, MRI
Nanotherm Iron oxide NPs with aminosilane coating Hyperthermia in solid tumors Clinical trial

that there exists the highest possibility that magnetic NPs will of nucleic acid delivery (remote control of delivery), compared
be applied in future with some of these drugs in clinics, the to the commercially available agents. The study came to the
remaining issues for chemotherapeutics are the side effects nearest to the multifunctional concept for the iron oxide nano-
and resistance acquired by different tumors. The current inves- particle gene delivery platform developed due to combining
tigation interests are therefore shifted to the combination of magnetically guided targeting, and possibility of providing gene
magnetic NPs with advanced therapeutic options. therapy with MRI and magnetic hyperthermia.
One of the most promising advanced solutions, as alterna- The first study for in vivo imaging of siRNA delivery and
tives to chemotherapies for cancer management, regards to silencing in tumors considered probes for in vivo transmission
therapeutic DNA to correct a genetic defect or RNA interfer- of siRNA and the synchronized imaging of its accumulation in
ence for gene silencing. For example, the use of siRNA mole­ tumors by NIR optical imaging and high-resolution MRI.[379] The
cules are meant to specifically turn off the abnormal gene probes comprised NIR dye/magnetic NPs/siRNA complexes to
expressions or genetic mutations in cancer cells. DNA and RNA enable the silencing and visualization of the process, providing
molecules alone are fairly ineffective due to their sensitivity in a reliable siRNA delivery and imaging approach that is impor-
complex physiological environments. Magnetic NPs are, thus, tant for the development of cancer theranostic agents. In addi-
increasingly investigated as carriers for gene delivery.[375] The tion, the targeting of two separate tumor models was achieved
first prerequisite for magnetic NPs to be effective carriers is the by modification of the nanoprobes with a membrane trans-
surface modification to allow the attachment of DNA or RNA location peptide for intracellular delivery. This study encour-
molecules. Some of the most popular ways employ cleavable aged other similar ones to use the all-in-one concept based on
linkers (in the case of stimuli-responsive delivery) or utilization magnetic NPs for simultaneous molecular imaging and siRNA
of electrostatic interactions between the therapeutic molecule delivery.[380] The iron oxide nanoplatform was decorated with a
and the surface of the nanoparticle. Alternatively, the active fluorescent dye, siRNA, and a Arg-Gly-Asp (RGD)-peptide tar-
molecules are incorporated into a degradable, in the tumor geting moiety to enable macroscopic–microscopic dual imaging
environment, outer shell, which releases the incorporated mole­­ of target cells and concurrent therapy. More recently, “stealth”
cule as it is broken down. NPs, i.e., PEG-coated iron oxide NPs for miRNA delivery to
Conjugation of DNA with magnetic NPs is mostly achieved overcome drug resistance in gastric cancer cells by enforcing
via the NPs’ surface modification with polyethyleneimine (PEI), miRNA expression in tumor cells are reported.[381] In this con-
especially with low-molecular-weight ones, as it shows strong text, miRNA delivery was combined with doxorubicin treatment
binding affinity and good protection of nucleic acids for trans- as a way to modulate the drug resistance and increase sensi-
fection purposes.[376] This approach is still in the investiga- tivity of tumor cells to doxorubicin therapy. Besides the invis-
tion phase, but it results in more and more promising studies ibility effect, PEG increased the biocompatibility of iron oxide
toward clinical trials. For instance, super-paramagnetic NPs NPs, whereas miRNA was conjugated via electrostatic interac-
with enhanced physiological stability can be achieved with tions. The produced miR16/iron oxide NPs significantly sup-
this method, combining targetable and convenient system for pressed the tumor growth in a drug-resistant gastric cancer
delivering multiple genes into the desired cells or tissues.[377] A mouse model allowing also in vivo imaging of the process.
stable and efficient coexpression of fluorescent proteins could Most recently, multifunctional iron oxide NPs were used to
be achieved by magnetofection. An excellent study for delivery deliver siRNA to downregulate the cell cycle-specific serine–
of both DNA and siRNA was published by Jiang et al.[378] The threonine-kinase, polo-like kinase-1 gene in pancreatic ductal
delivery system was based on iron oxide NPs of varying size adenocarcinoma in animal models.[382] This advanced magnetic
and cationic lipid-like materials, termed lipidoids. The study nanoparticle-based system was further upgraded with a pancre-
shed the light on optimizing the diameter of magnetic NPs atic-cancer-targeting ligand and a peptide for endosomal escape
for specific gene delivery. While siRNA molecules were intra- peptide for specific tumor recognition and effective intracellular
cellularly delivered in similar extents by using different sized delivery. Tumor growth ablation was directly visualized by MRI
NPs, spheres of 50–100 nm exhibited optimized DNA delivery. as a demonstration of simultaneously achieving tumor-specific
Applied magnetic fields expressively increased the efficiency gene therapy and monitoring of the tumor regression.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (22 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

3.5. Lab-on-Chip Applications modification or functionalization compared to flat surfaces or


macroscaled particles. This modification can be performed in
Since the advent of the original concept almost three decades order to modify a wide range of surface properties including
ago,[383] LOC technology is assumed to be one of the leading roughness, hydrophilicity, surface charge, surface energy, bio-
and most promising research areas as a demand for achieving compatibility, reactivity, among others.[397] In particular, the
affordable integrated ever-increasing automated miniatur- use of MNPs in LOC systems provides new possibilities of
ized healthcare systems providing monitoring, diagnosis, and capturing, transporting, labeling, and detecting entities in com-
therapy.[384] The vision beyond the development of LOC plat- plex aqueous systems, as they can be influenced and guided by
forms is to integrate common laboratory functions into one an external magnetic field by means of permanent magnet or
single chip within an area of only a few square centimeters with electromagnets, which is independent of any other biological,
the aptitude to identify toxins, biochemical products, or disease microfluidic, or chemical procedures.[398] Furthermore, mag-
symptoms, and allow biological screening essays or targeted netic manipulation approaches do not harm or change the
drug delivery.[385,386] Moreover, they demonstrated the poten- sample that is imperative, particularly, in biomedical applica-
tial and ability to be applied in a wide range of other techno- tions. The key to develop improved systems is the combination
logical applications beyond healthcare involving environmental of the proper affinity agents onto the surface of MNPs. Specially
monitoring, food, agriculture, and biochemical research.[387,388] hybrid MNPs consisting of ferromagnetic NPs of a few tens
LOC term is often indicated in the literature as an alternative to hundreds of nanometers in diameter embedded in a non-
name for Micro Total Analysis Systems or even microfluidic magnetic matrix, such as polymer, silica, or quartz, have been
systems. An overview of the various operation units that can widely developed not only to stabilize NPs and avoid agglom-
integrate an LOC, including sample preparation, sample injec- eration due to van der Waals forces but also to enable further
tion, sample manipulation, reaction, separation, and detection, functionalization to be used in a wide range of biotechnological
is presented in a series of relevant reviewed articles.[389–391] and biomedical applications, including magnetic separation,
LOC technologies provide numerous benefits over their mac- immunoassays, drug delivery, and hyperthermia studies.[33,399]
roscopic conventional counterparts, including minute con- These nano- and microsized particles with super-paramagnetic
sumption of the reagent and sample due to the miniaturized behavior are frequently called magnetic beads due to their
microchannel dimensions and therefore producing reduced spherical shapes and feature nonmagnetic behavior, unless
amount of waste, relatively fast reaction time by the increased when exposed to a magnetic field. Moreover, they can come
surface-area-to-volume ratio for efficient mass transport, par- with a wide range of sizes, physical and chemical properties,
allel operation for high-throughput analysis, and integration of and their surface functionalization can be tailored with bio-
multifunctional units such as valves, pumps, mixers, actuators, logical and chemical ligands to accomplish specific bioanalyt-
sensors, and detectors for in situ analysis.[318,392] Moreover, they ical tasks. Such beads can be usefully utilized as vehicle sub-
have the potential to be low cost, portable, and of simple and strates in bioassays, catalysis or even in vivo uses since they
practical handling. Microfluidic systems were originally fabri- can be easily recovered from a dispersion or as packed beds to
cated in silicon and glass, but as the field advanced, the focus enhance interaction of reactive surfaces with fluids passing by
shifted to cheaper, simpler, and less consuming manufacturing the microchannel of the microfluidic system.[400]
techniques involving polymer substrates, particularly polydi- An important application of microfluidic technology that has
methylsiloxane (PDMS) and other distinct materials.[391,393] shown promising potential is the synthesis of nanomaterials,
More recently, paper emerges as a promising alternative flex- including MNPs for a wide range of biotechnological applica-
ible and versatile material with unprecedented easy-to-use solu- tions, where critical stages such as nucleation, growth, and
tion for low-cost and point-of-care devices that come from its reaction conditions can be modulated in order to control and
mechanical flexibility and strength, low cost, high availability, tailor morphology, size, size distribution, and reproducibility.
and easy chemical functionalization. Its porous structure, com- In fact, in contrast to conventional batch methods, precise con-
posed of a network of hydrophilic cellulose fibers, enables the trol, rapid heat and mass transfer, high mixing efficiency, and
transport of fluids through capillary action, without the need large reaction interfaces can be achieved in continuous-flow
for external forces such as pumps.[394–396] The required func- microfluidic systems with online analysis. A PDMS T-junction
tion, degree of integration, and application are the main factors microfluidic system combined with an aluminum foil-based
to be considered when choosing a design and material, which UV light reflector was developed to controllably synthesize
are closely related to materials’ properties and manufacturing magnetic (γ-Fe2O3) hydrogel microparticles in spherical and
processes.[391] Moreover, biocompatibility, flexibility, optical nonspherical shapes. The MNPs were successfully encapsu-
transparency, air permeability, thermal stability, electrical con- lated in the hydrogel, providing the microparticles super-para-
ductivity, and nonspecific adsorption are physical features that magnetic properties that is required in most biotechnological
may set as the material choice. applications. Moreover, nonspherical particles have anisotropic
Parallel to the boom of LOC, nanomaterials attracted large responses under an external magnetic field, offering features in
attention in the scientific community and have been combined various fundamental studies and applications.[91] Hollow Fe/Ga-
with these platforms to improve the analytical performance based oxide nanospheres were also synthesized using a PDMS
and overcome the frequent constraint inherent to miniaturiza- microfluidic bioreactor comprising a micromixer, a microcon-
tion in terms of sensitivity and selectivity, especially when con- denser channel, microvalves, a microheater, and a microtem-
cerning the analysis of real samples.[397] This is mostly related perature sensor. The developed NPs feature a diameter of about
to their superior surface-to-volume area for further surface 157 ± 26 nm and saturated mass magnetization of 14.4 emu g−1

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (23 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

at 300 K increasing to 18.8 emu g−1 at 5 K. The microfluidic magnetic field. The successful culture of glioblastoma multi-
system may be a promising tool to produce hollow NPs for form cells proved that the developed microfibers can provide a
drug delivery purposes.[401] A natural-rubber-based microfluidic good structural integrity and environment for cell culture and
device was designed and fabricated for the synthesis of mag- tissue engineering applications that could be applied in drug
netic-decorated gold NPs (Fe3O4–AuNPs), which is a ferromag- screening for targeting cancer cells.[404] Relevant reviews that
netic material with extraordinary potential for applications in address nanomaterial synthesis in microfluidic systems can be
immunosensors, in photothermal ablation of tumor cells, and found in refs. [90,405–407].
as a contrast agent for imaging diagnosis. Monodispersed Fe3O4 Recent technological advances also provide new tools to
NPs (10 nm) homogeneously decorated with uniform AuNPs manipulate entities in micro/nanoliter volumes. In fact, the
(4 nm), free of organic solvent and surfactants, were obtained.[402] capability of capturing and isolating biological entities at the
Biomimetic cell membrane-coated NPs were also obtained by microscale level, combining LOC and MNP technologies, cre-
means of a PDMS microfluidic electroporation system. When ates the potential for cost effective, portable, and disposable
a mixture of Fe3O4 MNPs and red blood cell-membrane-derived point-of-care systems to monitor and detect various diseases.
vesicles (RBC vesicles) flow through the electroporation zone, In particular, the detection and isolation of circulating tumor
applied electric pulses promote the entry of the MNPs into cells (CTCs) play a key role in the diagnosis and prognosis of
the RBC vesicles. In vivo experiments of the resulting mate- cancer, while the high capture efficiency and purity of CTCs are
rials enhanced tumor MRI and photothermal therapy owing difficult to reach simultaneously among the various conven-
to the superior magnetic and photothermal properties of the tional isolation methods due to the small throughput, incapa-
MNP cores and the long blood circulation characteristics of bility to release captured cells, and dependence on expensive
the RBC membrane shells.[403] Another interesting and dif- instrumentation for enrichment or subsequent characteriza-
ferent microfluidic system was designed for fabricating, in a tion.[408] In an attempt to take a step forward to overcome the
one-step continuous process, sophisticated alginate microfibers aforementioned limitation, an inverted microchip integrating
(Figure 14a).[404] A fluid-focusing technique with two dimen- silicon nanowires (SiNW) and multifunctional magnetic nano-
sions, multi-inlets, and multijunctions was utilized to control composites based on anti-EpCAM-Fe3O4@C6/Ce6@silane
the unceasing laminar flow of alginate solutions without them (anti-epithelial cell adhesion molecule (EpCAM)) was designed
clogging the microchannels (Figure 14b). The diameter of the to enhanced capture efficiency and purity of CTCs.[408] The
microfibers (in the 177.4–364.3 µm range) is simply managed system allows real-time detection and photodynamic therapy
by regulating the flow rate and channel size of the microflu- of CTCs and showed improved capture purity of CTCs to 90%
idic system (Figure 1c–f). Diclofenac-loaded microfibers exhibit and CTCs’ capture efficiencies of 90.3% and 82% in culture
a drug release profile characteristic of a proper and steady medium and blood samples, respectively. A similar system
release. Furthermore, the release kinetics of diclofenac could composed of anti-EpCAM conjugated with magnetic upcon-
be actively controlled by magnetic iron-oxide-loaded micro- version NPs was introduced into a microfluidic chip for spe-
fibers, allowing for a rapid drug release by applying an external cific recognition and detection of CTCs with enhanced CTC

Figure 14.  a) Photograph of the microfluidic chip: 1, inlets of center channels; 2 and 3, inlets of side channels; 4, cross junction; 5, outlet; 6, obser-
vation channel; 7, bottom layer disk; 8, screw orifices; 9, the scale bar of 11 cm. b) Schematic representation of the microfiber formation: 1, 2 wt%
CaCl2 solution; 2, deionized water; 3, alginate solution; 4, sunflower seed oil; 5, observation channel; 6, microfibers. The formation of microfibers: A,
photograph of the microfiber in the observation channel; B, photograph of the second cross junction; C, photograph of the first cross junction. c,e)
Microscopic images of microfibers stained with rhodamine B. d,f) Scanning electron microscopy images of microfibers. Adapted with permission.[404]
Copyright 2012, the authors, published under the terms of CC-BY license.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (24 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

capture obtained by an integrated SiNW array that allows the the amount of CD4+ cells in blood and the packing level
further convenient release of captured CTCs for analysis and detected in the capture region was observed.[418] An interesting
reculturing. The systems demonstrate good consistency in real review about the use of nano- and microfluidic technologies for
clinical samples for lung cancer patients.[409] A magnetic-force- diagnosis of infectious diseases in developing countries can be
gradient-based microfluidic chip was also reported for simulta- found in ref. [419].
neously isolating CTCs in the sample and characterizes their Viral infectious diseases, such as severe acute respiratory
state with respect to EpCAM expression level. The systems syndromes including norovirus and influenza, have been a
used Fe3O4 NPs functionalized with (3-glycidylox-ypropyl)tri- severe threat to human health worldwide, and therefore the
methoxysilane for immobilization of EpCAm antibody on their capability of rapidly detecting this kind of infections is a vital
surface for the specific binding with CTCs. The system was able need to be addressed in order to provide immediate and appro-
to isolate 3 mL of the heterogeneous CTC sample in 1 h with priate clinical treatment. A suction-type microfluidic system
high isolating yield demonstrating its potential to shorten the using mAb-bound magnetic beads was developed for the rapid
date to diagnose the cancer metastasis in clinic.[410] A different and automatic immunological diagnosis of influenza infection.
approach through a microfluidic platform comprising three The system comprises a control and optical detection module
different modules demonstrated high-throughput separation to allow biosample incubation, purification, and optical analysis
of cancer cells from blood and on-chip organization of those automatically. Influenza concentration of 0.128 HAU with a
cells for streamlined analysis. While the first module employs detection time of 30 min was obtained with the system.[420] A
acoustic stranding waves to rapidly align cells, the second more promising integrated microfluidic device with dynamic
module separates magnetically labeled cells from unlabeled mixing was later developed to perform the automatic detec-
cells, offering purities beyond 85% for cells and 90% for binary tion of influenza infection using magnetic manganese fer-
mixtures of synthetic particles. The last module integrates an rite (MnFe2O4) NPs with ≈100 nm diameter. This NP-based
array of microwells with underlying micromagnets to capture immunoassay combined with layer-by-layer surface modi-
individual cells for on-chip analysis. The high capture efficiency fication achieved a detection limit of influenza as low as
of 80% of magnetically labeled cells demonstrates the potential 0.007 HAU within 20 min, showing promise for rapidly diagnosis
of this system not just for the isolation and evaluation of rare of infectious diseases. The fluorescence immunoassay signal
cancer cells, as it can be readily extended to address a variety was increased by approximately twofold when compared with
of applications across single cell biology and immunology.[411] commercial 4.5 µm magnetic beads due to the large surface-to-
Extensive reviews of the latest advances in microfluidic rare cell volume ratio.[421]
separation and processing can be found in refs. [412–415]. Magnetic microfluidic arrangements were also established
New techniques for HIV diagnosis and monitoring are also for the diagnosis of malaria, which is a mosquito- transmitted
needed to overcome the high cost, technically complex and infectious illness that affects more than 3 billion people, ≈50%
time-consuming conventional techniques to measure CD4+ cell of the total world’s population. An example is the system pre-
count such as flow cytometers, particularly when it comes to sented in Figure 15 that combines the use of microfluidic tech-
developing countries. So far, several CD4+ cell counting tech- nology for the separation of infected red blood cells (iRBCs) and
niques have been implemented using microfluidic systems, the magnetic resonance relaxometry system for the label-free
which require precise manipulation of blood on-chip[416] A and fast detection of malaria parasites. The iRBCs are separated
simple, rapid, and inexpensive CD4+ isolation and counting from the healthy red blood cells (hRBCs) through margination.
device based on antimouse CD4 magnetic beads bed immu- Moreover, it was shown that the existence of hemozoin crystal-
noassay was reported. CD4+ were successfully separated and lites or natural paramagnetic malaria pigment in iRBCs serves
captured at a maximum capability of about 700 cell µL−1. The as a natural biomarker for malaria recognition, where the bulk
system has the ability to avoid complex sample pretreatment magnetic susceptibility of the iRBCs is considerably higher than
and the entire analysis time was significantly reduced to 15 min. the hRBCs. This induces a substantial change in the transverse
Though the separation of CD4+ from mouse was taken as a relaxation rate. The microfluidic system demonstrates the capa-
model, it can also be used to separate and count human CD4+ bility of detecting ring stage parasites at parasitemia as low as
with the antihuman CD4 magnetic beads, with the objective 0.0005%. The approaches used are envisaged to provide a new
to monitor HIV in resource-limited settings.[417] A dual-force malaria diagnosis solution in the near future (Figure 15).[422]
cellular separation strategy using centrifuge–magnetophoresis Other microfluidic diagnosis systems were developed for the
was also developed and demonstrated separation efficiency detection and quantification of relevant biological entities. Glu-
up to 92% for cells expressing CD4+ from whole blood. The cose and creatinine were successfully measured at (2–8) × 10−3
system uses a combination of super-paramagnetic Dynabeads and (10−2–10) × 10−3 m, respectively, in a blood serum by means
CD4 MNPs that specifically bind phenotypic markers on target of enzyme-coated magnetic beads immobilized on the surface
cells and centrifugal force to isolate magnetically specific cel- of an electrolyte–insulator–semiconductor sensor though an
lular phenotype.[418] Another system to isolate magnetically external magnetic force on a microfluidic chip. The immobili-
tagged CD4+ cells directly from patient blood works by dual- zation method addresses the concern of long-term preservation
force CD4+ magnetophoresis, through which the interplay of of enzymes in microchannels.[423] Another low-cost and effi-
magnetic forces and flow controls the trajectory of target cells cient microfluidic system was designed to quantify glucose by
depending on whether the cells binds to a magnetic bead. High packing enzyme modified MNPs as an enzyme microreactor.
capture efficiency of about 93% was obtained from a back- The optimal linear ranges from 25 × 10−6 to 15 × 10−3 m with a
ground of nonmagnetic particles, and a linear relation between detection limit of 11 × 10−6 m and a reproducibility of 95.6%.[424]

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (25 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Figure 15.  Schematic representation of a) microfluidic microagitation device comprising two microfluidic components: a margination layer and a cover
layer. The margination layer consists of seven parallel margination channels. The yellow circles indicate the tubing ports’ locations. b) Zoom-in of the
inlet and filtration unit. The microstructures and pillars trap the coagulated blood and large debris to avoid channel blockage in the long and narrow
margination channels. c) Bifurcation outlet at the end of each margination channel. The hRBCs migrate to the axial axis of the microchannel and exit
via the middle outlet, while the stiffer iRBCs are directed to the sidewalls and exit via the side outlets. d) Complete microfluidic separation device with
one inlet, two outlets, and a margination channel. The insets show the schematic of the cross-sectional view of hRBC and iRBC distributions before
and after passing through the margination channel. The initially randomly distributed stiffer iRBCs are directed toward the sidewall, while the more
deformable hRBCs migrate toward the axial center of the microchannel. e) Bench top MRR system. The blood sample is centrifuged in a microcapillary
tube and loaded into MRR receiver coil placed inside a 0.5 T permanent magnet. The receiver coil is connected to a bench-top spectrometer for MRR
measurements. Adapted with permission.[422] Copyright 2015, the authors, published under the terms of CC-BY 4.0 license.

Another interesting microfluidic immunosensor was developed A different two-step microfluidic essay based on immuno-
for the electrochemical detection and quantification of immu- extraction and enrichment in situ was developed to detect the
noreactive trypsinogen (IRT) as marker for cystic fibrosis diag- specific presence of Salmonella typhimurium from a complex
nosis. The system features two distinct approaches: the incor- liquid sample based on a magnetic fluidize bed using Dyna-
poration of MNPs into the central microfluidic channels and beads anti-Salmonella. The system allows for a precise quanti-
performing as a bioaffinity scaffold for the immobilization of fication of the initial bacteria coupled with a very high sensi-
the antibody against the immunoreactive trypsin (anti-IRT) tivity, after a few hours versus 2 d in conventional cell culture
and the electrodeposition of copper NPs on a gold electrode. platforms.[427] Helicobacter pylori is another relevant micro-
Compared with the conventional enzyme-linked immuno- organism that affects around 10% of the world population
sorbent assay (ELISA) analysis, the developed immunosensor causing peptic ulcers and chronic gastritis. Noninvasive tests
reduces the total assay time in less than 40 min.[425] that include serological tests and the urea breath test have been
The detection and identification of bacteria is also crucial pointed out as viable alternatives to invasive methods such
in many fields of science and society. A automated microflu- as endoscopy with biopsies for histology, culture, and rapid
idic chip-based μ-Hall platform capable of measuring single, urease test. Recently, a microfluidic magnetic immunosensor
magnetically tagged bacteria directly in clinical specimens was coupled to a gold electrode for the fast and very sensitive quan-
developed (Figure 16).[426] Besides the minimal processing tification of human serum immunoglobulin G (IgG) anti-
and robust detection in different biological background, bodies to H. pylori was documented. The detection in serum
the microfluidic system features high-throughput detection samples is performed using a noncompetitive immunoassay
(107 cells min−1) in a simple and automatic compact and self- based on the utilization of purified H. pylori antigens coupled
contained device. The system was successfully tested for the with 3-aminopropyl-modified magnetic microspheres. The
detection of Gram-positive bacteria using vancomycin as the response obtained from the product reaction is directly pro-
targeting ligand. The overall detection limit of the system was portional to amount of IgG antibodies to H. pylori in serum
similar to that of the culture platform but the essay time was samples. The determined limits for electrochemical detection
50 times faster. Such results demonstrate the potential of this and the ELISA procedure were 0.37 and 2.1 U mL−1, respec-
powerful diagnosis tool for controlling infectious diseases and tively, and the within- and between-assay coefficients of varia-
promoting public health. tion were less than 5%. Moreover, the electrochemical detection

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (26 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

Another method to clearing bacteria and endotoxin from the


bloodstream, using MNPs modified with bis-Zn-dipicolylamine
(DPA), a synthetic ligand that binds to both Gram-positive and
Gram-negative bacteria, was developed in order to get a step
forward in bacterial sepsis, which is a serious clinical condition
that can lead to multiple organ malfunction and death despite
appropriate treatment with antibodies and fluid resuscitation.
A magnetic microfluidic system was designed to remove the
MNPs connected to Escherichia coli, a Gram-negative bacte-
rium usually involved in bacterial sepsis, from a bovine whole
blood resulting in almost 100% clearance.[429] Other magnetic
microfluidic systems have been reported in the literature for
the detection of relevant microorganisms, including Vibrio par-
ahaemolyticus,[430] Staphylococcus aureus,[431,432] Listeria monocy-
togenes,[433–435] among others.
This novel and exciting area of miniaturized applications of
magnetic beads with all their inherent benefits compared to
conventional counterparts offers many possibilities for future
development in an extensively wide range of biotechnological
fields, far beyond those previously exemplified, including tissue
engineering,[436,437] proteomics,[438–440] hyperthermia,[441,442]
and pharmacology,[404,443,444] among other biomedical
applications.[414,445–448]

4. Summary and Outlook


Magnetic NPs for advanced biomedical applications are an aus-
picious research field with huge and increasing interest. Rep-
resentative examples of the main obtained results in each area
of application have been summarized, indicating also the chal-
lenges that still remain to be overcome, such as the tendency to
aggregate or the poor magnetization of the smallest NPs used
in biomedical applications. Moreover, the selection of the proper
synthesis method is often among the most crucial aspects, as it
strongly determines nanoparticle’s characteristics and, therefore,
Figure 16.  Design and implementation of the bacterial μHall sensor. a) A also their functionality. Thus, it is important to establish proper
photograph of the μHall device shows an array of μHall sensors incorpo- large-scale synthesis protocols with accurate control of size and
rated onto a semiconductor substrate with PDMS microfluidics directly on shape of the different NPs, including their proper biofunction-
top. b) Eight μHall sensors, each measuring 8 µm × 8 µm were arranged alization. Though iron oxides are relatively less toxic compared
into an overlapping array across the fluidic channel. The dotted lines indi- with other semiconductor nanomaterials or transition metals,
cate the width of the sample flow. c) The voltage signal (VH) was simulated
concerns regarding toxicity still remain regarding Fe2O3-based
as a function of the vertical distance d between the center of a bacterium
and the surface of the Hall sensor. d) To enhance the μHall signal, a hydro- MNP composites.[449] Such an issue must be definitely solved in
dynamic focusing structure was used to bring individual pathogens close to the near future with clear and well-established procedures. Thus,
the μHall sensor. With hydrodynamic focusing, bacteria entering the chip the specific characterizations of size, shape, dose, and content
though the sample input port were pushed to the bottom of the channel of functional NPs should be classified in relation to their effi-
by the vertical sheath flow (flow rate, SV) and focused toward the center of ciency in disease treatments. Although such NPs are very prom-
the channel by the lateral sheaths (flow rate, SL). The finite-element simu- ising candidates for medicine and biology, they show a certain
lation shows the focusing of bacteria to a region of ≈100 µm × 7.5 µm,
within a physical channel measuring 200 µm × 15 µm. The flow rates were
degree of toxicity in both animals and humans. After some suc-
SV = 3S and SL = 8S, where S is the sample flow rate. e) A fluorescence cessful tests in animals with promising results, drug delivery
micrographic image of hydrodynamic focusing shows that focusing can through coating of MNPs is currently undergoing preliminary
be controlled by varying the relative flow rates between the vertical and human trials; nevertheless, it will take some time before them
lateral sheaths. F) With flow focusing, robust VH signals from individual be clinically available. Similarly, important steps must be given
bacteria could be observed. The graph shows the VH of a single Staphylo- in hyperthermia treatment. Despite such a procedure already
coccus aureus passing over a μHall sensor. Reproduced with permission.[426]
proven to be effective in animal trials (both at the regulatory
level and in the determination of specific materials and proce-
was done within 1 min, and total essay time is 25 min. These dures), it remains inaccessible to humans.[450] Theranostics and
results proved the potential usefulness of the fabricated micro- tissue engineering applications show themselves as highly inter-
chip for the early assessment of IgG antibodies to H. pylori.[428] esting areas, in which strong research efforts must be devoted to

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (27 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

highlighting novel diagnosis and therapeutic strategies. Further- [1] Z. Hedayatnasab, F. Abnisa, W. M. A. W. Daud, Mater. Des. 2017,
more, microfluidic systems are one of the areas that is consist- 123, 174.
ently supporting the whole path of magnetic nanoparticle-based [2] F. Zeinali Sehrig, et al., Artif. Cells, Nanomed., Biotechnol. 2016, 44,
biomedicine, offering proper solutions and reproducibility from 918.
[3] A. H. Lu, E. L. Salabas, F. Schüth, Angew. Chem., Int. Ed. 2007, 46,
nanoparticle synthesis to diagnostics, passing through devel-
1222.
oping tests in cell culture, drug development, and theranostics, [4] A. Yadollahpour, S. Rashidi, Orient. J. Chem. 2015, 31, 25.
among others. [5] P. Tartaj, M. Del Puerto Morales, S. Veintemillas-Verdaguer,
The future applications and challenges in magnetic nanopar- T. González-Carreño, C. J. Serna, J. Phys. D: Appl. Phys. 2003, 36,
ticle-based biomedicine pass through the definitive clarification R182.
of the toxicological aspects in nanoscale of NPs and nanosystems [6] B. Zhang, W. Yang, J. Yu, W. Guo, J. Wang, S. Liu, Y. Xiao, D. Shi,
both in in vivo and in vitro trials as well as the determination of Adv. Healthcare Mater. 2017, 6, 1600865.
the long-term stability of the functionalized MNPs before they can [7] P. Martins, M. Silva, S. Lanceros-Mendez, Nanoscale 2015, 7,
be widely introduced for their clinical use.[451] Further, interdisci- 9457.
plinary approaches are needed to guarantee that the experiments [8] D. T. Nguyen, K. S. Kim, Korean J. Chem. Eng. 2014, 31, 1289.
[9] X. Li, J. Wei, K. E. Aifantis, Y. Fan, Q. Feng, F. Z. Cui, F. Watari,
performed in laboratory can more clearly match the estimated
J. Biomed. Mater. Res., Part A 2016, 104, 1285.
conditions that would be faced in vivo, allowing proper steps [10] T. Neuberger, B. Schöpf, H. Hofmann, M. Hofmann,
toward applications. Substantial contributions through mathemat- B. Von Rechenberg, J. Magn. Magn. Mater. 2005, 293, 483.
ical modeling of more and smart complex systems, with the objec- [11] L. H. Reddy, J. L. Arias, J. Nicolas, P. Couvreur, Chem. Rev. 2012,
tive of understanding, more specifically, the complex interactions 112, 5818.
and effects, will determine whether, in the final analysis, a given [12] R. Langer, Science 1990, 249, 1527.
biomedical approach can be successfully used.[450] [13] S. Carregal-Romero, E. Caballero-D’Iaz, L. Beqa,
Thus, it has been shown that magnetic nanoparticle-based A. M. Abdelmonem, M. Ochs, D. Ḣ˙uhn, B. S. Suau, M. Valcarcel,
materials and biomedical approaches represent a wide and W. J. Parak, Annu. Rev. Anal. Chem. 2013, 53.
strong interdisciplinary research and application field. Impor- [14] C. S. Chiang, Y. S. Shen, J. J. Liu, W. C. Shyu, S. Y. Chen, Adv.
Healthcare Mater. 2016, 5, 2131.
tant advances have been achieved in the development of suit-
[15] M. Iranifam, TrAC, Trends Anal. Chem. 2013, 51, 51.
able MNPs and their functionalization. Interesting proofs- [16] S. Laurent, S. Dutz, U. O. Häfeli, M. Mahmoudi, Adv. Colloid Inter-
of-concept in a large variety of biomedical areas have demon- face Sci. 2011, 166, 8.
strated the full potential of this research area, representing an [17] K. Ulbrich, K. Holá, V. Šubr, A. Bakandritsos, J. Tuček, R. Zbořil,
important (re)evolution within the whole biomedical field. Chem. Rev. 2016, 116, 5338.
[18] S. Gil, J. F. Mano, Biomater. Sci. 2014, 2, 812.
[19] J. H. Ryu, H. Koo, I. C. Sun, S. H. Yuk, K. Choi, K. Kim, I. C. Kwon,
Adv. Drug Delivery Rev. 2012, 64, 1447.
Acknowledgements [20] H. Peng, J. Tang, R. Zheng, G. Guo, A. Dong, Y. Wang, W. Yang,
V.F.C. and A.F. contributed equally to this work. The authors thank the Adv. Healthcare Mater. 2017, 6, 1601289.
FCT—Fundação para a Ciência e Tecnologia—for financial support under [21] V. Iacovacci, G. Lucarini, L. Ricotti, A. Menciassi, Magnetic Field-Based
framework of the Strategic Funding UID/FIS/04650/2013, project PTDC/ Technologies for Lab-on-a-Chip Applications, (Eds.: M. Stoytcheva,
EEI-SII/5582/2014 and project UID/EEA/04436/2013 by FEDER funds R. Zlatev), InTechOpen, London, United Kingdom 2016.
through the COMPETE 2020—Programa Operacional Competitividade e [22] Z. Li, J. C. Barnes, A. Bosoy, J. F. Stoddart, J. I. Zink, Chem. Soc.
Internacionalização (POCI). Funds provided by FCT in the framework of Rev. 2012, 41, 2590.
EuroNanoMed 2016 call, Project LungChek ENMed/0049/2016 are also [23] Y. Panahi, M. Farshbaf, M. Mohammadhosseini, M. Mirahadi,
gratefully acknowledged. V.F.C., A.F., C.R., and P.M. also thank the FCT R. Khalilov, S. Saghfi, A. Akbarzadeh, Artif. Cells, Nanomed., Bio-
for the grants SFRH/BPD/98109/2013, SFRH/BPD/104204/2014, SFRH/ technol. 2017, 45, 788.
BPD/90870/2012 and SFRH/BPD/96227/2013, respectively. Finally, [24] P. Kerativitayanan, J. K. Carrow, A. K. Gaharwar, Adv. Healthcare
the authors acknowledge funding by the Spanish Ministry of Economy Mater. 2015, 4, 1600.
and Competitiveness (MINECO) through the project MAT2016-76039-
[25] A. J. McGrath, C. Dolan, S. Cheong, D. A. J. Herman, B. Naysmith,
C4-3-R (AEI/FEDER, UE) and from the Basque Government Industry
F. Zong, P. Galvosas, K. J. Farrand, I. F. Hermans, M. Brimble,
Department under the ELKARTEK program.
D. E. Williams, J. Jin, R. D. Tilley, J. Magn. Magn. Mater. 2017, 439,
251.
[26] X. Mao, J. Xu, H. Cui, Wiley Interdiscip. Rev.: Nanomed. Nanobio-
Conflict of Interest technol. 2016, 8, 814.
[27] K. El-Boubbou, D. Azar, A. Bekdash, R. J. Abi-Habib, J. Biomed.
The authors declare no conflict of interest. Nanotechnol. 2017, 13, 500.
[28] L. Mohammed, D. Ragab, H. Gomaa, Curr. Pharm. Des. 2016, 22,
3332.
[29] D. H. Kim, Y. Guo, Z. Zhang, D. Procissi, J. Nicolai, R. A. Omary,
Keywords A. C. Larson, Adv. Healthcare Mater. 2014, 3, 714.
drug release, hyperthermia, lab-on-chip, magnetic nanoparticles, tissue [30] C. Blanco-Andujar, A. Walter, G. Cotin, C. Bordeianu,
engineering D. Mertz, D. Felder-Flesch, S. Begin-Colin, Nanomedicine 2016, 11,
1889.
Received: July 14, 2017 [31] S. S. Banerjee, A. Jalota-Badhwar, S. D. Satavalekar, S. G. Bhansali,
Revised: September 28, 2017 N. D. Aher, R. R. Mascarenhas, D. Paul, S. Sharma, J. J. Khandare,
Published online: December 27, 2017 Adv. Healthcare Mater. 2013, 2, 800.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (28 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[32] B. D. Plouffe, S. K. Murthy, L. H. Lewis, Rep. Prog. Phys. 2015, 78, [69] H. Zhang, G. Zhu, Appl. Surf. Sci. 2012, 258, 4952.
016601. [70] S. Shabestari Khiabani, M. Farshbaf, A. Akbarzadeh, S. Davaran,
[33] V. F. Cardoso, V. Sebastián, C. J. R. Silva, G. Botelho, Artif. Cells, Nanomed., Biotechnol. 2017, 45, 6.
S. Lanceros-Méndez, Colloids Surf., B 2017, 157, 48. [71] V. Sreeja, P. A. Joy, Mater. Res. Bull. 2007, 42, 1570.
[34] Y. Pan, X. Du, F. Zhao, B. Xu, Chem. Soc. Rev. 2012, 41, 2912. [72] R. S. Melo, F. C. Silva, K. R. M. Moura, A. S. De Menezes,
[35] Y. W. Chu, D. A. Engebretson, J. R. Carey, J. Biomed. Nanotechnol. F. S. M. Sinfrônio, J. Magn. Magn. Mater. 2015, 381, 109.
2013, 9, 1951. [73] C. Okoli, M. Sanchez-Dominguez, M. Boutonnet, S. Järås,
[36] W. Sheng, W. Wei, J. Li, X. Qi, G. Zuo, Q. Chen, X. Pan, W. Dong, C. Civera, C. Solans, G. R. Kuttuva, Langmuir 2012, 28, 8479.
Appl. Surf. Sci. 2016, 387, 1116. [74] P. Laokul, S. Arthan, S. Maensiri, E. Swatsitang, J. Supercond.
[37] N. Bajwa, N. K. Mehra, K. Jain, N. K. Jain, Artif. Cells, Nanomed., Novel Magn. 2015, 28, 2483.
Biotechnol. 2016, 44, 758. [75] F. Foroughi, S. A. Hassanzadeh-Tabrizi, A. Bigham, Mater. Sci.
[38] R. Alshehri, A. M. Ilyas, A. Hasan, A. Arnaout, F. Ahmed, Eng., C 2016, 68, 774.
A. Memic, J. Med. Chem. 2016, 59, 8149. [76] Q. Zhou, Y. Hu, Y. Ma, Y. Weng, Adv. Mater. Res. 2011, 287, 494.
[39] H. Daraee, A. Eatemadi, E. Abbasi, S. F. Aval, M. Kouhi, [77] A. Najafi, K. Nematipour, J. Supercond. Novel Magn. 2017, 30,
A. Akbarzadeh, Artif. Cells, Nanomed., Biotechnol. 2016, 44, 410. 2647.
[40] Y. Wang, Q. Zhao, N. Han, L. Bai, J. Li, J. Liu, E. Che, L. Hu, [78] H. Alijani, M. H. Beyki, Z. Shariatinia, M. Bayat, F. Shemirani,
Q. Zhang, T. Jiang, S. Wang, Nanomedicine 2015, 11, 313. Chem. Eng. J. 2014, 253, 456.
[41] Z. Tang, J. Chen, Y. Bai, S.  Zhao, Smart Mater. Struct. 2016, 25, [79] G. Carraro, D. Barreca, C. MacCato, E. Bontempi, L. E. Depero,
085020. C. De Julián Fernández, A. Caneschi, CrystEngComm 2013, 15,
[42] K. Yan, P. Li, H. Zhu, Y. Zhou, J. Ding, J. Shen, Z. Li, Z. Xu, 1039.
P. K. Chu, RSC Adv. 2013, 3, 10598. [80] V. N. Dhage, M. L. Mane, A. P. Keche, C. T. Birajdar, K. M. Jadhav,
[43] S. Majidi, F. Z. Sehrig, S. M. Farkhani, M. S. Goloujeh, Phys. Rev. B: Condens. Matter Mater. Phys. 2011, 406, 789.
A. Akbarzadeh, Artif. Cells, Nanomed., Biotechnol. 2016, 44, 722. [81] M. L. Dinesha, H. S. Jayanna, S. Mohanty, S. Ravi, J. Alloys Compd.
[44] W. Wu, Z. Wu, T. Yu, C. Jiang, W. S. Kim, Sci. Technol. Adv. Mater. 2010, 490, 618.
2015, 16, 023501. [82] O. Volotskova, I. Levchenko, A. Shashurin, Y. Raitses, K. Ostrikov,
[45] J. Shan, L. Wang, H. Yu, J. Ji, W. A. Amer, Y. Chen, G. Jing, M. Keidar, Nanoscale 2010, 2, 2281.
H. Khalid, M. Akram, N. M. Abbasi, Mater. Sci. Technol. 2016, 32, [83] M. Bystrzejewski, O. Łabedź, W. Kaszuwara, A. Huczko, H. Lange,
602. Powder Technol. 2013, 246, 70.
[46] H. Rui, R. Xing, Z. Xu, Y. Hou, S. Goo, S. Sun, Adv. Mater. 2010, [84] S. M. Reddy, J. J. Park, S. M. Na, M. M. Maqableh, A. B. Flatau,
22, 2729. B. J. H. Stadler, Adv. Funct. Mater. 2011, 21, 4677.
[47] C. Xu, S. Sun, Adv. Drug Delivery Rev. 2013, 65, 732. [85] D. Ramimoghadam, S. Bagheri, S. B. A. Hamid, J. Magn. Magn.
[48] F. Zeinali Sehrig, S. Majidi, N. Nikzamir, M. Nikzamir, Mater. 2014, 368, 207.
A. Akbarzadeh, Artif. Cells, Nanomed., Biotechnol. 2016, 44, 918. [86] I. Morjan, R. Alexandrescu, F. Dumitrache, R. Birjega, C. Fleaca,
[49] J. Yu, F. Liu, Y. Muhammad Zubair, Y. L. Hou, Prog. Biochem. Bio- I. Soare, C. R. Luculescu, G. Filoti, V. Kuncer, L. Vekas, N. C. Popa,
phys. 2013, 40, 903. G. Prodan, V. Ciupina, J. Nanosci. Nanotechnol. 2010, 10, 1223.
[50] E. Tombácz, R. Turcu, V. Socoliuc, L. Vékás, Biochem. Biophys. Res. [87] G. Schinteie, V. Kuncser, P. Palade, F. Dumitrache, R. Alexandrescu,
Commun. 2015, 468, 442. I. Morjan, G. Filoti, J. Alloys Compd. 2013, 564, 27.
[51] N. T. K. Thanh, L. A. W. Green, Nano Today 2010, 5, 213. [88] K. B. Narayanan, N. Sakthivel, Adv. Colloid Interface Sci. 2010, 156, 1.
[52] E. Amstad, M. Textor, E. Reimhult, Nanoscale 2011, 3, 2819. [89] J. W. Moon, C. J. Rawn, A. J. Rondinone, L. J. Love, Y. Roh,
[53] A. Yadollahpour, Orient. J. Chem. 2015, 31, 271. S. M. Everett, R. J. Lauf, T. J. Phelps, J. Ind. Microbiol. Biotechnol.
[54] V. F. Cardoso, S. Irusta, N. Navascues, S. Lanceros-Mendez, 2010, 37, 1023.
Mater. Res. Express 2016, 3, 075402. [90] J. Ma, S. M. Y. Lee, C. Yi, C. W. Li, Lab Chip 2017, 17, 209.
[55] M. Faraji, Y. Yamini, M. Rezaee, J. Iran. Chem. Soc. 2010, 7, 1. [91] D. K. Hwang, D. Dendukuri, P. S. Doyle, Lab Chip 2008, 8, 1640.
[56] R. Massart, IEEE Trans. Magn. 1981, 17, 1247. [92] A. B. Salunkhe, V. M. Khot, S. H. Pawar, Curr. Top. Med. Chem.
[57] M. Karimi, S. A. Hassanzadeh-Tabrizi, A. Saffar-Teluri, J. Sol-Gel 2014, 14, 572.
Sci. Technol. 2017, 83, 124. [93] H. Markides, M. Rotherham, A. J. El Haj, J. Nanomater. 2012,
[58] Y. Li, K. Hu, B. Chen, Y. Liang, F. Fan, J. Sun, Y. Zhang, N. Gu, 2012.
Colloids Surf., A 2017, 520, 348. [94] A. Ruiz, P. C. Morais, R. Bentes de Azevedo, Z. G. M. Lacava,
[59] S. Lin, K. Lin, D. Lu, Z. Liu, J. Environ. Chem. Eng. 2017, 5, 303. A. Villanueva, M. del Puerto Morales, J. Nanopart. Res. 2014, 16,
[60] E. Suharyadi, E. A. Setiadi, N. Shabrina, T. Kato, S. Iwata, Adv. 2589.
Mater. Res. 2014, 896, 126. [95] M. Calero, L. Gutiérrez, G. Salas, Y. Luengo, A. Lázaro, P. Acedo,
[61] J. H. Kim, S. M. Kim, Y. I. Kim, J. Nanosci. Nanotechnol. 2014, 14, M. P. Morales, R. Miranda, A. Villanueva, Nanomedicine 2014, 10,
8739. 733.
[62] P. Arévalo, J. Isasi, A. C. Caballero, J. F. Marco, F. Martín- [96] A. Ruiz, Y. Hernández, C. Cabal, E. González, S. Veintemillas-
Hernández, Ceram. Int. 2017, 43, 10333. Verdaguer, E. Martínez, M. P. Morales, Nanoscale 2013, 5, 11400.
[63] D. Soundararajan, K. H. Kim, J. Magn. 2014, 19, 5. [97] F. M. Kievit, M. Zhang, Acc. Chem. Res. 2011, 44, 853.
[64] X. Wu, K. Zhou, W. Wu, X. Cui, Y. Li, J. Therm. Anal. Calorim. 2013, [98] W. Zhang, Adv. Exp. Med. Biol. 2014, 811, 19.
111, 9. [99] X. Duan, Y. Li, Small 2013, 9, 1521.
[65] N. A. Frey, S. Peng, K. Cheng, S. Sun, Chem. Soc. Rev. 2009, 38, [100] K. H. Kernstine, W. Stanford, B. F. Mullan, N. P. Rossi,
2532. B. H. Thompson, D. L. Bushnell, K. A. McLaughlin, J. A. Kern,
[66] A. P. Herrera, L. Polo-Corrales, E. Chavez, J. Cabarcas-Bolivar, Ann. Thorac. Surg. 1999, 68, 1022.
O. N. C. Uwakweh, C. Rinaldi, J. Magn. Magn. Mater. 2013, 328, 41. [101] P. Reimer, T. Balzer, Eur. Radiol. 2003, 13, 1266.
[67] J. Li, X. Shi, M. Shen, Part. Part. Syst. Charact. 2014, 31, 1223. [102] F. M. Kievit, O. Veiseh, N. Bhattarai, C. Fang, J. W. Gunn, D. Lee,
[68] Y. Zhang, Y. Liu, C. Fei, Z. Yang, Z. Lu, R. Xiong, D. Yin, J. Shi, J. R. G. Ellenbogen, J. M. Olson, M. Zhang, Adv. Funct. Mater. 2009,
Appl. Phys. 2010, 108, 084312. 19, 2244.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (29 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[103] J. Xie, J. Wang, G. Niu, J. Huang, K. Chen, X. Li, X. Chen, Chem. [134] O. Zvarec, S. Purushotham, A. Masic, R. V. Ramanujan,
Commun. 2010, 46, 433. A. Miserez, Langmuir 2013, 29, 10899.
[104] H. Lee, E. Lee, D. K. Kim, N. K. Jang, Y. Y. Jeong, S. Jon, J. Am. [135] H. Yabu, H. Ohshima, Y. Saito, ACS Appl. Mater. Interfaces 2014,
Chem. Soc. 2006, 128, 7383. 6, 18122.
[105] A. Moore, E. Marecos, A. Bogdanov Jr., R. Weissleder, Radiology [136] Q. Li, D. G. Barrett, P. B. Messersmith, N. Holten-Andersen, ACS
2000, 214, 568. Nano 2016, 10, 1317.
[106] J. F. Schultz, J. D. Bell, R. M. Goldstein, J. A. Kuhn, T. M. McCarty, [137] B. Jorge, World Population Ageing, (Ed: J. Bravo), United Nations,
Ann. Surg. Oncol. 1999, 6, 691. NY 2015.
[107] M. G. Harisinghani, J. Barentsz, P. F. Hahn, W. M. Deserno, [138] R. W. Y. Habash, R. Bansal, D. Krewski, H. T. Alhafid, Crit. Rev.
S. Tabatabaei, C. H. Van de Kaa, J. De la Rosette, R. Weissleder, Biomed. Eng. 2007, 35, 37.
N. Engl. J. Med. 2003, 348, 2491. [139] R. W. Y. Habash, R. Bansal, D. Krewski, H. T. Alhafid, Crit. Rev.
[108] S. K. Mouli, L. C. Zhao, R. A. Omary, C. S. Thaxton, Nat. Rev. Urol. Biomed. Eng. 2006, 34, 491.
2010, 7, 84. [140] T. Koltai, Onco. Targets Ther. 2016, 9, 6343.
[109] H. Chen, L. Wang, J. Yeh, X. Wu, Z. Cao, Y. A. Wang, M. Zhang, [141] X. L. Liu, C. T. Ng, P. Chandrasekharan, H. T. Yang, L. Y. Zhao,
L. Yang, H. Mao, Biomaterials 2010, 31, 5397. E. Peng, Y. B. Lv, W. Xiao, J. Fang, J. B. Yi, H. Zhang, K. H. Chuang,
[110] G. Liu, J. Gao, H. Ai, X. Chen, Small 2013, 9, 1533. B. H. Bay, J. Ding, H. M. Fan, Adv. Healthcare Mater. 2016, 5, 2092.
[111] K. R. Di Bona, Y. Xu, P. A. Ramirez, J. DeLaine, C. Parker, Y. Bao, [142] E. Y. K. Ng, S. D. Kumar, Biomed. Eng. Online 2017, 16, 1.
J. F. Rasco, Reprod. Toxicol. 2014, 50, 36. [143] Q. A. Pankhurst, J. Connolly, S. K. Jones, J. Dobson, J. Phys. D:
[112] E. Blanco, H. Shen, M. Ferrari, Nat. Biotechnol. 2015, 33, 941. Appl. Phys. 2003, 36, 167.
[113] J. Huang, L. Wang, R. Lin, A. Y. Wang, L. Yang, M. Kuang, W. Qian, [144] Q. A. Pankhurst, N. T. K. Thanh, S. K. Jones, J. Dobson, J. Phys. D:
H. Mao, ACS Appl. Mater. Interfaces 2013, 5, 4632. Appl. Phys. 2009, 42, 224001.
[114] M. Mahmoudi, S. Laurent, M. A. Shokrgozar, M. Hosseinkhani, [145] G. F. Goya, V. Grazú, M. R. Ibarra, Curr. Nanosci. 2008, 4, 1.
ACS Nano 2011, 5, 7263. [146] J. Rivas, M. Bañobre-López, Y. Piñeiro-Redondo, B. Rivas,
[115] R. Weissleder, D. D. Stark, B. L. Engelstad, B. R. Bacon, M. A. López-Quintela, J. Magn. Magn. Mater. 2012, 324, 3499.
C. C. Compton, D. L. White, P. Jacobs, J. Lewis, Am. J. Roentgenol. [147] M. Bañobre-López, A. Teijeiro, J. Rivas, Rep. Pract. Oncol. Radio-
1989, 152, 167. ther. 2013, 18, 397.
[116] Y. Chen, L. Gong, N. Gao, J. Liao, J. Sun, Y. Wang, L. Wang, P. Zhu, [148] C. S. S. R. Kumar, F. Mohammad, Adv. Drug Delivery Rev. 2011, 63,
Q. Fan, Y. A. Wang, W. Zeng, H. Mao, L. Yang, F. Gao, Int. J. 789.
Nanomed. 2015, 10, 6689. [149] M. Bañobre-López, Y. Piñeiro, M. A. López Quintela, J. Rivas,
[117] M. H. Schwenk, Pharmacotherapy 2010, 30, 70. in Handbook of Nanomaterials Properties, (Eds: B. Bhushan),
[118] D. Hetzel, W. Strauss, K. Bernard, Z. Li, A. Urboniene, L. F. Allen, Springer-Verlag, Berlin 2014, pp. 457–493.
Am. J. Hematol. 2014, 89, 646. [150] G. Vallejo-Fernandez, O. Whear, A. G. Roca, S. Hussain, J. Timmis,
[119] M. Harisinghani, R. W. Ross, A. R. Guimaraes, R. Weissleder, V. Patel, K. O’Grady, J. Phys. D: Appl. Phys. 2013, 46, 312001.
Neoplasia 2007, 9, 1160. [151] A. G. Roca, B. Wiese, J. Timmis, G. Vallejo-Fernandez, K. O’Grady,
[120] L. Zhu, Z. Zhou, H. Mao, L. Yang, Nanomedicine 2017, 12, 73. IEEE Trans. Magn. 2012, 48, 9464.
[121] O. L. Gobbo, K. Sjaastad, M. W. Radomski, Y. Volkov, [152] E. C. Abenojar, S. Wickramasinghe, J. Bas-Concepcion,
A. Prina-Mello, Theranostics 2015, 5, 1249. A. C. S. Samia, Prog. Nat. Sci.: Mater. Int. 2016, 26, 440.
[122] A. Kumar, P. K. Jena, S. Behera, R. F. Lockey, S. Mohapatra, [153] B. Mehdaoui, A. Meffre, L. Lacroix, J. Carrey, S. Lachaize,
S. Mohapatra, Nanomedicine 2010, 6, e64. M. Respaud, M. Gougeon, B. Chaudret, B. Mehdaoui, A. Meffre,
[123] H. Ejima, J. J. Richardson, K. Liang, J. P. Best, M. P. Van L. Lacroix, J. Carrey, S. Lachaize, M. Respaud, J. Appl. Phys. 2010,
Koeverden, G. K. Such, J. Cui, F. Caruso, Science 2013, 341, 154. 107, 09A324.
[124] A. M. Spokoyny, D. Kim, A. Sumrein, C. A. Mirkin, Chem. Soc. Rev. [154] R. Hergt, W. Andr, C. G. Ambly, I. Hilger, W. A. Kaiser, U. Richter,
2009, 38, 1218. H.-g. Schmidt, IEEE Trans. Magn. 1998, 34, 3745.
[125] M. J. Harrington, A. Masic, N. Holten-Andersen, J. H. Waite, [155] R. Hergt, S. Dutz, M. Robert, M. Zeisberger, J. Phys.: Condens.
P. Fratzl, Science 2010, 328, 216. Matter 2006, 18, S2919.
[126] J. Guo, Y. Ping, H. Ejima, K. Alt, M. Meissner, J. J. Richardson, [156] R. E. Rosensweig, J. Magn. Magn. Mater. 2002, 252, 370.
Y. Yan, K. Peter, D. Von Elverfeldt, C. E. Hagemeyer, F. Caruso, [157] W. Wu, Q. He, C. Jiang, Nanoscale Res. Lett. 2008, 3, 397.
Angew. Chem., Int. Ed. 2014, 53, 5546. [158] M. K. Yu, J. Park, S. Jon, Theranostics 2012, 2, 3.
[127] Y. Ju, J. Cui, M. Müllner, T. Suma, M. Hu, F. Caruso, Biomacromol- [159] K. Kekalo, I. Baker, R. Meyers, J. Shyong, Nano LIFE 2015, 5, 1.
ecules 2015, 16, 807. [160] F. Shubitidze, K. Kekalo, R. Stigliano, I. Baker, J. Appl. Phys. 2015,
[128] Y. Ping, J. Guo, H. Ejima, X. Chen, J. J. Richardson, H. Sun, 117, 094302.
F. Caruso, Small 2015, 11, 2032. [161] C. Blanco-Andujar, D. Ortega, P. Southern, Q. A. Pankhurst,
[129] J. H. Park, K. Kim, J. Lee, J. Y. Choi, D. Hong, S. H. Yang, N. T. K. Thanh, Nanoscale 2015, 7, 1768.
F. Caruso, Y. Lee, I. S. Choi, Angew. Chem., Int. Ed. 2014, 53, [162] F. L. Deepak, M. Bañobre-lópez, E. Carbo-argibay, M. F. Guimarães
12420. Cerqueira, Y. Piñeiro-redondo, J. Rivas, C. M. Thompson,
[130] W. Li, W. Bing, S. Huang, J. Ren, X. Qu, Adv. Funct. Mater. 2015, S. Kamali, C. Rodriguez-abreu, K. Kovnir, Y. V. Kolen’ko, J. Phys.
25, 3775. Chem. C 2015, 119, 11947.
[131] J. Shi, Y. Jiang, X. Wang, H. Wu, D. Yang, F. Pan, Y. Su, Z. Jiang, [163] Y. V. Kolen’ko, M. Bañobre-López, C. Rodríguez-Abreu, E. Carbo-
Chem. Soc. Rev. 2014, 43, 5192. Argibay, A. Sailsman, Y. Piñeiro-Redondo, M. F. Cerqueira,
[132] M. A. Rahim, M. Björnmalm, T. Suma, M. Faria, Y. Ju, K. Kempe, D. Y. Petrovykh, K. Kovnir, O. I. Lebedev, J. Rivas, J. Phys. Chem. C
M. Müllner, H. Ejima, A. D. Stickland, F. Caruso, Angew. Chem., 2014, 118, 8691.
Int. Ed. 2016, 55, 13803. [164] P. Guardia, R. Di Corato, L. Lartigue, C. Wilhelm, A. Espinosa,
[133] K. V. Korpany, D. D. Majewski, C. T. Chiu, S. N. Cross, A. S. Blum, M. Garcia-hernandez, F. Gazeau, L. Manna, T. Pellegrino, ACS
Langmuir 2017, 33, 3000. Nano 2012, 6, 3080.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (30 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[165] R. Hergt, S. Dutz, R. Michael, J. Phys.: Condens. Matter 2008, 20, [191] P. Hugounenq, M. Levy, D. Alloyeau, E. Dubois, C. Ricolleau,
385214. C. Wilhelm, F. Gazeau, R. Bazzi, J. Phys. Chem. C 2012, 116, 15702.
[166] M. Gonzales-weimuller, M. Zeisberger, K. M. Krishnan, J. Magn. [192] A. Cervadoro, M. Cho, J. Key, C. Cooper, C. Stigliano, S. Aryal,
Magn. Mater. 2009, 321, 1947. A. Brazdeikis, J. F. Leary, P. Decuzzi, ACS Appl. Mater. Interfaces
[167] D. Bonvin, A. Arakcheeva, A. Millán, R. Piñol, H. Hofmann, 2014, 6, 12939.
M. M. Ebersold, RSC Adv. 2017, 7, 13159. [193] J. Lee-h, J.-t. Jang, J.-s. Choi, S. H. Moon, S.-h. Noh, J.-w. Kim,
[168] S. L. Saville, B. Qi, J. Baker, R. Stone, R. E. Camley, K. L. Livesey, J.-G. Kim, I.-S. Kim, I. K. Park, J. Cheon, Nat. Nanotechnol. 2011,
L. Ye, T. M. Crawford, O T. Mefford, J. Colloid Interface Sci. 2014, 6, 418.
424, 141. [194] A. Me, B. Mehdaoui, V. Kelsen, P. F. Fazzini, J. Carrey, S. Lachaize,
[169] Z. Nemati, J. Alonso, L. M. Martinez, H. Khurshid, E. Garaio, M. Respaud, B. Chaudret, Nano Lett. 2012, 12, 4722.
J. A. Garcia, M. H. Phan, H. Srikanth, J. Phys. Chem. C 2016, 120, [195] E. Mazario, N. Mene, P. Herrasti, M. Can, V. Connord, J. Carrey,
8370. J. Phys. Chem. C 2013, 117, 11405.
[170] Y. Piñeiro-Redondo, M. Bañobre-López, I. Pardiñas-Blanco, [196] X. Lasheras, M. Insausti, I. G. De Muro, E. Garaio, F. Plazaola,
G. Goya, M. A. López-Quintela, J. Rivas, Nanoscale Res. Lett. 2011, M. Moros, L. De Matteis, M. De Fuente, L. Lezama, J. Phys. Chem.
6, 383. C 2016, 120, 3492.
[171] S. V. Jadhav, S.-h. Lee, D. S. Nikam, R. A. Bohara, S. H. Pawar, [197] Y. Qu, J. Li, J. Ren, J. Leng, C. Lin, D. Shi, ACS Appl. Mater. Inter-
Y.-S. Yu, New J. Chem. 2017, 41, 1598. faces 2014, 6, 16867.
[172] D. Cabrera, J. Camarero, D. Ortega, F. J. Teran, J. Nanopart. Res. [198] I. Sharifi, H. Shokrollahi, S. Amiri, J. Magn. Magn. Mater. 2012,
2015, 17, 1. 324, 903.
[173] L. C. Branquinho, M. S. Carria, A. S. Costa, N. Zufelato, [199] R.-J. Chung, H.-Y. Wang, K.-T. Wu, J. Med. Biol. Eng. 2013, 34, 251.
M. H. Sousa, R. Miotto, R. Ivkov, A. F. Bakuzis, Sci. Rep. 2013, 3, 1. [200] T. Kondo, K. Mori, M. Hachisu, T. Yamazaki, D. Okamoto,
[174] R. Grillo, J. Gallo, D. G. Stroppa, E. Carbó-Argibay, R. Lima, M. Watanabe, K. Gonda, H. Tada, Y. Hamada, M. Takano,
L. F. Fraceto, M. Bañobre-López, ACS Appl. Mater. Interfaces 2016, N. Ohuchi, Y. Ichiyanagi, J. Appl. Phys. 2015, 117, 17D157.
8, 25777. [201] C. Martinez-boubeta, K. Simeonidis, D. Serantes,
[175] M. Palihawadana-arachchige, H. Nemala, V. M. Naik, R. Naik, I. Conde-leborán, I. Kazakis, G. Stefanou, L. Peña, R. Galceran,
M. Palihawadana-arachchige, H. Nemala, V. M. Naik, J. Appl. Phys. L. Balcells, C. Monty, D. Baldomir, M. Mitrakas, M. Angelakeris,
2017, 121, 023901. Adv. Funct. Mater. 2012, 22, 3737.
[176] G. Hemery, A. C. Keyes Jr., E. Garaio, I. Rodrigo, J. A. Garcia, [202] E. Kita, T. Oda, T. Kayano, S. Sato, M. Minagawa, H. Yanagihara,
F. Plazaola, E. Garanger, O. Sandre, Inorg. Chem. 2017, 56, 8232. M. Kishimono, C. Mitsumata, S. Hashimoto, K. Yamada,
[177] I. Andreu, E. Natividad, L. Solozábal, O. Roubeau, ACS Nano N. Ohkohchi, J. Phys. D: Appl. Phys. 2010, 43, 474011.
2015, 9, 1408. [203] D. Yoo, J.-H. Lee, T. H. Shin, J. Cheon, Acc. Chem. Res. 2011, 44,
[178] D. F. Coral, P. Mendoza Zélis, M. Marciello, M. del Puerto 863.
Morales, A. Craievich, F. H. Sánchez, M. B. Fernández van Raap, [204] J. Kolosnjaj-tabi, R. Di Corato, L. Lartigue, I. Marangon,
Langmuir 2016, 32, 1201. P. Guardia, A. K. A. Silva, N. Luciani, O. Clément, P. Flaud, J.
[179] C. Guibert, V. Dupuis, V. Peyre, J. Fresnais, J. Phys. Chem. C 2015, V. Singh, P. Decuzzi, T. Pellegrino, C. Wilhelm, F. Gazeau, ACS
119, 28148. Nano 2014, 4268.
[180] I. Conde-Leboran, D. Baldomir, C. Martinez-Boubeta, [205] R. Das, J. Alonso, Z. N. Porshokouh, V. Kalappattil, D. Torres,
O. Chubykalo-Fesenko, M. del Puerto Morales, G. Salas, M.-h. Phan, E. Garaio, A. Jose, J. Luis, S. Llamazares, H. Srikanth,
D. Cabrera, J. Camarero, F. J. Teran, D. Serantes, J. Phys. Chem. C J. Phys. Chem. C 2016, 120, 10086.
2015, 119, 15698. [206] S. H. Moon, S.-h. Noh, J.-h. Lee, T.-h. Shin, Y. Lim, J. Cheon, Nano
[181] R. R. Wildeboer, P. Southern, Q. A. Pankhurst, J. Phys. D: Appl. Lett. 2017, 17, 800.
Phys. 2014, 47, 495003. [207] D. J. Carnevale, M. Shatruk, G. F. Strouse, Chem. Mater. 2016, 28,
[182] M. Filippousi, M. Angelakeris, M. Katsikini, E. Paloura, 5480.
I. Efthimiopoulos, Y. Wang, D. Zamboulis, G. Van Tendeloo, J. [208] F. Liu, J. Zhu, W. Yang, Y. Dong, Y. Hou, C. Zhang, H. Yin, Angew.
Phys. Chem. C 2014, 118, 16209. Chem., Int. Ed. Engl. 2014, 53, 2176.
[183] M. Cano, R. Núñez-lozano, R. Lumbreras, V. González-rodríguez, [209] M. Jeun, Y. J. Kim, K. H. Park, S. H. Paek, S. Bae, J. Nanosci. Nano-
A. Delgado-garcía, J. M. Jiménez-hoyuela, G. De Cueva-méndez, technol. 2013, 13, 5719.
Nanoscale 2017, 9, 812. [210] D. Soukup, S. Moise, E. Céspedes, J. Dobson, N. D. Telling, ACS
[184] F. Sonvico, C. Dubernet, D. Jaillard, J. Degrouard, J. Hoebeke, Nano 2015, 9, 231.
E. Duguet, P. Colombo, P. Couvreur, Bioconjugate Chem. 2005, 16, [211] M. Torres-lugo, C. Rinaldi, M. Creixell, A. C. Boh, ACS Nano 2011,
1181. 7124.
[185] D. Fourmy, J. Carrey, Nanomedicine 2015, 10, 893. [212] M. Bañobre-López, D. Rodrigues, B. Espiña, J. Azeredo, J. Rivas,
[186] T. Sadhukha, T. S. Wiedmann, J. Panyam, Biomaterials 2013, 34, Biofouling 2013, 29, 1225.
5163. [213] B. Sanz, M. P. Calatayud, T. E. Torres, M. L. Fanarraga,
[187] M. Colombo, S. Carregal-Romero, M. F. Casula, M. del Puerto M. R. Ibarra, G. F. Goya, Biomaterials 2017, 114, 62.
Morales, I. B. Böhm, J. T. Heverhagen, D. Prosperi, W. J. Parak, [214] H. L. Rodríguez-Luccioni, M. Latorre-Esteves, J. Méndez-Vega,
Chem. Soc. Rev. 2012, 41, 4306. M. Torres-Lugo, Int. J. Nanomed. 2011, 6, 373.
[188] K. H. Bae, M. Park, M. J. Do, N. Lee, J. H. Ryu, G. W. Kim, C. Kim, [215] M. E. de Sousa, A. Carrea, P. Mendoza Zélis, D. Muraca,
T. G. Park, T. Hyeon, ACS Nano 2012, 5, 5266. O. Mykhaylyk, Y. E. Sosa, R. G. Goya, F. H. Sánchez, R. A. Dewey,
[189] I. S. Smolkova, N. E. Kazantseva, V. Babayan, J. Vilcakova, M. B. Fernández van Raap, J. Phys. Chem. C 2016, 120, 7339.
N. Pizurova, P. Saha, Cryst. Growth Des. 2017, 17, 2332. [216] A. Riedinger, P. Guardia, A. Curcio, M. A. Garcia, R. Cingolani,
[190] R. Das, N. Rinaldi-montes, J. Alonso, Z. Amghouz, E. Garaio, L. Manna, T. Pellegrino, Nano Lett. 2013, 13, 2399.
A. Garcia, P. Gorria, J. A. Blanco, M.-h. Phan, H. Srikanth, ACS [217] P. Liu, Y. Dong, P. Du, B. Mu, Ind. Eng. Chem. Res. 2012, 51,
Appl. Mater. Interfaces 2016. 3350.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (31 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[218] R. Piñol, C. D. S. Brites, R. Bustamante, A. Martínez, N. J. O. Silva, [246] P. Saint-Cricq, S. Deshayes, J. I. Zink, A. M. Kasko, Nanoscale
J. L. Murillo, R. Cases, J. Carrey, C. Estepa, C. Sosa, F. Palacio, 2015, 7, 13168.
L. D. Carlos, A. Millán, ACS Nano 2015, 3134. [247] T. T. T. N’Guyen, H. T. T. Duong, J. Basuki, V. Montembault,
[219] C. J. Sunderland, M. Steiert, J. E. Talmadge, A. M. Derfus, S. Pascual, C. Guibert, J. Fresnais, C. Boyer, M. R. Whittaker,
S. E. Barry, Drug Dev. Res. 2006, 67, 70. T. P. Davis, L. Fontaine, Angew. Chem., Int. Ed. Engl. 2013, 52,
[220] R. A. Bini, R. Fernando, C. Marques, F. J. Santos, J. A. Chaker, 14152.
M. Jafelicci Jr., J. Magn. Magn. Mater. 2012, 324, 534. [248] P. Saint-Cricq, S. Deshayes, J. I. Zink, A. M. Kasko, Nanoscale
[221] D. Singh, J. M. Mcmillan, A. V. Kabanov, M. Sokolsky-papkov, 2015, 13168.
H. E. Gendelman, Nanomedicine 2014, 9, 501. [249] S. Kossatz, J. Grandke, P. Couleaud, A. Latorre, A. Aires,
[222] S. B. Campbell, M. Patenaude, T. Hoare, Biomacromolecules 2013, K. Crosbie-staunton, R. Ludwig, H. Dähring, V. Ettelt, A. Lazaro-
14, 644. carrillo, M. Calero, M. Sader, J. Courty, Y. Volkov, A. Prina-mello,
[223] A. M. Hawkins, C. E. Bottom, Z. Liang, D. A. Puleo, J. Z. Hilt, Adv. A. Villanueva, Á. Somoza, A. L. Cortajarena, R. Miranda, I. Hilger,
Healthcare Mater. 2012, 2012, 96. Breast Cancer Res. 2015, 17, 1.
[224] T. Hoare, B. P. Timko, J. Santamaria, G. F. Goya, S. Irusta, S. Lau, [250] C. Sanson, O. Diou, J. Thévenot, E. Ibarboure, A. Soum, A. Brûlet,
C. F. Stefanescu, D. Lin, R. Langer, D. S. Kohane, Nano Lett. 2011, S. Miraux, E. ThiaudièreTan, S. Tan, A. Brisson, Â. V. Dupuis,
11, 1395. O. O. Sandre, S. Lecommandoux, ACS Nano 2011, 5, 1122.
[225] S. A. Rovers, R. Hoogenboom, F. Kemmere, J. T. F. Keurentjes, [251] K. Hayashi, M. Nakamura, H. Miki, S. Ozaki, M. Abe,
Soft Matter 2012, 8, 1623. T. Matsumoto, W. Sakamoto, T. Yogo, K. Ishimura, Theranostics
[226] A. E. Mengesha, R. J. Wydra, J. Z. Hilt, P. M. Bummer, Pharm. Res. 2014, 4, 834.
2013, 30, 3214. [252] N. D. Thorat, R. A. Bohara, M. Radz Noor, D. Dhamecha,
[227] M. Bañobre-López, Y. Piñeiro-Redondo, M. Sandri, A. Tampieri, T. Soulimane, S. A. M. Tofail, ACS Biomater. Sci. Eng. 2017, 3, 1332.
R. Di Santis, V. A. Dediu, J. Rivas, IEEE Trans. Magn. 2014, 50, [253] B. Clares, R. A. Biedma-Ortiz, E. Sáez-Fernández, J. C. Prados,
5400507. C. Melguizo, L. Cabeza, R. Ortiz, J. L. Arias, Eur. J. Pharm. Biop-
[228] S. K. Samal, M. Dash, T. Shelyakova, H. A. Declercq, harm. 2013, 85, 329.
M. Uhlarz, M. Bañobre-López, P. Dubruel, M. Cornelissen, [254] K. Hayashi, Y. Sato, W. Sakamoto, T. Yogo, ACS Biomater. Sci. Eng.
T. Herrmannsdörfer, J. Rivas, G. Padeletti, S. De Smedt, 2017, 3, 95.
K. Braeckmans, D. L. Kaplan, V. A. Dediu, ACS Appl. Mater. Inter- [255] A. Espinosa, R. Di Corato, J. Kolosnjaj-tabi, P. Flaud, T. Pellegrino,
faces 2015, 7, 6282. C. Wilhelm, ACS Nano 2016, 10, 2436.
[229] R. Costa, C. Ribeiro, A. C. Lopes, P. Martins, V. Sencadas, [256] J. Kolosnjaj-tabi, A. Espinosa, O. Cle, R. Di Corato, C. Wilhelm,
R. Soares, S. Lanceros-Mendez, J. Mater. Sci.: Mater. Med. 2013, A. K. A. Silva, C. Me, D. I. Corato, E. T. Al, ACS Nano 2015, 9, 2904.
24, 395. [257] W. Walther, U. Stein, Adv. Drug Delivery Rev. 2009, 61, 641.
[230] G. A. Koning, A. M. M. Eggermont, L. H. Lindner, T. L. M. Hagen, [258] P. T. Yin, B. P. Shah, K.-B. Lee, Small 2014, 10, 4106.
Pharm. Res. 2010, 27, 1750. [259] S. Toraya-Brown, M. R. Sheen, P. Zhang, L. Chen, J. R. Baird,
[231] B. Kneidl, M. Peller, G. Winter, L. H. Lindner, M. Hossann, Int. J. E. Demidenko, M. J. Turk, P. J. Hoopes, J. R. Conejo-garcia,
Nanomed. 2014, 9, 4387. S. Fiering, Nanomedicine 2014, 10, 1273.
[232] D.-h. Kim, E. A. Vitol, J. Liu, S. Balasubramanian, D. J. Gosztola, [260] R. Ludwig, et al., Int. J. Nanomed. 2017, 12, 1009.
E. E. Cohen, V. Novosad, E. A. Rozhkova, Langmuir 2013, 29, 7425. [261] B. Wang, J.-J. Yin, X. Zhou, I. Kurash, Z. Chai, Y. Zhao, W. Feng,
[233] J. Huang, Y. Li, A. Orza, Q. Lu, P. Guo, L. Wang, L. Yang, H. Mao, J. Phys. Chem. C 2013, 117, 383.
Adv. Funct. Mater. 2016, 26, 3818. [262] J. Puig, C. E. Hoppe, L. A. Fasce, Y. Piñeiro-Redondo,
[234] S. Carregal-Romero, P. Guardia, X. Yu, R. Hartmann, T. Pellegrino, M. Bañobre-López, M. A. López-Quintela, J. Rivas, R. J. J. Williams,
W. J. Parak, Nanoscale 2015, 7, 570. J. Phys. Chem. C 2012, 116, 13421.
[235] S. Jiang, A. A. Eltoukhy, K. T. Love, R. Langer, D. G. Anderson, [263] F. Assa, H. Jafarizadeh-Malmiri, H. Ajamein, H. Vaghari,
Nano Lett. 2013, 13, 1059. N. Anarjan, O. Ahmadi, A. Berenjian, Crit. Rev. Biotechnol. 2017,
[236] G. Béalle, R. Di Corato, J. Kolosnjaj-tabi, V. Dupuis, O. Clément, 37, 492.
F. Gazeau, C. Wilhelm, C. Ménager, Langmuir 2012, 28, 11834. [264] J. Barbosa, D. M. Correia, R. Gonçalves, C. Ribeiro, G. Botelho,
[237] K. Katagiri, Y. Imai, K. Koumoto, T. Kaiden, K. Kono, S. Aoshima, P. Martins, S. Lanceros-Mendez, Adv. Healthcare Mater. 2016, 5,
Small 2011, 7, 1683. 3027.
[238] S.-H. Hu, S.-Y. Chen, X. Gao, ACS Nano 2012, 6, 2558. [265] P. J. Cregg, K. Murphy, A. Mardinoglu, Appl. Math. Model. 2012,
[239] Y. Qu, J. Li, J. Ren, J. Leng, C. Lin, D. Shi, Nanoscale 2014, 6, 36, 1.
12408. [266] K. Hola, Z. Markova, G. Zoppellaro, J. Tucek, R. Zboril, Biotechnol.
[240] R. R. de Almeida, J. Gallo, A. C. C. da Silva, A. K. O. da Silva, Adv. 2015, 33, 1162.
O. D. L. Pessoa, T. Araújo, L. K. A. M. Leal, P. B. A. Fechine, [267] X. Mou, Z. Ali, S. Li, N. He, J. Nanosci. Nanotechnol. 2015, 15, 54.
M. Bañobre-López, N. M. P. S. Ricardo, J. Braz. Chem. Soc. 2017, [268] M. K. Jaiswal, M. Gogoi, H. Dev Sarma, R. Banerjee, D. Bahadur,
28, 1547. Biomater. Sci. 2014, 2, 370.
[241] N. Griffete, J. Fresnais, A. Espinosa, C. Wilhelm, A. Bée, [269] A. Bakandritsos, A. Papagiannopoulos, E. N. Anagnostou,
C. Ménager, Nanoscale 2015, 7, 18891. K. Avgoustakis, R. Zboril, S. Pispas, J. Tucek, V. Ryukhtin,
[242] A. K. Hauser, R. J. Wydra, N. A. Stocke, K. W. Anderson, Z. Hilt, J. N. Bouropoulos, A. Kolokithas-Ntoukas, T. A. Steriotis,
Controlled Release 2016, 219, 76. U. Keiderling, F. Winnefeld, Small 2012, 8, 2381.
[243] B. A. M. Derfus, G. Von Maltzahn, T. J. Harris, T. Duza, [270] L. A. Tai, P. J. Tsai, Y. C. Wang, Y. J. Wang, L. W. Lo, C. S. Yang,
K. S. Vecchio, E. Ruoslahti, S. N. Bhatia, Adv. Mater. 2007, 19, Nanotechnology 2009, 20.
3932. [271] M. Magro, R. Campos, D. Baratella, G. Lima, K. Holà, C. Divoky,
[244] M. Nair, R. Guduru, P. Liang, J. Hong, V. Sagar, S. Khizroev, Nat. R. Stollberger, O. Malina, C. Aparicio, G. Zoppellaro, R. Zbořil,
Commun. 2013, 4, 1707. F. Vianello, Chem. – Eur. J. 2014, 20, 11913.
[245] S. T. Meikle, Y. Piñeiro, M. Bañobre-López, J. Rivas, M. Santin, [272] M. Arruebo, R. Fernández-Pacheco, M. R. Ibarra, J. Santamaría,
Acta Biomater. 2016, 40, 235. Nano Today 2007, 2, 22.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (32 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[273] K. J. Widder, A. E. Senyei, D. G. Scarpelli, Proc. Soc. Exp. Biol. Med. [305] R. Brito-Pereira, C. Ribeiro, S. Lanceros-Mendez, P. Martins,
1978, 158, 141. Composites, Part B 2017, 120, 97.
[274] T. Kato, R. Nemoto, H. Mori, R. Abe, K. Unno, A. Goto, H. Murota, [306] S. Gil, J. M. Silva, J. F. Mano, ACS Biomater. Sci. Eng. 2015, 1,
M. Harada, M. Homma, Appl. Biochem. Biotechnol. 1984, 10, 199. 1016.
[275] K. J. Widder, R. M. Morris, G. A. Poore, D. P. Howard, A. E. Senyei, [307] Y. Li, G. Huang, X. Zhang, B. Li, Y. Chen, T. Lu, T. J. Lu, F. Xu, Adv.
Eur. J. Cancer Clin. Oncol. 1983, 19, 135. Funct. Mater. 2013, 23, 660.
[276] P. K. Gupta, C. T. Hung, N. S. Rao, J. Pharm. Sci. 1989, 78, 290. [308] N. Zhang, J. Lock, A. Sallee, H. Liu, ACS Appl. Mater. Interfaces
[277] U. O. Häfeli, S. M. Sweeney, B. A. Beresford, J. L. Humm, 2015, 7, 20987.
R. M. Macklis, Nucl. Med. Biol. 1995, 22, 147. [309] J. Meng, Y. Zhang, X. Qi, H. Kong, C. Wang, Z. Xu, S. Xie, N. Gu,
[278] A. Jătariu, C. Peptu, M. Popa, A. Indrei, Rev. Med.-Chir. Soc. Med. H. Xu, Nanoscale 2010, 2, 2565.
Nat. Iasi 2009, 113, 1160. [310] R. De Santis, A. Gloria, T. Russo, U. D’Amora, S. Zeppetelli,
[279] A. S. Lübbe, C. Bergemann, W. Huhnt, T. Fricke, H. Riess, L. Ambrosio, AIP Conf. Proc. 2010, 1255, 420.
J. W. Brock, D. Huhn, Cancer Res. 1996, 56, 4694. [311] U. D’Amora, T. Russo, A. Gloria, V. Rivieccio, V. D’Antò, G. Negri,
[280] F. Wiekhorst, C. Seliger, R. Jurgons, U. Steinhoff, D. Eberbeckt, L. Ambrosio, R. De Santis, Bioactive Mater. Med. 2017, 2, 138.
L. Trahms, C. Alexiou, J. Nanosci. Nanotechnol. 2006, 6, 3222. [312] Y. Sapir-Lekhovitser, M. Y. Rotenberg, J. Jopp, G. Friedman,
[281] A. Nacev, C. Beni, O. Bruno, B. Shapiro, J. Magn. Magn. Mater. B. Polyak, S. Cohen, Nanoscale 2016, 8, 3386.
2011, 323, 651. [313] H. Y. Xu, N. Gu, Front. Mater. Sci. 2014, 8, 20.
[282] D. B. Pacardo, F. S. Ligler, Z. Gu, Nanoscale 2015, 7, 3381. [314] A. Ortolani, M. Bianchi, M. Mosca, S. Caravelli, M. Fuiano,
[283] L. Yang, H. Liu, Powder Technol. 2013, 240, 54. M. Marcacci, A. Russo, Joints 2016, 4, 228.
[284] S. Bhatnagar, V. V. K. Venuganti, J. Nanosci. Nanotechnol. 2015, 15, [315] C. A. Cezar, E. T. Roche, H. H. Vandenburgh, G. N. Duda,
1925. C. J. Walsh, D. J. Mooney, Proc. Natl. Acad. Sci. USA 2016, 113,
[285] R. Tietze, J. Zaloga, H. Unterweger, S. Lyer, R. P. Friedrich, 1534.
C. Janko, M. Pöttler, S. Dürr, C. Alexiou, Biochem. Biophys. Res. [316] Z. Liu, L. Huang, L. Liu, B. Luo, M. Liang, Z. Sun, S. Zhu, X. Quan,
Commun. 2015, 468, 463. Y. Yang, T. Ma, J. Huang, Z. Luo, Int. J. Nanomed. 2014, 10, 43.
[286] C. A. Cezar, S. M. Kennedy, M. Mehta, J. C. Weaver, L. Gu, [317] Y. Sapir, B. Polyak, S. Cohen, Nanotechnology 2014, 25.
H. Vandenburgh, D. J. Mooney, Adv. Healthcare Mater. 2014, 3, [318] J. Meng, B. Xiao, Y. Zhang, J. Liu, H. Xue, J. Lei, H. Kong,
1869. Y. Huang, Z. Jin, N. Gu, H. Xu, Sci. Rep. 2013, 3, 2278.
[287] N. S. Satarkar, J. Z. Hilt, J. Controlled Release 2008, 130, 246. [319] S. Panseri, A. Russo, G. Giavaresi, M. Sartori, F. Veronesi, M. Fini,
[288] H. Oliveira, E. Pérez-Andrés, J. Thevenot, O. Sandre, E. Berra, D. M. Salter, A. Ortolani, A. Strazzari, A. Visani, C. Dionigi,
S. Lecommandoux, J. Controlled Release 2013, 169, 165. N. Bock, M. Sandri, A. Tampieri, M. Marcacci, J. Biomed. Mater.
[289] M.-H. K. Jordan A, P. Wust, M. Johannsen, in Nanotechnology Res., Part A 2012, 100, 2655.
for Cancer Therapy, (Ed.: M. M. Amiji), CRC Press, Weinheim, [320] X. B. Zeng, H. Hu, L. Q. Xie, F. Lan, W. Jiang, Y. Wu, Z. W. Gu, Int.
Germany 2017. J. Nanomed. 2012, 7, 3365.
[290] X. Hua, Q. Yang, Z. Dong, J. Zhang, W. Zhang, Q. Wang, S. Tan, [321] N. Bock, A. Riminucci, C. Dionigi, A. Russo, A. Tampieri, E. Landi,
H. D. Smyth, Drug Delivery 2017, 24, 511. V. A. Goranov, M. Marcacci, V. Dediu, Acta Biomater. 2010, 6, 786.
[291] Z. Tang, Y. Gao, D. Li, S. Zhou, Adv. Healthcare Mater. 2017, 6, [322] F. Heidari, M. E. Bahrololoom, D. Vashaee, L. Tayebi, Ceram. Int.
1600919. 2015, 41, 3094.
[292] M. Karimi, A. Ghasemi, P. Sahandi Zangabad, R. Rahighi, [323] P. Periyathambi, W. S. Vedakumari, S. Kumar Baskar, S. Bojja,
S. M. Moosavi Basri, H. Mirshekari, M. Amiri, Z. Shafaei Pishabad, T. P. Sastry, Mater. Lett. 2015, 139, 108.
A. Aslani, M. Bozorgomid, D. Ghosh, A. Beyzavi, A. Vaseghi, [324] M. Ishii, R. Shibata, Y. Shimizu, T. Yamamoto, K. Kondo,
A. R. Aref, L. Haghani, S. Bahrami, M. R. Hamblin, Chem. Soc. Rev. Y. Inoue, N. Ouchi, T. Tanigawa, N. Kanemura, A. Ito, H. Honda,
2016, 45, 1457. T. Murohara, Int. J. Cardiol. 2014, 175, 545.
[293] L. J. Santos, R. L. Reis, M. E. Gomes, Trends Biotechnol. 2015, 33, [325] J. J. Kim, R. K. Singh, S. J. Seo, T. H. Kim, J. H. Kim, E. J. Lee,
471. H. W. Kim, RSC Adv. 2014, 4, 17325.
[294] C. J. Mortimer, C. J. Wright, Biotechnol. J. 2017, 12, 1600693. [326] A. I. Gonçalves, M. T. Rodrigues, P. P. Carvalho, M. Bañobre-López,
[295] R. Gonçalves, P. Martins, X. Moya, M. Ghidini, V. Sencadas, E. Paz, P. Freitas, M. E. Gomes, Adv. Healthcare Mater. 2016,
G. Botelho, N. D. Mathur, S. Lanceros-Mendez, Nanoscale 2015, 5, 213.
7, 8058. [327] S. Aliramaji, A. Zamanian, M. Mozafari, Mater. Sci. Eng., C 2017,
[296] E. A. Lee, H. Yim, J. Heo, H. Kim, G. Jung, N. S. Hwang, Arch. 70, 736.
Pharm. Sci. Res. 2014, 37, 120. [328] J. K. Xu, F. F. Zhang, J. J. Sun, J. Sheng, F. Wang, M. Sun,
[297] L. Ferreira, J. M. Karp, L. Nobre, R. Langer, Cell Stem Cell 2008, 3, Molecules 2014, 19, 21506.
136. [329] J. Parssinen, H. Hammarén, R. Rahikainen, V. Sencadas,
[298] N. J. Sniadecki, Endocrinology 2010, 151, 451. C. Ribeiro, S. Vanhatupa, S. Miettinen, S. Lanceros-Méndez,
[299] N. J. Sniadecki, A. Anguelouch, M. T. Yang, C. M. Lamb, Z. Liu, V. P. Hytönen, J. Biomed. Mater. Res., Part A 2015, 103, 919.
S. B. Kirschner, Y. Liu, D. H. Reich, C. S. Chen, Proc. Natl. Acad. [330] J. Nunes-Pereira, S. Ribeiro, C. Ribeiro, C. J. Gombek, F. M. Gama,
Sci. USA 2007, 104, 14553. A. C. Gomes, D. A. Patterson, S. Lanceros-Méndez, Polym. Test.
[300] N. J. Sniadecki, C. M. Lamb, Y. Liu, C. S. Chen, D. H. Reich, 2015, 44, 234.
Rev. Sci. Instrum. 2008, 79, 044302. [331] D. M. Correia, V. Sencadas, C. Ribeiro, P. M. Martins, P. Martins,
[301] P. Tseng, J. W. Judy, D. Di Carlo, Nat. Methods 2012, 9, 1113. F. M. Gama, G. Botelho, S. Lanceros-Méndez, J. Colloid Interface
[302] B. Hu, A. J. El Haj, J. Dobson, Int. J. Mol. Sci. 2013, 14, 19276. Sci. 2016, 476, 79.
[303] L. Bonnemay, C. Hoffmann, Z. Gueroui, Wiley Interdiscip. Rev.: [332] Y. Wei, X. Zhang, Y. Song, B. Han, X. Hu, X. Wang, Y. Lin, X. Deng,
Nanomed. Nanobiotechnol. 2015, 7, 342. Biomed. Mater. 2011, 6, 055008.
[304] C. Ribeiro, V. Correia, P. Martins, F. M. Gama, S. Lanceros- [333] P. Martins, Y. V. Kolen’Ko, J. Rivas, S. Lanceros-Mendez, ACS Appl.
Mendez, Colloids Surf., B 2016, 140, 430. Mater. Interfaces 2015, 7, 15017.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (33 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[334] R. Gonçalves, P. Martins, D. M. Correia, V. Sencadas, J. L. Vilas, [367] J. T. Jang, H. Nah, J. H. Lee, S. H. Moon, M. G. Kim, J. Cheon,
L. M. León, G. Botelho, S. Lanceros-Méndez, RSC Adv. 2015. 5, Angew. Chem., Int. Ed. 2009, 48, 1234.
35852. [368] J. S. Choi, J. H. Lee, T. H. Shin, H. T. Song, E. Y. Kim, J. Cheon,
[335] F. J. O’Brien, Mater. Today 2011, 14, 88. J. Am. Chem. Soc. 2010, 132, 11015.
[336] D. Singh, J. M. McMillan, A. V. Kabanov, M. Sokolsky-Papkov, [369] J. Yang, C. H. Lee, H. J. Ko, J. S. Suh, H. G. Yoon, K. Lee,
H. E. Gendelman, Nanomedicine 2014, 9, 501. Y. M. Huh, S. Haam, Angew. Chem., Int. Ed. 2007, 46, 8836.
[337] K. Shimizu, A. Ito, T. Yoshida, Y. Yamada, M. Ueda, H. Honda, [370] M. K. Yu, Y. Y. Jeong, J. Park, S. Park, J. W. Kim, J. J. Min, K. Kim,
J. Biomed. Mater. Res., Part B 2007, 82, 471. S. Jon, Angew. Chem., Int. Ed. 2008, 47, 5362.
[338] H. Ito, R. Kato, K. Ino, H. Honda, J. Biosci. Bioeng. 2010, 109, 182. [371] P. C. Liang, Y. C. Chen, C. F. Chiang, L. R. Mo, S. Y. Wei,
[339] A. Ito, R. Kato, K. Ino, H. Honda, Tissue Eng., Part C 2009, 15, 413. W. Y. Hsieh, W. L. Lin, Int. J. Nanomed. 2016, 11, 2021.
[340] K. Shimizu, A. Ito, H. Honda, J. Biosci. Bioeng. 2007, 104, 171. [372] R. Mangaiyarkarasi, S. Chinnathambi, S. Karthikeyan, P. Aruna,
[341] Y. Yamamoto, A. Ito, H. Fujita, E. Nagamori, Y. Kawabe, S. Ganesan, J. Magn. Magn. Mater. 2016, 399, 207.
M. Kamihira, Tissue Eng., Part A 2011, 17, 107. [373] N. Kohler, C. Sun, A. Fichtenholtz, J. Gunn, C. Fang, M. Zhang,
[342] A. Ito, H. Akiyama, Y. Yamamoto, Y. Kawabe, M. Kamihira, in Small 2006, 2, 785.
20th Anniversary MHS 2009 and Micro-Nano Global COE—2009 [374] Y. F. Rao, W. Chen, X. G. Liang, Y. Z. Huang, J. Miao, L. Liu, Y. Lou,
Int. Symp. on Micro–NanoMechatronics and Human Science, X. G. Zhang, B. Wang, R. K. Tang, Z. Chen, X. Y. Lu, Small 2015,
IEEE, Kyushu University, Japan 2009. https://doi.org/10.1109/ 11, 239.
MHS.2009.5352027. [375] D. Kami, S. Takeda, Y. Itakura, S. Gojo, M. Watanabe, M. Toyoda,
[343] M. Sato, A. Ito, H. Akiyama, Y. Kawabe, M. Kamihira, Tissue Eng., Int. J. Mol. Sci. 2011, 12, 3705.
Part A 2013, 19, 307. [376] S. C. McBain, H. H. P. Yiu, A. El Haj, J. Dobson, J. Mater. Chem.
[344] H. Fujita, K. Shimizu, Y. Yamamoto, A. Ito, M. Kamihira, 2007, 17, 2561.
E. Nagamori, J. Tissue Eng. Regener. Med. 2010, 4, 437. [377] Y. Wang, H. Cui, K. Li, C. Sun, W. Du, J. Cui, X. Zhao, W. Chen,
[345] A. Ito, M. Hayashida, H. Honda, K. I. Hata, H. Kagami, M. Ueda, PLoS One 2014, 9, e106612.
T. Kobayashi, Tissue Eng. 2004, 10, 873. [378] S. Jiang, A. A. Eltoukhy, K. T. Love, R. Langer, D. G. Anderson,
[346] K. Shimizu, A. Ito, M. Arinobe, Y. Murase, Y. Iwata, Y. Narita, Nano Lett. 2013, 13, 1059.
H. Kagami, M. Ueda, H. Honda, J. Biosci. Bioeng. 2007, 103, 472. [379] Z. Medarova, W. Pham, C. Farrar, V. Petkova, A. Moore, Nat. Med.
[347] A. Ito, K. Ino, M. Hayashida, T. Kobayashi, H. Matsunuma, 2007, 13, 372.
H. Kagami, M. Ueda, H. Honda, Tissue Eng. 2005, 11, 1553. [380] J. H. Lee, K. Lee, S. H. Moon, Y. Lee, T. G. Park, J. Cheon, Angew.
[348] V. Mironov, V. Kasyanov, R. R. Markwald, Trends Biotechnol. 2008, Chem., Int. Ed. 2009, 48, 4174.
26, 338. [381] Z. Sun, X. Song, X. Li, T. Su, S. Qi, R. Qiao, F. Wang, Y. Huan,
[349] K. Shimizu, A. Ito, M. Arinobe, Y. Murase, Y. Iwata, Y. Narita, W. Yang, J. Wang, Y. Nie, K. Wu, M. Gao, F. Cao, Nanoscale 2014,
H. Kagami, M. Ueda, H. Honda, Biotechnol. Bioeng. 2007, 6, 14343.
96, 803. [382] U. M. Mahajan, S. Teller, M. Sendler, R. Palankar, C. Van Den
[350] A. Ito, Y. Takizawa, H. Honda, K. I. Hata, H. Kagami, M. Ueda, Brandt, T. Schwaiger, J. P. Kühn, S. Ribback, G. Glöckl, M. Evert,
T. Kobayashi, Tissue Eng. 2004, 10, 833. W. Weiss, N. Hosten, F. Dombrowski, M. Delcea, F. U. Weiss,
[351] A. Ito, E. Hibino, C. Kobayashi, H. Terasaki, H. Kagami, M. Ueda, M. M. Lerch, J. Mayerle, Gut 2016, 65, 1838.
T. Kobayashi, H. Honda, Tissue Eng. 2005, 11, 489. [383] A. Manz, N. Graber, H. M. Widmer, Sens. Actuators, B 1990, 1, 244.
[352] E. Castro, J. F. Mano, J. Biomed. Nanotechnol. 2013, 9, 1129. [384] C. D. Chin, V. Linder, S. K. Sia, Lab Chip 2012, 12, 2118.
[353] A. Ito, Y. Takizawa, H. Honda, K. I. Hata, H. Kagami, M. Ueda, [385] S. Cardoso, D. C. Leitao, T. M. Dias, J. Valadeiro, M. D. Silva,
T. Kobayashi, Biochem. Eng. J. 2004, 20, 119. A. Chicharo, V. Silverio, J. Gaspar, P. P. Freitas, J. Phys. D: Appl.
[354] A. K. Gupta, R. R. Naregalkar, V. D. Vaidya, M. Gupta, Phys. 2017, 50.
Nanomedicine 2007, 2, 23. [386] P. Yager, T. Edwards, E. Fu, K. Helton, K. Nelson, M. R. Tam,
[355] B. Sumer, J. Gao, Nanomedicine 2008, 3, 137. B. H. Weigl, Nature 2006, 442, 412.
[356] T. Lammers, S. Aime, W. E. Hennink, G. Storm, F. Kiessling, [387] T. Jamshaid, E. T. T. Neto, M. M. Eissa, N. Zine, M. H. Kunita,
Acc. Chem. Res. 2011, 44, 1029. A. E. El-Salhi, A. Elaissari, TrAC, Trends Anal. Chem. 2016, 79, 344.
[357] S. Mura, P. Couvreur, Adv. Drug Delivery Rev. 2012, 64, 1394. [388] R. Monošík, L. Angnes, Microchem. J. 2015, 119, 159.
[358] T. Lammers, V. Subr, K. Ulbrich, W. E. Hennink, G. Storm, [389] D. Mark, S. Haeberle, G. Roth, F. Von Stetten, R. Zengerle, Chem.
F. Kiessling, Nano Today 2010, 5, 197. Soc. Rev. 2010, 39, 1153.
[359] B. L. Viglianti, S. A. Abraham, C. R. Michelich, P. S. Yarmolenko, [390] S. Haeberle, R. Zengerle, Lab Chip 2007, 7, 1094.
J. R. MacFall, M. B. Bally, M. W. Dewhirst, Magn. Reson. Med. 2004, [391] P. N. Nge, C. I. Rogers, A. T. Woolley, Chem. Rev. 2013, 113, 2550.
51, 1153. [392] M. A. M. Gijs, Microfluid. Nanofluid. 2004, 1, 22.
[360] B. L. Viglianti, A. M. Ponce, C. R. Michelich, D. Yu, S. A. Abraham, [393] J. Zhou, D. A. Khodakov, A. V. Ellis, N. H. Voelcker, Electrophoresis
L. Sanders, P. S. Yarmolenko, T. Schroeder, J. R. MacFall, 2012, 33, 89.
D. P. Barboriak, O. M. Colvin, M. B. Bally, M. W. Dewhirst, Magn. [394] D. R. Ballerini, X. Li, W. Shen, Microfluid. Nanofluid. 2012, 13, 769.
Reson. Med. 2006, 56, 1011. [395] D. M. Cate, J. A. Adkins, J. Mettakoonpitak, C. S. Henry, Anal.
[361] M. De Smet, E. Heijman, S. Langereis, N. M. Hijnen, H. Grüll, Chem. 2015, 87, 19.
J. Controlled Release 2011, 150, 102. [396] A. K. Yetisen, M. S. Akram, C. R. Lowe, Lab Chip 2013, 13, 2210.
[362] Y. Onuki, I. Jacobs, D. Artemov, Y. Kato, Biomaterials 2010, 31, [397] C. Ruffert, Micromachines 2016, 7, 21.
7132. [398] R. Ganguly, I. K. Puri, Wiley Interdiscip. Rev.: Nanomed. Nanobio-
[363] D. Ho, X. Sun, S. Sun, Acc. Chem. Res. 2011, 44, 875. technol. 2010, 2, 382.
[364] J. Xie, G. Liu, H. S. Eden, H. Ai, X. Chen, Acc. Chem. Res. 2011, 44, [399] Y. Jiang, H. Wang, S. Li, W. Wen, Sensors 2014, 14, 6952.
883. [400] M. A. M. Gijs, F. Lacharme, U. Lehmann, Chem. Rev. 2010, 110,
[365] Q. Chen, H. Ke, Z. Dai, Z. Liu, Biomaterials 2015, 73, 214. 1518.
[366] D. Yoo, J. H. Lee, T. H. Shin, J. Cheon, Acc. Chem. Res. 2011, 44, [401] C. H. Weng, C. C. Huang, C. S. Yeh, H. Y. Lei, G. B. Lee, Microfluid.
863. Nanofluid. 2009, 7, 841.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (34 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advhealthmat.de

[402] F. C. Cabrera, A. F. A. A. Melo, J. C. P. De Souza, A. E. Job, ized Systems for Chemistry and Life Sciences, MicroTAS 2014, San
F. N. Crespilho, Lab Chip 2015, 15, 1835. Antonio, Texas, USA 2014, 437.
[403] L. Rao, B. Cai, L. L. Bu, Q. Q. Liao, S. S. Guo, X. Z. Zhao, [428] S. V. Pereira, G. A. Messina, J. Raba, J. Chromatogr. B: Anal.
W. F. Dong, W. Liu, ACS Nano 2017, 11, 3496. Technol. Biomed. Life Sci. 2010, 878, 253.
[404] Y. S. Lin, K. S. Huang, C. H. Yang, C. Y. Wang, Y. S. Yang, [429] J. J. Lee, K. J. Jeong, M. Hashimoto, A. H. Kwon, A. Rwei,
H. C. Hsu, Y. J. Liao, C. W. Tsai, PLoS One 2012, 7, 0033184. S. A. Shankarappa, J. H. Tsui, D. S. Kohane, Nano Lett. 2014, 14, 1.
[405] K. S. Krishna, Y. Li, S. Li, C. S. S. R. Kumar, Adv. Drug Delivery Rev. [430] B. Park, S. J. Choi, Biosens. Bioelectron. 2017, 90, 269.
2013, 65, 1470. [431] L. Chen, C. Chen, P. Wang, L. F. Wu, T. Song, J. Magn. Magn.
[406] J. Wang, Y. Song, Small 2017, 13, 1604084. Mater. 2017, 427, 90.
[407] J. Boken, S. K. Soni, D. Kumar, Crit. Rev. Anal. Chem. 2016, 46, 538. [432] C. H. Wang, K. Y. Lien, J. J. Wu, G. B. Lee, Lab Chip 2011, 11, 1521.
[408] H. Xu, B. Dong, S. Xu, X. Sun, J. Sun, Y. Yang, L. Xu, X. Bai, [433] L. Malic, X. Zhang, D. Brassard, L. Clime, J. Daoud, C. Luebbert,
S. Zhang, Z. Yin, H. Song, Biomaterials 2017, 138, 69. V. Barrere, A. Boutin, S. Bidawid, J. Farber, N. Corneau, T. Veres,
[409] C. Wang, M. Ye, L. Cheng, R. Li, W. Zhu, Z. Shi, C. Fan, J. He, Lab Chip 2015, 15, 3994.
J. Liu, Z. Liu, Biomaterials 2015, 54, 55. [434] L. Malic, X. Zhang, D. Brassard, L. Clime, J. Daoud, A. Boutin,
[410] B. Kwak, J. Lee, D. Lee, K. Lee, O. Kwon, S. Kang, Y. Kim, Biosens. C. Luebbert, V. Barrere, S. Bidawid, N. Corneau, J. Farber, T. Veres,
Bioelectron. 2017, 88, 153. in 18th Int. Conf. on Miniaturized Systems for Chemistry and Life Sci-
[411] C. W. Shields, J. L. Wang, K. A. Ohiri, E. D. Essoyan, B. B. Yellen, ences, MicroTAS 2014, Texas, USA 2014, 2546.
A. J. Armstrong, G. P. López, Lab Chip 2016, 16, 3833. [435] D. A. Kanayeva, R. Wang, D. Rhoads, G. F. Erf, M. F. Slavik,
[412] M. Antfolk, T. Laurell, Anal. Chim. Acta 2017, 965, 9. S. Tung, Y. Li, J. Food Prot. 2012, 75, 1951.
[413] C. Wyatt Shields Iv, C. D. Reyes, G. P. López, Lab Chip 2015, 15, 1230. [436] F. Khademolhosseini, C. C. Liu, C. J. Lim, M. Chiao, Biomed.
[414] M. Hejazian, W. Li, N. T. N’Guyen, Lab Chip 2015, 15, 959. Microdevices 2016, 18, 1.
[415] K. A. Hyun, H. I. Jung, Lab Chip 2014, 14, 45. [437] S. H. Song, J. Choi, H. I. Jung, Electrophoresis 2010, 31, 2762.
[416] S. Tasoglu, U. A. Gurkan, S. Wang, U. Demirci, Chem. Soc. Rev. [438] H. C. Tekin, M. A. M. Gijs, Lab Chip 2013, 13, 4711.
2013, 42, 5788. [439] D. L. Arruda, W. C. Wilson, C. Nguyen, Q. W. Yao, R. J. Caiazzo Jr.,
[417] D. Gao, H. F. Li, G. S. Guo, J. M. Lin, Talanta 2010, 82, 528. I. Talpasanu, D. E. Dow, B. C. S. Liu, Expert Rev. Mol. Diagn. 2009,
[418] M. Glynn, D. Kirby, D. Chung, D. J. Kinahan, G. Kijanka, J. Ducrée, 9, 749.
Journal of Laboratory Automation 2014, 19, 285. [440] L. Johansson, K. Gunnarsson, S. Bijelovic, K. Eriksson, A. Surpi,
[419] W. G. Lee, Y. G. Kim, B. G. Chung, U. Demirci, A. Khademhosseini, E. Göthelid, P. Svedlindh, S. Oscarsson, Lab Chip 2010, 10, 654.
Adv. Drug Delivery Rev. 2010, 62, 449. [441] C. Y. Lin, K. C. Ho, in 2007 NSTI Nanotechnology Conf. and Trade
[420] K. Y. Lien, L. Y. Hung, H. Y. Lei, G. B. Lee, in 14th International Con- Show—NSTI Nanotech 2007, Technical Proc., 2007.
ference on Miniaturized Systems for Chemistry and Life Sciences 2010, [442] R. O. Rodrigues, M. Bañobre-López, J. Gallo, P. B. Tavares,
MicroTAS 2010, Groningen, The Netherlands 2010, 94. A. M. T. Silva, R. Lima, H. T. Gomes, J. Nanopart. Res. 2016, 18 , 194.
[421] L. Y. Hung, J. C. Chang, Y. C. Tsai, C. C. Huang, C. P. Chang, [443] D. Lombardi, P. S. Dittrich, Anal. Bioanal. Chem. 2011, 399, 347.
C. S. Yeh, G. B. Lee, Nanomedicine 2014, 10, 819. [444] L. Agiotis, I. Theodorakos, S. Samothrakitis, S. Papazoglou,
[422] T. F. Kong, W. Ye, W. K. Peng, H. W. Hou, Marcos, P. R. Preiser, I. Zergioti, Y. S. Raptis, J. Magn. Magn. Mater. 2016, 401, 956.
N. T. Nguyen, J. Han, Enhancing malaria diagnosis through micro- [445] M. Stipsitz, G. Kokkinis, C. Gooneratne, J. Kosel, S. Cardoso,
fluidic cell enrichment and magnetic resonance relaxometry detection. F. Cardoso, I. Giouroudi, Key Eng. Mater. 2015, 644, 207.
Scientific Reports, 2015, 5, 11425. [446] W. Li, C. M. Mao, J. H. Yu, Z. J. Sun, in ITME2009—Proc. 2009
[423] Y. H. Lin, C. H. Chiang, M. H. Wu, T. M. Pan, J. D. Luo, IEEE Int. Symp. on IT in Medicine and Education, 2009, 1150.
C. C. Chiou, Applied Physics Letters 2011, 99, 253704. [447] I. Giouroudi, F. Keplinger, Int. J. Mol. Sci. 2013, 14, 18535.
[424] J. Sheng, L. Zhang, J. Lei, H. Ju, Anal. Chim. Acta 2012, 709, 41. [448] T. Kokalj, E. Pérez-Ruiz, J. Lammertyn, New Biotechnol. 2015, 32,
[425] M. L. S. Benuzzi, S. V. Pereira, J. Raba, G. A. Messina, Microchim. 485.
Acta 2016, 183, 397. [449] W. Wu, C. Z. Jiang, V. A. L. Roy, Nanoscale 2016, 8, 19421.
[426] D. Issadore, H. J. Chung, J. Chung, G. Budin, R. Weissleder, [450] A. Aseri, S. K. Garg, A. Nayak, S. K. Trivedi, J. Ahsan, Int. J. Pharm.
H. Lee, Adv. Healthcare Mater. 2013, 2, 1224. Sci. Rev. Res. 2015, 31, 119.
[427] I. Pereiro, J. Kucerova, L. Alexandre, B. Dupuy, Z. Bilkova, [451] N. V. Long, Y. Yang, T. Teranishi, C. M. Thi, Y. Cao, M. Nogami,
J. L. Viovy, L. Malaquin, S. Descroix, in 18th Int. Conf. on Miniatur- J. Nanosci. Nanotechnol. 2015, 15, 10091.

Adv. Healthcare Mater. 2018, 7, 1700845 1700845  (35 of 35) © 2017 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

Вам также может понравиться