You are on page 1of 9

Send Orders for Reprints to reprints@benthamscience.

ae
239

Current Drug Targets, 2018, 19, 239-247


REVIEW ARTICLE
ISSN: 1389-4501
eISSN: 1873-5592

Functionalized Magnetic Nanostructures for Anticancer Therapy Impact


Factor:
3.236

BENTHAM
SCIENCE

Eugenia Dumitra Teodor1,*, Florentina Gatea1, Anton Ficai2 and Gabriel Lucian Radu3
1
Centre of Bioanalysis, National Institute for Biological Sciences, Bucharest, Romania; 2Department of Science and
Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University
POLITEHNICA of Bucharest, Bucharest, Romania; 3Department of Analytical Chemistry and Instrumental Analysis,
Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Bucharest, Romania

Abstract: Background: In the last years, the production and applications of nanoparticles based on
iron oxides in the field of biomedicine presented a great interest due to their particular properties. Be-
cause of the expansion of the pharmaceutical industry numerous new systems for drugs delivery have
appeared, and those centered on magnetic nanoparticles are in a particular attention and in different
ARTICLE HISTORY promising developmental stages.
Objective: In this mini review, some representative, interesting and feasible magnetic nanostructures ob-
Received: September 16, 2015
Revised: November 10, 2015 tained recently (from last 5-6 years) with possible use in antitumor/anticancer therapy are presented.
Accepted: November 19, 2015
Current Drug Therapy

Results: The synthesis of these nanostructures with magnetic properties implies very simple assembling
DOI: procedures and presents one of the lowest cytotoxic profiles. Magnetic nanostructures displayed possible
10.2174/1389450117666160208145835
appliance in a large diversity of biotechnological and medical fields, both for diagnose and therapy.
Conclusion: Different types of magnetic nano-carriers loaded with different antitumor/anticancer
agents and the cases tested in vivo are considered.

Keywords: Functionalized magnetic nanostructures, anticancer therapy, drug delivery, combined drug release, imaging and
hyperthermia.

1. INTRODUCTION applications (in vitro and in vivo), and other magnetic


nanostructures based on metal alloys (e.g. FeCo, FePt,
In the last 10 years, many aspects about nanotechnologies CoPt3) are used, at this time, only for applications in vitro
had a great impact on the researchers, mainly, but even on because of their high toxicity toward living cells [2-6]. The
administrations, industries, investors, finance organizations, magnetic nanoparticles have specific physical and chemical
and for ordinary people. Numerous studies and experiments properties which make them applicable for different objec-
were done; thus, to have a contribution in the field of new tives as well as magnetic resonance imaging and hyper-
magnetic nanostructures with biomedical applications it is thermia, drug delivery, cell targeting, cell labelling and bio
important to have a multidisciplinary approach and to have a separation [1, 7-11]. For all mentioned purposes, high
harmonized knowledge in many domains such us physics, magnetic saturation and appropriate surface functionaliza-
chemistry and biochemistry, biology, materials science, for tion of nanoparticles are required; in addition, high bio-
the reason that this issue is very complex. In this context, it compatibility is necessary [12].
is important to design nanoparticle formulations with mag- In recent years, the design and application of new nano-
netic behavior, appropriate sizes and charging for penetrate sized carrier systems provoked emergent attention in
cells/tissues and, in the perspective of clinical applications, nanotechnologies area [13, 14]. The nanostructures are tar-
with the capacity to avoid or cross the main biological barri- geted to deliver different substances in a passive or in an
ers which disturb nanoparticles to reach their target tissue or active way. For tumor tissues passive targeted drug delivery
organ [1]. can be used because of the reduced lymphatic systems of
tumor tissues and their permeable vasculature with pore di-
For biomedical applications, nanoparticles based on iron
mensions ranging between 100 and 780 nm [15, 16]. These
oxides are the main widely explored because of their specific
features allow what is called the “enhanced permeability and
magnetic behavior and insignificant cytotoxicity. Magnetite
retention” effect, which permits greater accumulation of de-
(Fe3O4) and maghemite (γ -Fe2O3), are the most frequently livered nanoparticles at the solid tumor site. On the other
used for synthesis of magnetic nanoparticles with biomedical hand, for active targeted drug delivery is necessary a compli-
cated system which implies a covalent conjugation of target-
*Address correspondence to this author at the Centre of Bioanalysis, Faculty ing molecules on the nanoparticle surface, molecule which
Name, National Institute for Biological Sciences, Bucharest, Romania; can identify and can link to particular ligands specifically
Tel: +40 0212200900; Fax: +40 0212200900; E-mail: eu_teodor@yahoo.com expressed in cancer cells [17, 18].

1873-5592/18 $58.00+.00 © 2018 Bentham Science Publishers


240 Current Drug Targets, 2018, Vol. 19, No. 3 Teodor et al.

Speaking about “drug delivery” that term refers to the Singh et al. (2011) published an elaborated study about
biologic active agents of interest which are immobilized in, utilization of paclitaxel loaded magnetic nanoparticles con-
or linked to an organic or bio-polymeric shell or to an inor- jugated with lectin for leukemia therapy. It is known that to
ganic particle. In most of such cases the therapeutic sub- use the magnetic nanoparticles as carriers for drugs delivery
stances are more protected by immobilization and their effi- and to avoid the in vivo aggregation of nanoparticles, these
ciency can be enhanced, and in that way, novel targeted must be covered with a hydrophobic or hydrophilic polym-
therapies could be possible [19]. Many methods were pro- eric shell [57]. In addition, the obtained magnetic nanoparti-
posed for a good strategy for delivery of drugs. After Laurent cles ought to maintain their magnetic properties and aqueous
et al., 2011 [20], this strategy could be: (1) modification of dispersibility besides the capacity of high drug loading, an-
drugs using chemical procedures [21, 22]; (2) drug immobi- ticipated drug release profile and a good tolerance of normal
lization in vesicles [23-25] or drug immobilization in inor- cells and tissues [12]. Their earlier studies have demon-
ganic hollow nano-shells [26-28]; (3) systems with con- strated that covering the magnetic nanoparticles with glyc-
trolled release of drugs based on polymers [29-31] or based eryl mono-oleate, which is a lipid polymer with a long am-
on vectors hydrogel-type [32-35], and (4) integrated systems phiphilic chain, the aqueous dispersibility is supplied and
at small-scale such as microchip systems [36-39]. Moreover, both types’ hydrophilic and hydrophobic drugs can be incor-
due to systemic circulation, the ways of delivery of drugs are porated [58, 59]. The usage of specific molecular signature
an important subject in the case of delivery of active sub- aids in supplying the suitable therapeutic amount of drug at
stances on living systems. For improving the discrimination the targeted site, and helps the benefit of receptor mediated
in chemotherapy, intravenous [40, 41], intraperitoneal [42], endocytosis. Further, the efficiency of cellular uptake for
pulmonary [43], transdermal [44-46], nose [47, 48], vaginal magnetic nanoparticles with paclitaxel entrapped and for
[49] and eye [50, 51] paths of administration were ap- paclitaxel loaded MNP conjugated with lectin was compared
proached. with simply paclitaxel on a leukemic cell line (K562) [57].
Wang et al. (2011) have developed an innovative
Even though there are substantial progresses in targeted
DNRMNPs (Daunorubicin-magnetic nanoparticles) formula-
drug delivery systems, there are, yet, many conventional
tion for sustained release of daunorubicin, an antitumor agent
practices, and have high benefits for more efficient strategies
from anthracycline class. First, the iron oxide core was cov-
for delivery of drugs. The general opinion is that the critical ered with oleic acid containing daunorubicin, and then the
zones of expectations for efficient systems for drugs delivery nanoparticles were stabilized by Pluronic F-127®. The
should be: (1) high magnetization and narrow size distribu- authors concluded that obtained DNR-MNP formulations are
tion, (2) optimized zeta-potential values, (3) long half-life or very efficient systems for drug delivery because after intra-
long period of circulation, (4) great bio accessibility and par- venous injection they observed that the dosing interval of
ticular targeting, (5) capacity for intracellular distribution, drug is extended and the side effects for high plasma daun-
(6) receptiveness to stimuli (i.e. efficiency), (7) imaging ca- orubicin amounts are decreased and all these facts improved
pacity, and (8) biodegradation [52-54]. Wahajuddin & Arora patient compliance for a potential application in systemic
(2012), Tietze et al. (2015) published large reviews about administration [60].
superparamagnetic iron oxide nanoparticles (SPIONs), mag-
netic core fabrication, coatings, drugs loading, and utility as Tumor cells have developed metabolic aptitudes to sur-
drug delivery vehicles, so we will refer here only to other vive under unfavorable microenvironments therefore they
cases of magnetic nanostructures with possible use in anti- developed a phenotype more aggressive. An early-
tumor therapy [54, 55]. recognized and distinctive biochemical marker of the altered
phenotype of tumor cells is the increased utilization of glu-
1.1. Different Types of Functionalized Magnetic cose [61]. Consequently, a number of scientists had the idea
Nanoparticles for Antitumor Drug Delivery to propose that glucose transporter could be a target for di-
agnose and therapy and could moderate the emerging of tu-
Recently, Akbarzadeh et al. (2012) reported the associa- mor [62, 63].
tion of magnetic nanoparticles (iron oxide) and doxorubicin For improving some innovative vectorized magnetic
hydrochloride as the anticancer agent. As polymeric support nanostructures and using the idea of increased glucose con-
was used poly (D, L-lactic-co-glycolic acid) poly (ethylene sumption of tumor cells, Shan et al. (2012) described the
glycol) (PLGA-PEG) and the obtained nanoparticles with synthesis and the characterization (using transmission elec-
encapsulated anticancer drug were used for local treatment. tron microscopy and infrared spectroscopy) of an innovative
In the same work was reported the synthesis by ring-opening conjugated SPIO NPs with 2-deoxy-D-glucose (2-DG), ab-
polymerization using as initiator polyethylene glycol (with breviated as γ-Fe2O3-DMSA-DG NPs, which was assessed
different molecular mass) and D, L-lactide and glycolide to on human breast cancer cells line MDA-MB-231 [64]. The
obtain some similar PLGA-PEG triblock copolymers. The synthesis of γ- Fe2O3 -DMSA-DG NPs was done by assem-
obtained data pointed out that the chains of copolymer suc- bling the γ- Fe2O3 NPs coated with meso-2, 3-
cessfully entrapped the Fe3O4 nanoparticles and the resulted dimercaptosuccinic acid (DMSA) by conjugation of surface
nanoparticles are biocompatible and have appropriate dimen- carboxyl groups of DMSA with the functional groups of 2-
sions for in vivo applications. The authors concluded that the DG. Later, the same group [65] reported an improved syn-
efficacy and the targeting ability of obtained nanoparticles in thesis and methodically study of the composition and charac-
lung cancer treatment during future experiments in vivo will teristics of γ- Fe2O3 -DMSA-DG NPs. They assessed the
be assessed [56]. obtained nanostructures on other cancer cells line (Human
Functionalized Magnetic Nanostructures for Anticancer Therapy Current Drug Targets, 2018, Vol. 19, No. 3 241

cervical cancer cells/HeLa) regarding its ability to improve solid tumors. The main complication which appears in leu-
the targeting effect for tumor cells in comparison with the γ- kemia chemotherapy is the phenomenon of resistance to dif-
Fe2O3-DMSA NPs in vitro. ferent chemotherapeutic agents of leukemic cells.
Rouhollah et al. (2013) used polyamidoamine dendrimer- This fact is determined by the same mechanism of induc-
coated magnetic nanoparticles (DcMNPs) as an operative ing apoptosis (by initiating the intrinsic mitochondrial, or
system of delivering doxorubicin to a target tissue. The sta- cytochrome c/Apaf- 1/caspase-9 pathway) for the majority of
bility of nanoparticles, the drug loading and drug release the anti-cancer agents. The apoptotic pathways in chronic
capacities were considered. It was noticed that the obtained myeloid leukemia are mostly blocked due to Bcr-Abl gene
low-generation nanoparticles had pH-sensitive characteris- expression, p53 mutation and deficiency of Fas receptor and
tics for drug release. functional Apaf-1 [74]. A hopeful strategy to avoid the
mechanism of resistance and in the same time to reduce the
The results are promising for the understanding and the
side effects during higher chemotherapy dosage is the com-
improvement of new pH- sensitive systems for the delivery
bination of usual chemotherapy with new approaches to ini-
of drugs with the intention of defeating the drug resistance
tiate apoptosis in leukemia cells.
for the duration of cancer therapy [66].
Recently, another promising approach was developed by In some cases, enzymes or natural antitumor substances
were used as anticancer agents. Teodor et al. (2009) obtained
Unterweger et al. (2014) who generated a novel magnetic
biocompatible hydrogel-magnetic nanoparticles based on
carrier system for cisplatin [67]. Nanoparticles were coated
chitosan and hyaluronic acid with L- asparaginase (anti-
by a hybrid structure of dextran and hyaluronic acid and
leukemia agent) entrapped (Fig. 1). The synthesized hydro-
showed excellent biocompatibility. Cisplatin functionaliza-
gel-magnetic nanoparticles presented dimensions lower than
tion led to a noticeable change in the biological outcome,
making these particles potent drug- delivery vehicles with 30 nm in dried phase, and lower than 300 nm in swelled
phase, being able to penetrate the cells and tissues (particu-
the desired efficacy against tumor cells [67]. The synthesized
larly tumor tissues) [35]. The obtained nanostructures are
nanoparticles were assessed only in vitro. The same team has
very stable in water and in physiological medium and the
a promising approach using fatty acid-coated SPIONs, sub-
enzyme activity could be long time preserved. The L-
sequent albumin shielding and adsorptive mitoxantrone func-
asparaginase release was preliminary assessed in neutral and
tionalization, in order to obtain both antitumor-effective
magnetic nanoparticles and reliable and easy scale-up syn- acid environment (in vitro) [75].
thesis pathways [68]. Particles derived from this approach
have proven their stability over time under appropriate stor-
age conditions [69]. These features are necessary for the at-
tempted manufacturing as investigational medicinal products
to be conforming to good manufacturing practice facilities.
Gu et al. (2014) synthesized a composite based on Fe3O4
and hydroxyapatite with mesoporous properties. The
Fe3O4/hydroxyapatite composite was investigated for effi-
ciency in drug release behavior using as model drug doxoru-
bicin (a chemotherapeutic agent). The release experiments
showed a low burst effect for doxorubicin and the composite
presented a pH-dependent behavior. The authors emphasize
that the method for the preparation of composite was acces-
sible, reliable and ecological [70].
Wu et al. (2014) synthesized clusters by assembling
Fe3O4 with SiO which could covalently link to bridges of
polyethylene glycol dicarboxylic acid modified by 3-
aminopropyltriethoxysilane and 2,2,2-trifluoroethanol. These
bridges can electrostatic interact with doxorubicin and form
clusters which were tested for delivery of doxorubicin and
demonstrated to be recyclable. These clusters could effec-
tively release doxorubicin and inhibit the progression of can-
cer cells in a breast cancer cells line MCF-7 [71]. The same
team, Zou et al. (2015) synthesized and characterized
doxorubicin loaded-mesoporous magnetic nanoparticles with
apoptotic effect on breast cancer cells [72]. Moreover, Elbi-
aly et al. (2015) synthesized doxorubicin loaded magnetic
gold nanoparticles with excellent antitumor properties which Fig. (1). TEM image (a) and schematic illustration (b) of hydrogel
were tested in vivo on nude mice [73]. magnetic nanoparticles.
Chemotherapy continues to be the leading policy for leu-
Chen et al. (2009) reported that MNPs-Fe3O4 could en-
kemia treatment since hematological malignant tumor cannot
dorse the apoptosis induced by gambogic acid (GA) in leu-
be resolved by surgical intervention or radiation therapy as
242 Current Drug Targets, 2018, Vol. 19, No. 3 Teodor et al.

kemic cells in vitro, and the combination of MNPs-Fe3O4 established that this nano-vector could improve magnetic
and GA could have a synergistic effect on apoptosis induc- resonance imaging (MRI) contrast in vitro, and could allow
tion caused by the regulation of different proliferative and the study of MRI treatment efficacy in vivo. The prominent
anti apoptotic gene products, as well as caspase-3, bax, bcl-2, specificity and effectiveness of this vectorized nano-system
NF-κB, and surviving [76]. The authors concluded that com- make it possible for using in gene therapy for malignant tu-
bining MNPs-Fe3O4 with GA could provide a less toxic mors treatment [93, 94].
agent for leukemia treatment. The same team (Wang et al.,
2011) continued the studies and demonstrated that the 1.2. Combined Drug Release, Hyperthermia and Imaging
MNPs-Fe3O4 combined with GA could be a potential ap-
Among the imaging techniques, magnetic resonance im-
proach in the pancreatic cancer treatment [77].
aging (MRI) is a compelling procedure for medical diagnos-
Yallapu et al. (2013) proposed for therapy of pancreatic tic applicable on soft tissue [95]. The MRI technique in-
cancer a formulation composed from magnetic nanoparticles cludes also the benefit of using nonionizing radiation and
loaded with curcumin, MNP-CUR, which was in vitro and in presents high sensitivity, great specificity, capacity of multi-
vivo assessed for therapeutic effectiveness. Bio-distribution planar imaging and elevated anatomical resolution. The bio-
studies demonstrated that a significant amount of MNP-CUR medicine domain witnessed an increasing attention, in the
formulation was able to reach the pancreatic xenograft tu- last years, in the utilization of magnetic nanomaterials in bio
mor(s), which suggests its clinical translational potential separation, in magnetic cells and tissues targeting, in hyper-
[78]. Another experiment with curcumin was realized by thermia therapy, in tumor drug delivery and treatment, and
Sundar et al. (2014), who used 3-aminopropyltriethoxy si- other different diseases effective treatment [20, 52, 96-102].
lane (APTES) coated magnetite nano-powders as carriers for
Foy et al. (2010) proposed the development of a formula-
anticancer drug curcumin. Only physico-chemical charac-
tion based on magnetic nanoparticles with both properties,
terization, drug loading and drug release were done [79]. Zhu
et al. (2012) presented a study with the same curcumin and namely imaging and drug delivery, which could be used for
”image-guided drug therapy” [103]. Their proposed MNPs
with doxorubicin as model anticancer agent. The intracellu-
are based on a magnetic core covered with oleic acid (OA)
lar delivery of curcumin and doxorubicin, which are hydro-
and stabilized with an amphiphilic block copolymer. For-
phobic, from the shell of magnetic nanostructures was as-
merly, they confirmed that respectively MNPs assemblage
sessed on the glioblastoma U-87 MG cell line [80].
can present extended MRI contrast for tumors and hydro-
Another aspect which is in attention nowadays is RNA phobic anticancer drug can be attached to MNPs shells for
interference (RNAi), a modality for living cells to regulate sustained drug delivery [104, 105]. Later, they developed
gene expression via small RNA molecules. Short interfering improved MNPs with optical imaging characteristics via
RNA (siRNA) is defined as ”double stranded, non-coding novel near infrared dyes to quantitatively determine their
RNA, which binds to complementary mRNA to direct gene long-term bio-distribution and tumor localization in mice
silencing through argonaute, an endonuclease within the with xenograft breast tumors, with and without an external
RNA-induced silencing complex” (RISC) [81, 82]. In the magnetic field.
gene therapy area, RNAi mechanism is developing very fast
Sun et al. (2010) described the production of a new sort
due to its great therapeutic potential in the management of a
of multifunctional fluorescent magnetic nanoparticles, (~30
variety of maladies based on abnormal gene expression, and
nm in diameter), in which multiple fluorescent CdTe quan-
tumors are a most important objective where this knowledge
tum dots are attached around individual silica-coated super-
could be used [83-85]. Treatment with siRNA is capable to
control pathogenesis and can induce apoptosis in cancer cells paramagnetic Fe3O4 nanoparticles. In addition, on the surface
of obtained nanoparticles were attached activated carboxylic
by silencing the genes involved in uncontrolled cell growth.
groups for facile bio-conjugation with bio-molecules [106].
Because RNA molecules are anionic and hydrophilic, not
like usual chemotherapeutic agents, their internalization by The obtained nanostructures displayed both magnetic and
cells through passive diffusion is not possible [85]. There is fluorescent properties, which are encouraging for their appli-
considerable attention in obtaining secure and efficient de- ance in bio separation, magnetic targeting and imaging. The
livery devices to facilitate both delivery and intracellular carboxyl groups attached on the nanoparticles surface pre-
trafficking of siRNA. A wide range of systems for siRNA sented chemical activity and were able to link different bio-
delivery, as well as liposomes, viruses, cationic polymers logically active compounds such as antibodies, enzymes, and
and dendrimers, cell-penetrating peptides, gold nanoparti- peptides, allowing the use of the nano-systems in identifica-
cles, magnetic nanoparticles and semiconductor quantum tion of specific targets. The Fe3O4/CdTe magnetic/ fluores-
dots were investigated [86-92]. cent nanostructures associated with some specific antibodies
were successfully employed for immuno-labeling and fluo-
The main difficulty for siRNA application in therapy of
rescent imaging of HeLa cells [106]. A similar experiment
cancer continues to be the specific and useful delivery of
was done by Chen et al. (2012) based on rotavirus structural
siRNA to target cancer cells. The vector-type nanostructures,
protein VP4-coated Fe3O4 nanoparticles for dual modality
can be suitable to overcome these difficulties if are appropri-
magnetic resonance/fluorescence cellular imaging and drug
ately designed. Veiseh et al. (2010) reported the assembling
of a siRNA nano-vector with magnetic core which was as- (doxorubicin) delivery [107] or by other team works for fluo-
rescence and photoacoustic imaging [108, 109].
sessed for internalization capacity of siRNA by tumor cells
and for intracellular transferring of siRNA towards enhanced Zhao et al. (2013) explored the feasibility and effective-
knockdown of targeted gene expression. In addition, they ness of chemotherapy combined with magnetic and thermal
Functionalized Magnetic Nanostructures for Anticancer Therapy Current Drug Targets, 2018, Vol. 19, No. 3 243

effect obtained through combination of magnetic nanoparti- investigation of a PLGA drug delivery system for cancer
cles with methotrexate, an anti- cancer drug for breast cancer treatment is ongoing (including organ targeting, controlling
comprehensive treatment. The in vitro cytotoxicity results on of drug release and in vivo evaluation of the treatment effi-
human breast cancer cell MCF-7 indicated that the bi-modal cacy of chemotherapy in tumor models) [113].
cancer treatment approach using combined magnetic fluid A hybrid combination which contains magnetic nanopar-
hyperthermia and chemotherapy is more effective than
ticles (iron oxide), poly (vinyl alcohol) and doxorubicin en-
mono-modal treatment, indicating a thermal enhancement
trapped was obtained by Wang et al. (2014) [114]. The com-
effect of hyperthermia together with the drug cytotoxicity
posite was tested in trans-catheter arterial release using a
[110].
liver tumor model led to embolization of the liver tumor
Meenach et al. (2013) reported the synthesis of different blood vessels in vivo. After the embolization the composite
hydrogel nanocomposites which simultaneously can exhibit was disassembled and then the nanostructures were capable
hyperthermia and can deliver paclitaxel as anticancer agent to penetrate the tumor tissue and release the doxorubicin.
[111]. The synthesized nanostructures were assembled from The composite presented low cytotoxicity and was receptive
poly (ethylene glycol) (PEG) - based hydrogels containing to MRI, so that it could consider a promising theranostic
PEG (n = 1000) methyl ether methacrylate and PEG (n = agent [114].
400) di-methacrylate ester, and the magnetic nanoparticles Collagen, hydroxyapatite, cisplatin and magnetite were
(iron oxide) immobilized in polymeric structure. The capac-
utilized to obtain antitumor drug delivery systems (DDS) for
ity to induce hyperthermia of the obtained hydrogel-
bone cancer treatment. There are some very important ad-
nanostructures was function of network crosslinking; hydro-
vantages correlated with the application of mentioned DDS.
gels with lower swelling capacity showed higher hyperther-
First of all, collagen and hydroxyapatite are efficient in bone
mia in comparison with hydrogels with higher swelling ca-
defect regeneration especially after the surgical resection of
pacity. The release of paclitaxel showed to be dependent of the cancerous tissue and also act as a support for cisplatin
the structure of hydrogel network and the drug exhibited
delivery. Cisplatin and magnetite are two potential antitumor
non-Fickian release from the hydrogel-nanostructures. The
materials with complementary activity. Moreover, the syner-
experiments were done using hyperthermia, paclitaxel, and
gic activity is more important because the produced hyper-
both hyperthermia and drug (to observe eventually synergis-
thermia is beneficial not only due to the antitumor activity
tic cytotoxicity) on three cell lines: M059K (glioblastoma),
but also can enhance the delivery of the cisplatin. Based on a
MDA MB 231 (breast carcinoma), and A549 (lung adeno- recent patent [115], as well as some unpublished papers, the
carcinoma). The experiments showed that paclitaxel associ-
delivery rate can be controlled by the applied electromag-
ated with hyperthermia was more efficient on A549 cell line,
netic field. The delivery rate of cisplatin can be intensified
but the other cell lines did not presented the same reaction.
when hyperthermia is produced. For instance, the delivery
A similar experiment presented Sahoo et al. (2014) who rate can be intensified by applying an adequate, medical fre-
obtained nano-systems based on superparamagnetic manga- quency alternative electromagnetic field after a while (for
nese ferrite (MnFe2O4) nanoparticles covered with instance 20 arbitrary units). At this moment, along with the
mesoporous silica and with fluorescent moieties attached. temperature increase, the delivery rate increase (Fig. 2).
These nanostructures can be used in cancer cell imaging and From the point of view of the antitumor activity of these
in cancer diagnosis and can deliver doxorubicin to cancer systems, the improvement is due to the produced hyperther-
cells. In addition was proved that these nano-systems were mia as well as the increased cisplatin delivery [116].
specifically captured by HeLa cancer cells in contrast with The use of these DDS is also recommended because can
normal cells. In vitro biological experiments revealed that
also assure strong, short term antitumor activity and, at long
the nanoparticles with folate attached and loaded with
term, sustained antitumor activity, due to the presence of
doxorubicin presented high efficiency for both targeting and
magnetite and/or silver nanoparticles. Based on the literature
killing the cancer cells. The in vivo studies using mice con-
data, silver exhibits antimicrobial and antitumor activity and
firmed the usefulness and the biocompatibility of the de-
its use in many implantable materials or devices can assure a
scribed nano-systems [112]. long term activity [117, 118]. The antitumor activity of dif-
Ye et al. (2014) have synthesized PLGA-SPION-Mn:ZnS ferent systems is presented in Fig. (3).
vesicles as a multifunctional drug delivery system consisting
Multifunctional systems with more antitumor compo-
of a biodegradable polymeric shell containing a payload of
nents can be obtained by combining the activity of cytostat-
multiple imaging agents and an anti-cancer drug. Poly (lac-
ics (with different antitumor mechanism), magnetite (which
tic-co-glycolic acid) (PLGA) vesicles were fabricated by induces antitumor activity due to the produced hyperthermia)
encapsulating inorganic imaging agents of SPIONs, manga-
and nanoparticles (with intrinsic or photo thermal induced
nese-doped zinc sulfide (Mn:ZnS) quantum dots (QDs) and
antitumor activity). It can be seen that, the highest antitumor
the anticancer drug busulfan entrapped in PLGA nanoparti-
activity (expressed as arbitrary units) can be obtained by
cles using an emulsion evaporation method. The quantum
combining all the three mentioned mechanisms, and this
dots enhanced the fluorescence visualization of cell uptake.
activity is dose dependent. If one of the components is miss-
PLGA vesicles have also been demonstrated to have high ing, the antitumor activity is considerably lower. Usually,
entrapment efficiency for the lipophilic drug busulfan and a
these systems are designed containing cytostatics; if not, a
sustained release of drug. Some in vivo MRI and histological
much lower antitumor activity is obtained even at short term.
studies were done in a rat model to elucidate the bio distribu-
At long term point of view, the antitumor activity is still pre-
tion of PLGA vesicles. The authors stipulated that further
sent, but at much lower level, corresponding to the activity
244 Current Drug Targets, 2018, Vol. 19, No. 3 Teodor et al.

100 1.57 25 1.2


120 1.81.58 25 1.4
140 1.59 25 1.6
160 1.6 25 1.8
1.6

Concentraation,AU 1.4

1.2
The two profiles corespond to the release in the presence (generating 42oC) and
in the absence (37oC) of the alternating electromagnetic field; the alternating
1 electromagnetic field is aplied at 25AU

0.8 AU - arbitrary concentration/time units


37oC
0.6
42oC
Electromagneticfieldapplication
0.4

0.2

0
0 20 40 60 80 100 120 140 160

Ti
Time,AU
AU

Fig. (2). Cisplatin delivery profile from magnetic cisplatin loaded DDS.

induced by magnetite and nanoparticles. Based on the above CONCLUSION


mentioned facts, function of the initial composition, the anti-
Magnetic nanoparticles-based drug delivery systems con-
tumor activity can be designed at different antitumor level
sist of a very promising category of nanoparticles for medi-
(high enough to assure curative role) while, at long term it
cal applications. Highly complicated nano-scaled drug deliv-
can assure recurrence preventive purposes.
ery systems requiring complex materials science and engi-
Based on the paper published by Andronescu et al. neering and large work teams were designed in the past few
(2010) [119] these systems can be easily used in clinics be- years. They are in strong contrast to very simple nano-
cause, surgical removal of the tumor bonny tissue is an es- pharmaceuticals that have already been clinically applied,
sential step in the treatment protocol of bone cancer and, in such as DOXIL®18, a doxorubicin-encapsulated liposome.
this step, without any supplementary surgical intervention, The deficient clinical assessments of promising formulations
these systems can be implanted. The use of these drug deliv- still remain a major problem for practical application of de-
ery systems is highly recommended because the cytostatics, signed antitumor nanostructures. More in vitro and in vivo
which are the most toxic components of these systems, are reproducible experiments and more medical investigations of
delivered loco-regionally and consequently the systemic tox- already obtained nanostructures with reproducible character-
icity is lower, compared with the cases where, the cytostatics istics instead of continuous process to obtain new more so-
are administrated via a systemic route [120]. phisticated nanostructures need to be done. Complex, multi-
component drug delivery systems can be promising antitu-
mor alternatives with good curative short term activity and
Shortterm long term recurrence preventive anti-tumor systems.

C=Cytostatics CONSENT FOR PUBLICATION


H+N+C H=Hyperthermia
ntitumoralActivity,AU

Not applicable.
N=Nanoparticles
CONFLICT OF INTEREST
Longterm The authors declare no conflict of interest, financial or
H/N+C otherwise.
H+N+C

H/N+C ACKNOWLEDGEMENTS
H/N H+N
An

All the funds are provided by Romanian Agency for Sci-


H+N entific Research.
H/N
REFERENCES
[1] Krishnan KM. Biomedical nanomagnetics: a spin through possibili-
Time,AU ties in imaging, diagnostics, and therapy. IEEE Trans Magn 2010;
46(7): 2523-8.
Fig. (3). Short/long term activity of magnetic, cytostatic delivery [2] Behrens S, Bönnemann H, Matoussevitch N, et al. Surface engi-
drug delivery systems loaded with silver nanoparticles. neering of Co and FeCo nanoparticles for biomedical application. J
Phys Condens Matter 2006; 18: S2543-61.
Functionalized Magnetic Nanostructures for Anticancer Therapy Current Drug Targets, 2018, Vol. 19, No. 3 245

[3] Gu H, Ho PL, Tsang KWT, Wang L, Xu B. Using bifunctional [28] Akerman ME, Chan WCW, Laakkonen P, Bhatia SN, Ruoslahti E.
magnetic nanoparticles to capture vancomycin-resistant enterococci Nanocrystal targeting in vivo. Proc Natl Acad Sci USA 2002; 99:
and other gram-positive bacteria at ultralow concentration. J Am 12617-21.
Chem Soc 2003; 125: 15702-03. [29] Uhrich KE, Cannizzaro SM, Langer RS, Shakesheff KM. Polym-
[4] 4 Martins MA, Neves MC, Esteves ACC, et al. Biofunctionalized eric systems for controlled drug release. Chem Rev 1999; 99: 3181-
ferromagnetic CoPt3/polymer nanocomposites. Nanotechnology 98.
2007; 18: 215609. [30] Veronese FM, Pasut G. PEGylation, successful approach to drug
[5] Guo D, Wu C, Li X, Jiang H, Wang X, Chen B. In vitro cellular delivery. Drug Discov Today 2005; 10: 1451-8.
uptake and cytotoxic effect of functionalized nickel nanoparticles [31] Richardson TP, Peters MC, Ennett AB, Mooney DJ. Polymeric
on leukemia cancer cells. J Nanosci Nanotechnol 2008; 8: 2301-07. system for dual growth factor delivery. Nat Biotechnol 2001; 19:
[6] Zeisberger M, Dutz S, Müller R, Hergt R, Matoussevitch N, Bön- 1029-34.
nemann H. Metallic cobalt nanoparticles for heating applications. J [32] Vinogradov SV, Bronich TK, Kabanov AV. Nanosized cationic
Magn Magn Mater 2007; 311: 224-7. hydrogels for drug delivery: preparation, properties and interactions
[7] Latorre M, Rinaldi C. Applications of magnetic nanoparticles in with cells. Adv Drug Deliv Rev 2002; 54: 135-47.
medicine: magnetic fluid hyperthermia. P R Health Sci J 2009; [33] Gupta P, Vermani K, Garg S. Hydrogels: from controlled release to
28(3): 227–38. pH-responsive drug delivery. Drug Discov Today 2002; 7: 569-79.
[8] Jain P, Lee K, El-Sayed I, El-Sayed M. Calculated absorption and [34] Qiu Y, Park K. Environment-sensitive hydrogels for drug delivery.
scattering properties of gold nanoparticles of different size, shape, Adv Drug Deliv Rev 2001; 53: 321-39.
and composition: applications in biological imaging and biomedi- [35] Teodor E, Litescu SC, Lazar V, Somoghi R. Hydrogel-magnetic
cine. J Phys Chem B 2006; 110(14): 7238-48. nanoparticles with immobilized L-Asparaginase for biomedical ap-
[9] Merritt WM, Lin YG, Spannuth WA, et al. Effect of interleukin-8 plications, J Mat Sci Mat Med 2009; 20: 1307-14.
gene silencing with liposome-encapsulated small interfering RNA [36] Santini JT, Cima MJ, Langer R. A controlled release microchip.
on ovarian cancer cell growth. J Nat Cancer Inst 2008; 100(5): 359- Nature 1999; 397:335-8.
72. [37] LaVan DA, McGuire T, Langer R. Small-scale systems for in vivo
[10] Brigger I, Dubernet C, Couvreur P. Nanoparticles in cancer therapy drug delivery. Nat Biotechnol 2003; 21: 1184-91.
and diagnosis. Adv Drug Deliv Rev 2002; 54(5): 631-51. [38] Grayson ACR, Choi IS, Tyler BM, Wang PP, Brem H, Cima MJ,
[11] Ferrari M. Cancer nanotechnology: opportunities and challenges. Langer R. Multi-pulse drug delivery from a resorbable polymeric
Nat Rev Cancer 2005; 5(3): 161-71. microchip device. Nat Mater 2003; 2: 767- 72.
[12] Gupta A, Gupta M. Synthesis and surface engineering of iron oxide [39] Razzacki SZ, Thwar PK, Yang M, Ugaz VM, Burns MA. Inte-
nanoparticles for biomedical applications. Biomateri- grated microsystems for controlled drug delivery. Adv Drug Deliv
als.2005:26:3995-4021. Rev 2004; 56: 185-98.
[13] Torchilin VP. Targeted pharmaceutical nanocarriers for cancer [40] Collins JM. Pharmacologic rationale for regional drug delivery. J
therapy and imaging. AAPS J. 2007; 9(2): E128-47. Clin Oncol 1984; 2: 498-504.
[14] Yokoyama M, Okano T. Introduction: targetable drug carriers: [41] Egan TD. Intravenous drug delivery systems: Toward an intrave-
present status and a future perspective. Adv Drug Deliver Rev nous ''vaporizer''. J Clin Anesthesia 1996; 8: S8-S14.
1996; 21(2): 77-80. [42] Markman M. Intraperitoneal antineoplastic drug delivery: rationale
[15] Montet X, Figueiredo JL, Alencar H, et al. Tomographic fluores- and results. Lancet Oncol 2003; 4: 277-83.
cence imaging of tumor vascular volume in mice. Radiol 2007; [43] Edwards DA, Hanes J, Caponetti G, et al. Large porous particles
242(3): 751-8. for pulmonary drug delivery. Science 1997; 276: 1868-71.
[16] Bremer C, Ntziachristos V, Weitkamp B, et al. Optical imaging of [44] Prausnitz MR, Bose VG, Langer R, Weaver JC. Electroporation of
spontaneous breast tumors using protease sensing ‘smart’ optical mammalian skin – a Mechanism to enhance transdermal drug-
probes. Invest Radiol 2005; 40(6): 321-7. delivery. Proc Natl Acad Sci USA 1993; 90: 10504-8.
[17] Shukla R, Thomas TP, Peters JL, et al. HER2 specific tumor target- [45] Henry S, McAllister DV, Allen MG, Prausnitz MR. Microfabri-
ing with dendrimer conjugated anti-HER2 mAb. Bioconjug Chem cated microneedles: A novel approach to transdermal drug deliv-
2006; 17(5): 1109-15. ery. J Pharm Sci 1998; 87: 922-5.
[18] Yousefpour P, Atyabi F, Vasheghani-Farahani E, et al. Targeted [46] Prausnitz MR, Mitragotri S, Langer R. Current status and future
delivery of doxorubicin-utilizing chitosan nanoparticles surface potential of transdermal drug delivery. Nature Rev Drug Discov
functionalized with anti-Her2 trastuzumab. Int J Nanomedicine 2004; 3: 115-24.
2011; 6: 1977–90. [47] Illum L. Nasal drug delivery: new developments and strategies.
[19] Mahmoudi M, Sant S, Wang B, et al. Superparamagnetic iron Drug Discovery Today 2002; 7: 1184-9.
oxide nanoparticles (SPIONs): Development, surface modification [48] Illum L. Nasal drug delivery - possibilities, problems and solutions.
and applications in chemotherapy. Adv Drug Deliv Rev 2011; 63: J Control Release 2003; 87: 187-98.
24-46. [49] Brannonpeppas L. Novel vaginal drugrelease applications. Adv
[20] Laurent S, Bridot JL, Elst LV, Muller RN. Magnetic iron oxide Drug Deliv Rev 1993; 11: 169-77.
nanoparticles for biomedical applications. Future Med Chem 2010; [50] Loftssona T, Jarvinen T. Cyclodextrins in ophthalmic drug deliv-
2: 427-49. ery. Adv Drug Deliv Rev 1999; 36: 59-79.
[21] Means GE, Feeney RE. Chemical modifications of proteins: His- [51] Le Bourlais C, Acar L, Zia H, Sado PA, Needham T, Leverge R.
tory and applications. Bioconjugate Chem 1990; 1: 2-12. Ophthalmic drug delivery systems - Recent advances. Progress
[22] Chau Y, Dang NM, Tan FE, Langer R. Investigation of targeting Retin Eye Res 1998; 17: 33-58.
mechanism of new dextranpeptide-methotrexate conjugates using [52] Liu F, Laurent S, Fattahi H, Elst LV, Muller RN. Superparamag-
biodistribution study in matrix-metalloproteinase overexpressing netic nanosystems based on iron oxide nanoparticles for biomedical
tumor xenograft model. J Pharm Sci 2006; 95: 542-51. imaging. Nanomed (Lond) 2011; 6: 519-28.
[23] Gregoriadis G. Engineering liposomes for drug delivery: Progress [53] Santhosh P.B., Ulrih N.P., Multifunctional superparamagnetic iron
and problems. Trends Biotechnol 1995; 13: 527-37. oxide nanoparticles: Promising tools in cancer theranostics, Cancer
[24] Adams ML, Lavasanifar A, Kwon GS. Amphiphilic block copoly- Lett 2013; 336: 8-17.
mers for drug delivery. J Pharm Sci 2003; 92: 1343-55. [54] Tietze R, Zaloga J, Unterweger H, et al. Magnetic nanoparticle-
[25] Rosler A, Vandermeulen GWM, Klok HA. Advanced drug delivery based drug delivery for cancer therapy. Biochem Biophys Res
devices via self-assembly of amphiphilic block copolymers. Adv Commun 2015; 468: 463-70.
Drug Deliv Rev 2001; 53: 95-108. [55] Whajuddin, Arora S. Superparamagnetic iron oxide nanoparticles:
[26] Wu GH, Milkhailovsky A, Khant HA, Fu C, Chiu W, Zasadzinski magnetic nanoplatforms as drug carriers. Int J Nanomed 2012; 7:
JA. Remotely triggered liposome release by near-infrared light ab- 3445–71.
sorption via hollow gold nanoshells. J Am Chem Soc 2008; 130: [56] Akbarzadeh A, Mikaeili H, Zarghami N, Mohammad R, Barkhord-
8175-7. ari A, Davaran S. Preparation and in vitro evaluation of doxorubi-
[27] Son SJ, Bai X, Lee SB. Inorganic hollow nanoparticles and nano- cinloaded Fe3O4 magnetic nanoparticles modified with biocom-
tubes in nanomedicine. Part 1. Drug/gene delivery applications. patible copolymers, Int J Nanomed 2012; 7: 511-26.
Drug Discov Today 2007; 12: 650-6.
246 Current Drug Targets, 2018, Vol. 19, No. 3 Teodor et al.

[57] Singh A, Dilnawaz F, Sahoo SK. Long circulating lectin conju- [81] Semple SC, Akinc A, Chen J, et al. Rational design of cationic
gated paclitaxel loaded magnetic nanoparticles: A new theranostic lipids for siRNA delivery. Nat Biotechnol 2010; 28: 172-6.
avenue for leukemia therapy. PLoS ONE 2011; 6(11): e26803 [82] Scherer LJ, Rossi JJ. Approaches for the sequence-specific knock-
[58] Dilnawaz F, Singh A, Mohanty C, Sahoo SK. Dual drug loaded down of mRNA. Nat Biotechnol 2003; 21: 1457-65.
superparamagnetic iron oxide nanoparticles for targeted cancer [83] Hannon GJ. RNA interference. Nature 2002; 418: 244-51.
therapy. Biomaterials 2010; 31: 3694–706. [84] Dykxhoorn DM, Chowdhury D, Lieberman J. RNA interference
[59] Singh A, Dilnawaz F, Mewar S, et al. Composite polymeric mag- and cancer: endogenous pathways and therapeutic approaches. Adv
netic nanoparticles for co-delivery of hydrophobic and hydrophilic Exp Med Biol 2008; 615: 299-329.
anticancer drugs and MRI imaging for cancer therapy. ACS Appl [85] Oh YK, Park TG. siRNA delivery systems for cancer treatment.
Mater Interfaces 2011; 3: 842-56. Adv Drug Deliv Rev 2009; 61: 850-62.
[60] Wang J, Chen B, Chen J, et al. Synthesis and antitumor efficacy of [86] Whitehead KA, Langer R, Anderson DG. Knocking down barriers:
daunorubicin-loaded magnetic nanoparticles. Int J Nanomed 2011; advances in siRNA delivery. Nat Rev Drug Discov 2009; 8: 129-
6: 203-11. 38.
[61] Bell GI, Burant CF, Takeda J, Gould GW. Structure and function [87] Tseng YC, Mozumdar S, Huang L. Lipid-based systemic delivery
of mammalian facilitative sugar transporters. J Biol Chem. 1993; of siRNA. Adv Drug Deliv Rev 2009; 61: 721-31.
268:19161–4. [88] Howard KA. Delivery of RNA interference therapeutics using
[62] Gold J. Inhibition of Walker 256 intramuscular carcinoma in rats polycation-based nanoparticles. Adv Drug Deliv Rev 2009; 61:
by administration of hydrazine sulfate. Oncology 1971; 25: 66-71. 710-20.
[63] Fanciulli M, Valentini A, Bruno T, Citro G, Zupi G, Floridi A. [89] Mok H, Lee SH, Park JW, Park TG. Multimeric small interfering
Effect of the antitumor drug lonidamine on glucose metabolism of ribonucleic acid for highly efficient sequence-specific gene silenc-
adriamycin-sensitive and -resistant human breast cancer cells. On- ing. Nat Mater 2010; 9: 272-8.
col Res 1996; 8: 111–20. [90] Yezhelyev MV, Qi L, O’Regan RM, Nie S, Gao X. Proton-sponge
[64] Shan XH, Hu H, Xiong F, et al. Targeting Glut1-overexpressing coated quantum dots for siRNA delivery and intracellular imaging.
MDA-MB-231 cells with 2-deoxy-D-g1ucose modified SPIOs. Eur J Am Chem Soc 2008; 130: 9006-12.
J Radiol. 2012; 81(1): 95-9. [91] Lee JS, Green JJ, Love KT, Sunshine J, Langer R, Anderson DG.
[65] Xiong F, Zhu ZY, Xiong C, Hua XQ, Shan XH, Zhang Y, Gu N. Gold, poly(beta-amino ester) nanoparticles for small interfering
Preparation, characterization of 2-deoxy-D-glucose functionalized RNA delivery. Nano Lett 2009; 9: 2402-6.
dimercaptosuccinic acid-coated maghemite nanoparticles for tar- [92] Giljohann DA, Seferos DS, Prigodich AE, Patel PC, Mirkin CA.
geting tumor cells. Pharm Res 2012; 29(4): 1087-97. Gene regulation with polyvalent siRNA-nanoparticle conjugates. J
[66] Rouhollah K, Pelin M, Serap Y, Gozde U, Ufuk G. Doxorubicin Am Chem Soc 2009; 131: 2072-73.
loading, release, and stability of polyamidoamine dendrimer-coated [93] Veiseh O, Kievit FM, Fang C, et al. Chlorotoxin Bound Magnetic
magnetic nanoparticles. J Pharm Sci 2013; 102(6): 1825-35. Nanovector Tailored for Cancer Cell Targeting, Imaging, and
[67] Unterweger H, Tietze R, Janko C, et al. Development and charac- siRNA. Deliv Biomat 2010; 31(31): 8032–42
terization of magnetic iron oxide nanoparticles with a cisplatin- [94] Zhao P, Wang HY, Gao H, Li CZ, Zhang YZ. Reversal of
bearing polymer coating for targeted drug delivery. Int J Nanomed multidrug resistance by magnetic chitosan-Fe3O4 nanoparticle-
2014; 9: 3659-76. encapsulated MDR1 siRNA in glioblastoma cell line. J Neurol Res
[68] Zaloga J, Janko C, Nowak J, et al. Development of a lauric 2013; 35: 821-8
acid/albumin hybrid iron oxide nanoparticle system with improved [95] Jin Y, Jia C, Huang SW, O’Donnell M, Gao X. Multifunctional
biocompatibility. Int J Nanomed 2014; 9: 4847-66. nanoparticles as coupled contrast agents. Nat Commun 2010; 1: 41.
[69] Zaloga J, Janko C, Agarwal R, et al. Different storage conditions [96] Fattahi H, Laurent S, Liu F, Arsalani N, Elst LV, Muller RN. Mag-
influence biocompatibility and physicochemical properties of iron netoliposomes as multimodal contrast agents for molecular imaging
oxide nanoparticles. Int J Mol Sci 2015; 16(5): 9368-84. and cancer nanotheragnostics. Nanomedicine (Lond) 2011; 6: 529-
[70] Gu L, X He, Z Wu. Mesoporous Fe3O4/hydroxyapatite composite 44.
for targeted drug delivery. Mat Res Bull 2014; 59: 65-8. [97] Issadore D, Shao H, Chung J, et al. Self-assembled magnetic filter
[71] Wu J, Wang Y, Jiang W, Xu S, Tian R. Synthesis and characteriza- for highly efficient immunomagnetic separation. Lab Chip 2011;
tion of recyclable clusters of magnetic nanoparticles as doxorubicin 11: 147-51.
carriers for cancer therapy. Appl Surf Sci 2014; 321: 43-9. [98] Das M, Dhak P, Gupta S, et al. Highly biocompatible and water-
[72] Zou Y, Liu P, Liu C-H, Zhi X-T. Doxorubicin-loaded mesoporous dispersible, amine functionalized magnetite nanoparticles, prepared
magnetic nanoparticles to induce apoptosis in breast cancer cells. by a low temperature, air-assisted polyol process: a new platform
Biomed Pharmacother 2015; 69: 355-60. for bio-separation and diagnostics. Nanotechnology 2010; 21:
[73] Elbialy NS, Fathy MM, Khalil WM. Doxorubicin loaded magnetic 125103.
gold nanoparticles for in vivo targeted drug delivery. Int J Pharma- [99] Yang X, Hong H, Grailer JJ, et al. cRGD-functionalized, DOX -
ceutics 2015; 490: 190-9. conjugated, and 64Cu - labeled superparamagnetic iron oxide
[74] Jia L, Patwari Y, Kelsey SM, Newland AC. Trail-induced apopto- nanoparticles for targeted anticancer drug delivery and PET/MR
sis in Type I leukemic cells is not enhanced by overexpression of imaging. Biomaterials 2011; 32: 4151-60.
bax. Biochem Biophys Res Commun 2001; 283: 1037-45. [100] Yallapu MM, Othman SF, Curtis ET, Gupta BK, Jaggi M, Chauhan
[75] Teodor E, Truica G, Lupescu I. The release of L-asparaginase from SC. Multi-functional magnetic nanoparticles for magnetic reso-
hydrogel-magnetic nanoparticles. Roum Biotechnol Lett 2008; 13: nance imaging and cancer therapy. Biomaterials 2011; 32: 1890-
3907-13. 905.
[76] Chen B, Liang Y, Wu W, et al. Synergistic effect of magnetic [101] Elsherbini AA, Saber M, Aggag M, El-Shahawy A, Shokier HA.
nanoparticles of Fe3O4 with gambogic acid on apoptosis of K562 Magnetic nanoparticle-induced hyperthermia treatment under mag-
leukemia cells, Int J Nanomed 2009; 4: 251-9. netic resonance imaging. Magn Reson Imaging 2011; 29: 272-80.
[77] Wang C, Zhang H, Chen B, Yin H, Wang W. Study of the en- [102] Maity D, Chandrasekharan P, Yang CT, et al. Facile synthesis of
hanced anticancer efficacy of gambogic acid on Capan-1 pancreatic water-stable magnetite nanoparticles for clinical MRI and magnetic
cancer cells when mediated via magnetic Fe3O4 nanoparticles. Int J hyperthermia applications. Nanomedicine (Lond) 2010; 5: 1571-84.
Nanomed 2011; 6: 1921-35. [103] Foy SP, Manthe RL, Foy ST, Dimitrijevic S, Krishnamurthy N,
[78] Yallapu MM, Ebeling MC, Khan S, et al. Novel curcumin loaded Labhasetwar V. Optical imaging and magnetic field targeting of
magnetic nanoparticles for pancreatic cancer treatment. Mol Cancer magnetic nanoparticles in tumors. ACS Nano 2010; 4(9): 5217-24.
Ther 2013; 12: 1471-80. [104] Jain TK, Morales MA, Sahoo SK, Leslie-Pelecky DL, Labhasetwar
[79] Sundar S, Mariappan R, Piraman S. Synthesis and characterization V. Iron oxide nanoparticles for sustained delivery of anticancer
of amine modified magnetite nanoparticles as carriers of curcumin- agents. Mol Pharmaceutics 2005; 2: 194-205.
anticancer drug. Powder Technol 2014; 266: 321-8. [105] Jain TK, Richey J, Strand M, Leslie-Pelecky DL, Flask CA, Lab-
[80] Zhu X-M, Yuan J, Leung K C-F, et al. Hollow superparamagnetic hasetwar V. Magnetic nanoparticles with dual functional proper-
iron oxide nanoshells as a hydrophobic anticancer drug carrier: in- ties: Drug delivery and magnetic resonance imaging. Biomaterials
tracelluar pH-dependent drug release and enhanced cytotoxicity. 2008; 29: 4012-21.
Nanoscale 2012; 4: 5744-54.
Functionalized Magnetic Nanostructures for Anticancer Therapy Current Drug Targets, 2018, Vol. 19, No. 3 247

[106] Sun P, Zhang H, Liu C, et al. Preparation and characterization of drugs for drug delivery and imaging. Biomaterials 2014; 35: 3885-
Fe3O4/CdTe magnetic/fluorescent nanocomposites and their appli- 94.
cations in immunolabeling and fluorescent imaging of cancer cells. [114] Wang YX, Zhu XM, Liang Q, Cheng CH, Wang W, Leung KC. In
Langmuir 2010; 26(2): 1278-84. vivo chemoembolization and magnetic resonance imaging of liver
[107] Chen W, Cao Y, Liu M, et al. Rotavirus capsid surface protein tumors by using iron oxide nanoshell/doxorubicin/poly(vinyl alco-
VP4-coated Fe3O4 nanoparticles as a theranostic platform for cel- hol) hybrid composites. Angew Chem Int Ed Engl 2014; 53(19):
lular imaging and drug delivery. Biomaterials 2012; 33: 7895-902. 4812-5.
[108] Ma Y, Tong S, Bao G, Gao C, Dai Z. Indocyanine green loaded [115] Ficai A, Andronescu E, Ghitulica CD, Ficai D, Voicu G, Albu MG.
SPIO nanoparticles with phospholipid-PEG coating for dual-modal Process for preparing composite multi-purpose materials with pos-
imaging and photothermal therapy. Biomaterials 2013; 34: 7706- sible applicability in the treatment of bone cancer; Patent No:
14. RO127725-A2; RO127725-B1;
[109] Zhang M, Cao Y, Wang L, Ma Y, Tu X, Zhang Z. Manganese [116] Ficai D, Sonmez M, Albu MG, Mihaiescu DE, Ficai A, Bleotu C.
doped iron oxide theranostic nanoparticles for combined T1 mag- Antitumoral materials with regenerative function obtained using a
netic resonance imaging and phototherma therapy. ACS Appl Ma- layer-by-layer technique. Drug Des Dev Ther 2015; 9: 1269-79.
ter Interfaces 2015; 7(8): 4650-8. [117] Patrascu JM, Nedelcu IA, Sonmez M, et al. Composite scaffolds
[110] Zhao L, Huo M, Liu J, et al. In vitro investigation on the magnetic based on silver nanoparticles for biomedical applications. J Nano-
thermochemotherapy mediated by magnetic nanoparticles com- materials 2015; 1-8.
bined with methotrexate for breast cancer treatment. J Nanosci [118] Nedelcu IA, Ficai A, Sonmez M, Ficai D, Oprea O, Andronescu E.
Nanotechnol 2013; 13(2): 741-5. Silver based materials for biomedical applications. Curr Org Chem
[111] Meenach SA, Shapiro JM, Hilt JZ, Anderson KW. Characterization 2014; 18(2): 173-84.
of PEG-iron oxide hydrogel nanocomposites for dual hyperthermia [119] Andronescu E, Ficai M, Voicu G, Ficai D, Maganu M, Ficai A.
and paclitaxel delivery. J Biomater Sci Polym Ed 2013; 24(9): Synthesis and characterization of collagen/ hydroxyapatite: mag-
1112-26. netite composite material for bone cancer treatment. J Mater Sci-
[112] Sahoo B, Devi K SP, Dutta S, Maiti TK, Pramanik P, Dhara D, Mater M 2010; 21: 2237-42.
Biocompatible mesoporous silica-coated superparamagnetic man- [120] Andronescu E, Ficai A, Albu MG, et al. Collagen-
ganese ferrite nanoparticles for targeted drug delivery and MR im- hydroxyapatite/cisplatin drug delivery systems for locoregional
aging applications, J Colloid Interface Sci 2014; 431: 31-4. treatment of bone cancer. Technol Cancer Res Treat 2013; 12: 275-
[113] Ye F, Barrefelt Å, Asem H, et al. Biodegradable polymeric vesicles 84.
containing magnetic nanoparticles, quantum dots and anticancer

PMID: 26853321

You might also like