You are on page 1of 17

Journal of Superconductivity and Novel Magnetism

https://doi.org/10.1007/s10948-018-4841-2

REVIEW PAPER

A Review on Iron Oxide Nanoparticles and Their Biomedical


Applications
P. Sangaiya1 · R. Jayaprakash1

Received: 21 March 2018 / Accepted: 11 August 2018


© Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
The iron oxide nanoparticles have a great attraction in biomedical applications due to their non-toxic role in the
biological systems. The iron oxide nanoparticles have both magnetic behaviour and semiconductor property which lead
to multifunctional biomedical applications. The iron oxide nanoparticles used in biomedical fields such as antibacterial,
antifungal and anticancer were reviewed. The uses of hematite (α-Fe2 O3 ), maghemite (γ -Fe2 O3 ) and magnetite (Fe3 O4 )
nanoparticles, for an inhibition time in biological activities, are listed in this work. Also, this review explains the use of iron
oxide nanoparticles in the biomedical fields with particular attention to the application of hematite and superparamagnetic
iron oxide nanoparticles. In this review, analysis reveals that the role of iron oxide in biological activity is good due to its
biocompatibility, biodegradability, ease of synthesis and different magnetic behaviours. The change of properties of iron
oxide nanoparticles such as particle size, morphology, surface, agglomeration and electronic properties has specific impact
in biomedical application. The review mainly focused in and discussed about antibacterial, anticancer, bone marrow and cell
labelling activities. From this review work, the iron oxide nanoparticle may be specialised in particular bacterial and cancer
treatments. Also discussed are the iron oxide nanoparticle-specific biomedical applications like human placenta, insulin and
retinal locus treatments.

Keywords Hematite · Maghemite · Magnetite · Antimicrobial · Anticancer · Cell labelling

1 Introduction biogeochemical cycles of iron in the environment. There


are different phases of iron oxide crystallites exhibited as
The iron oxide nanoparticles play a role in the biomedi- hematite (α-Fe2 O3 ), maghemite (γ -Fe2 O3 ), goethite FeOH
cal system and have a vibrant position in the day-to-day (OH) and magnetite (Fe3 O4 ) [1]. The hematite phase is con-
needs. There are plenty of nanoparticles fulfill the advanced sidered as the more stable n-type semiconductor behaviour.
development in the field of biological application. The iron Nowadays, researchers have given more attention towards
oxide nanoparticles play a major role in different applica- the stable hematite iron oxide for biomedical application.
tions, such as magnetic, electrochemical, gas sensor, energy The reason for choosing this α-Fe2 O3 (hematite) iron oxide
storage, cancer therapy and magnetic storage, and biomedi- as a specific nanoparticle is only due to its low cost
cal treatments. Among the different types of nanoparticles, and being non-toxic. This hematite iron oxide nanoparti-
hematite (iron oxide) is one of the most common, natu- cle possesses a band gap of 2.1–2.2 eV [2]. The influence
ral and environmental-friendly. It has a significant part in of environmental-friendly iron oxide nanoparticles can be
investigated in the biological system. In this aspect, many
researchers have made different attempts in biological appli-
cations [3]. The uses of magnetic and antiferromagnetic
 P. Sangaiya
iron oxide nanoparticles in biomedical areas date back four
sangaiahbose@gmail.com
decades ago. The role of nanoparticles on antibacterial
R. Jayaprakash
agents is to kill some bacterial species causing no dam-
Jayaprakash.rajan.2015@gmail.com
age to the human host cells. Iron oxide nanoparticles are
1 Nano Technology Laboratory, Department of Physics, Sri
recently used as sensors for metabolites, hyperthermia and
Ramakrishna Mission Vidyalaya College of Arts and Science, biomolecules, toxicity and magnetic nanotoxicology. Thus,
Coimbatore, Tamil Nadu, 641020 India this review has discussed the importance and enhancement
J Supercond Nov Magn

of iron oxide nanoparticles on antimicrobials, therapeutic


activities on cancer cell and other specific treatments. The
iron oxide nanoparticles synthesised in different ways may
enhance physical and chemical properties, such as chemi-
cal precipitation, sol-gel and forced hydrolysis techniques,
hydrothermal technique, surfactant-mediated/template syn-
thesis, biomimetic mineralisation, precipitation by anhy-
drous solution, microemulsion technique, flow injection
syntheses, electrochemical methods, aerosol/vapour meth-
ods and sonochemical technique [4]. Also, many researchers
focused on green synthesis like fruit extracts used for bio-
logical studies [5]. This review is mainly focused on the
Fig. 1 Differences between antiviral, antibacterial and antifungal
role of iron oxide in nanoparticles to the great extent of activity [14]
antimicrobial, antitumour and cancer treatments. The spe-
cific applications of bone marrow cells, cell labelling and
human placenta, as well as insulin are also discussed. M. Mohamed Rafi et al. [8] have synthesised α-Fe2 O3
nanoparticles with the thermal decomposition route. They
have chosen Fe2 SO4 ·7H2 O as the source of iron. They used
2 Role of Iron Oxide Nanoparticle in agarose dextran and gelatin as template components of the
Antimicrobial Activity polysaccharide. The average crystalline size of α-Fe2 O3 as
10 nm was achieved by them. The antibacterial activity
There are plenty of nanomaterials emerging at the present of α-Fe2 O3 nanoparticles on six bacteria, including these
scenario to act as specific components in bio-analytical Gram-positive S. aureus, A. hydrophila and Streptococcus
devices. Bacterial infections are among the most impor- pyogenes and three Gram-negative P. aeruginosa, E.
tant infectious diseases. The extensive research studies have faecalis and E. coli, was determined. The inhibition of α-
been made by S. Z. Moghadamtousi et al. [6], achieving Fe2 O3 nanoparticles over the above-mentioned bacteria was
new antimicrobial medicines, which is isolated from differ- tested by them. They found that α-Fe2 O3 nanoparticles did
ent sources, and the differences of physical and structural not create any change in the dosage of 12.5 mL, but the
variations of antibacterial, antifungal and antiviral isolations possible inhibition effect occurred due to an increase in
play an important role in antimicrobial studies. Figure 1 dosage. The novel biopolymer templates were synthesised
shows the specific three classifications from biomedical by using a simple thermal decomposition method. As
applications. According to the progress in the development evidenced from FTIR, good interaction between hematite
of antibacterial agents, there are still special attentions to and polysaccharide functional groups controls the hematite
find new antibacterial agents due to the development of crystal growth. The XRD measurements revealed a pure-
multidrug-resistant bacteria. In many cases, the change in phase α-Fe2 O3 . HRSEM and HRTEM confirmed the
the property of material in the nanorange certainly occu- dumbbell nature of the iron oxide nanoparticles obtained
pies a definite position in this field. It is clearly understood at 800 ◦ C, and the lowest particle size was found to be
due to the enhancement of performances of sensitivity 77 nm for gelatin-Fe2 O3 . The magnetisation measurements
and detection limit down to single molecule detection. At of the iron oxide nanoparticles prepared at 800 ◦ C exhibit
the same time, the different combinations of nanomateri- a ferromagnetic behaviour at room temperature for all the
als, according to its characteristic feature, are capable to samples. The hematite nanoparticles have an outstanding
increase the performances of microbial application. The 17 antimicrobial efficiency against some bacterial pathogens. It
bacterial species are named as follows: Vibrio harveyi, Vib- is concluded that further exploration of this field is needed
rio alginolyticus, Vibrio vulnificus, Vibrio parahaemolyti- to develop eco-friendly bionanomaterials for biomedicines.
cus, Vibrio cholerae, Bacillus subtilis, Bacillus cereus, M. Arakha et al. [9] have studied the growth kinetic
Aeromonas hydrophila, Streptococcus agalactiae, Staphy- analysis. They studied the effect of interaction pattern at
lococcus aureus, Staphylococcus intermedius, Staphylococ- the iron oxide nanoparticle-bacterium interface initially by
cus epidermidis, Edwardsiella tarda, Pseudomonas aerug- following the growth kinetics of B. subtilis and E. coli in the
inosa, Enterococcus faecalis, Klebsiella pneumoniae and absence and presence of different negative surface potential
Escherichia coli [7]. Among these bacterial species, the iron oxide nanoparticles and p-iron oxide nanoparticle
five bacteria respond for iron oxide nanoparticles, such as concentrations. They also performed the study by preparing
Bacillus subtilis, Bacillus cereus, Aeromonas hydrophila, the mother cultures of test organisms in nutrient broth
Escherichia coli and Staphylococcus aureus. on taking a loop full of bacteria from the specified slant
J Supercond Nov Magn

culture, which were cultured overnight at 37 ◦ C and 150 rpm potential of E. coli cells was measured by dynamic light
agitation. They have studied it for different concentrations scattering. The repercussions of iron oxide nanoparticle due
of both negative surface potential iron oxide nanoparticles to progressive surface adsorption were further identified by
and positive surface potential iron oxide nanoparticles as the researchers from the potential shift 42 to 278 mV.
2.5 μM, 5 μM, 10 μM, 25 μM and 50 μM (micrometres). The antibacterial activity of glycol chitosan with iron
The stock solution of iron oxide nanoparticles was prepared oxide nanoparticles was studied by B. S. Inbaraj et al. [11],
by them on dispersing iron oxide nanoparticles in sterilised particularly for E. coli O157:H7, Salmonella enteritidis
nutrient broth and sonicated for 10 min, followed by and S. aureus. The magnetic property of iron oxide
UV radiation sterilisation. The reaction mixtures without nanoparticles widely used for this activity was reduced
nanoparticles were taken as controls. The bacterial mother after coating with iron oxide nanoparticles for future
cultures of 20 μL (microlitres) were added by them to biomedical applications. The reason for utility of iron oxide
the different reaction mixtures, and the reaction volumes nanoparticle is due to the rapid and efficient action as well
were also adjusted by adding nutrient broth to a final as the biomedical application due to the magnetic field
volume of 300 μL with nanoparticles. They performed which remains unabsorbed in the body. They mentioned
the growth kinetic studies by measuring an optical density that this property facilitated access to deep regions in the
at 600 nm using a plate reader at regular time interval. living tissues. The size effect was studied by W. Zhang
At the approximately mid log phase of bacterial growth, et al. [12], and the adsorption of hematite nanoparticles
respective concentrations of nanoparticles were added by on E. coli cells was due to nanoparticles as the adsorbent
them in the respective reaction mixtures and its different in adsorption experiments onto E. coli cells. They used
doted colours which show growth reaction are shown in the equation as Kb = Kc s/(1 − s) to calculate the
Fig. 2. The influence of nanoparticle on bacterial growth total interaction energies between hematite nanoparticles
has been estimated by them from the data collection in the and E. coli cells. They found that the total interaction
growth curve. energy for each size describes the variation of the interfacial
W. Zhang et al. [10] have mentioned that E. coli cells had energy as a function of the interacting distance when a
noticeable deformation with hematite treatment for 45 min single hematite nanoparticle approaches an E. coli cell.
with a statistical significance. Their study said that the Also, S. Arokiyaraj et al. [13] tested leaf extract with
hematite-treated cells became significantly harder or stiffer iron oxide into E. coli cells by co-precipitation method. E.
than the untreated ones, which was evidenced by indentation Fazio et al. [14] have prepared polymeric-based iron oxide
and spring constant measurements. The average indentation nanoparticles by varying the ablation parameters of pulsed
of the hematite-treated E. coli cells was 120 nm, which laser ablation. They found that polymeric phase increased
was significantly lower than that of the untreated cells. The the iron oxide nanoparticle’s stability. The antimicrobial
spring constant of hematite-treated E. coli cells (0.28 nN/nm activity of iron oxide nanoparticles on Staphylococcus
and 0.11 nN/nm) was about 20 times higher than that of aureus was also studied by C. Narendhar et al. [15] and
untreated ones (0.01 nN/nm and 0.01 nN/nm). The zeta N. Tran et al. [16] on MTT assay. Their results indicated

Fig. 2 Fluorescence
microscopic images of B.
subtilis and E. coli in the
absence and presence of n-IONP
and p-IONP [9]
J Supercond Nov Magn

that the iron oxide nanoparticle prepared by pulsed laser were very effective and less toxic than the other metal oxide
ablation technique in water shows a potential bacterial nanoparticles, and the concentration/size of nanoparticles
effect on Staphylococcus aureus. They also mentioned that have played an important role on antimicrobial studies. In
a minimum effect was observed for the bacterial growth in the future, a rapid and efficient action can be expected
the PVA-Fe2 O3 nanocolloids. They suggested about PVA- as the iron oxide magnetic field remains unabsorbed in
Fe2 O3 nanoparticle to be non-toxic, and so, this could the body, which facilitates access to deep regions in the
be integrated in the drug carrier, multiblock polymeric living tissues. Also, the semiconducting and magnetic
nanocomposites. The antibacterial activities of iron oxide iron oxide nanoparticles are advantageous over commercial
nanoparticles made interesting materials for the potential antibiotics owing to their smaller dosage requirement
applications as efficient vectors for drug delivery-specific and elimination of possible harmful side effects within
platforms for drug targeting studies. M. P. Kumar et al. [17] short time duration. The recent studies of iron oxide
modified the surface of iron oxide nanoparticles promised nanoparticles show enhanced antimicrobial action against
to offer useful biomedical applications after biocide coating, various microorganisms than the commercial drug. Also,
and the particles are found to be 100% effective to deactivate the diagnosis method of iron oxide nanoparticles was
methicillin-resistant Staphylococcus aureus bacteria within found to be an easier way in detecting microorganisms at
2 h. A. S. Ahmed et al. [18] presented their investigation low level concentration from drinking water samples. The
of iron oxide nanoparticles for their antibacterial potential present review study demonstrates the potential application
against Bacillus cereus and Klebsiella pneumoniae. They of magnetic iron oxide nanoparticles to control the
found that 5 μg/mL and 40 μg/mL concentrations of treatment of fungal infections and antimicrobial indications,
iron oxide nanoparticles exhibit a 40–50% loss in viable and iron oxide nanomaterial has promising potential for
bacterial cells, and that 80 μg/mL concentrations acted battling widespread bacterial infection either by themselves
as bactericidal for causing a 90–99% loss in the viability or used in combination with the commercially existing
of antimicrobial applications. G. Silvia et al. [19] have antibiotics. The biocompatibility of pure and different
reported the leaf extracts of Cynometra ramiflora were used doped superparamagnetic iron oxide nanoparticles could
to synthesise iron oxide nanoparticles. The leaf extracts with be suitable for antimicrobial applications and medical
iron oxide nanoparticles were very effective in inhibiting devices. In summary, iron oxide nanoparticles have high
E. coli and S. epidermidis which may lead to interest of antimicrobial activity and excellent safety to mammalian
iron oxide property changes with the addition of other cells. The greater antimicrobial activity was observed for
doping materials like Ag, Ti and Co. The researchers S. S. hematite compared to conventional magnetite nanoparticles.
Tawab et al. [20] reported natural antioxidant gallic acid-
assisted magnetite prepared by in situ and post-synthesis
method. The particle size of 5 nm and 8 nm for in situ 3 Iron Oxide’s Role on Anticancer Activity
synthesis of gallic acid-functionalised magnetite was found, Studies
and experimental antioxidant activity and antimicrobial
studies proved the outstanding antimicrobial activity on In the nanotechnology broad field of the growing mod-
bacterial and fungal strains. K. H. Prakash et al. [21] tested ern world, the growing technology also brings unidentified
the silver-loaded hematite nanocomposites by hydrothermal social issues, such as atmospheric contaminations, and plays
process for biomedical application. Y. A. M. Abdulhady et a vital role on human health issues. The social needs lead
al. [22] prepared titanium-coated iron oxide nanoparticles to new invention and more attention on specific iron oxide
by co-precipitation method and evaluated them against nanoparticles. The iron oxide nanoparticle and its applica-
four pathogenic bacterial. species. Their report showed tions were used in a wide range of biomedical applications.
promising antibacterial activities against Gram-positive and These include anticancer, antitumour, bone marrow cell and
Gram-negative strains which offers a high reduction percent cell binding applications. This review focused on iron oxide
after 30 min by 150 μg/mL of nanoparticles. B. Mayank et nanoparticles which exhibit omnipresent and unique phys-
al. [23] first reported that they tried multiple mechanisms ical and chemical properties when their dimensions are
of bactericidal activity, and that iron oxide nanoparticles reduced to between several and hundreds of nanometres. M.
exhibited minimal/no cytotoxicity to human cells and were Malekigorji et al. [24] clarified that superparamagnetic iron
effective on bacteria. In this work, antibacterial properties oxide nanoparticles (SPIONs) are small, synthetic α-Fe2 O3
of iron oxide and cobalt oxide nanoparticles enhanced the (hematite), γ -Fe2 O3 (maghemite) or Fe3 O4 (magnetite) par-
bactericidal activity against pathogenic bacterial strains B. ticles with a core diameter ranging from 10 to 20 nm.
subtilis, S. aureus, E. coli and Salmonella typhi and, also, Also, X. P. Nguyen et al. [25] discussed the fabrication and
were highly biocompatible and found to be non-toxic to structure characterisation of Fe3 O4 and Fe2 O3 nanoparti-
the human cell line MCF-7. The iron oxide nanoparticles cles encapsulated by several organic and inorganic materials
J Supercond Nov Magn

such as oleic acid (OL), starch (ST), dextran (D), chitosan iron oxide detect cancer. They are as follows: (1) detec-
(CS), O-carboxymethyl chitosan (OCMCS) and the co- tion of receipts over-expressed on the cancer cell surface in
polymer of poly(styrene-co-acrylic acid) (St-co-AA). The the tumour mass, (2) detection of unusual angiogenesis in
stability and toxicity were also considered. J. S.-H. Huang the tumour micro-environment, (3) detection of circulating
and R.-S. Juang [26] have reviewed the recent design strate- tumour cells and (4) detection of soluble tumour biomark-
gies for biochemical and biomedical applications of mul- ers, and these stages are shown in Fig. 3. The behaviour
tifunctional magnetic nanoparticles. These nanoparticles of iron oxide nanoparticles depends on many parameters
were combined with other materials which enhanced the such as coating, targeting molecules, overall particle size,
potential and broadened the applications of such compos- delivery route, doping, coating, surfactant, delivery root and
ite bi-functional nanomaterials. The magnetic nanoparticles biomarkers.
were conjugated with proper ligands or smart polymers. The The cytotoxicity of hematite nanoparticles on Hek293
bi-functional nanomaterials exhibit highly selective binding cells was identified by H. Yan and B. Zhang [29].
and high sensitivity. They are using the nanoparticles for The nanoparticles were prepared by them using the raw
magnetic resonance imaging (MRI) diagnosis and therapy materials Fe(NO3 )3 and BaCl2 which were dissolved in
(hyperthermic and chemotherapeutic drug release). Also, 30 mL deionised water containing PVP. They use NaOH
they mentioned the remarkable use of iron oxide nanopar- as the reducing agent. The autoclave method was adopted
ticles, which is one of the achievements of their diagnostic for their hematite nanoparticles, and the particle size around
and therapeutic purposes. The use of nanoparticles should 180 nm was achieved by them. They tested the viability
be eliminated by biological systems without any other detri- of Hek293 cells by exposing hematite nanoparticles at the
mental effects. The researchers pointed out that magnetic dosage levels 100 μg/mL to 500 μg/mL. The exposure
nanoparticles could also provide a strong contrast effect in time given by them is 1 day, and they found that the
T2-weighted images due to high magnetisation, conjugated hematite nanoparticles did not show the toxicity at the low
cancer-targeting agent and core-satellite hybrid nanostruc- dosage level, i.e. < 300 μg/mL; they reported significant
tures. H. Chen et al. [27] made review investigation based cytotoxicity was beginning to start at higher dosage levels
on specific needs to improve cancer management that pro- > 350 μg/mL. Finally, they found that the considerable
vide good detection sensor devices to detect tumours in their decreases were observed for the 500 μg/mL dosage level
early stage, precise and dynamic monitoring of patients’ with increasing hematite nanoparticles. In conclusion, those
response to treatment, simultaneous tracking to the levels researchers reported that hematite nanoparticles can induce
of multiple biomarkers in vivo and high sensitivity that can the oxidative stress in Hek293 cell and decrease both
be translated into capability in practice to identify cancer the antioxidative capacity and activity of antioxidative
at early or pre-syndrome stage which requires further clini- enzymes in Hek293 cells. The monodispersed hematite
cal development and corroboration. This review focused on nanoparticles were successfully prepared via a simple
iron oxide nanoparticles used in cancer imaging and diagno- hydrothermal method. The α-Fe2 O3 nanoparticles have a
sis. Also, the unique properties of iron oxide nanoparticles homogeneous dispersion in water or ethanol solution with
have impact with controlled size, composition, morphol- a diameter distribution of 180 nm. They reported that
ogy and physical properties. Also, Z. Bakhtiary et al. [28] hematite nanoparticles showed a dose- and time-dependent
have pointed out about four different stages to help how cytotoxicity to Hek293 cells, which was attributed to the

Fig. 3 Four different stages in


identifying cancer using iron
oxide nanoparticles [28]
J Supercond Nov Magn

lipid peroxidation of the cells due to the interaction with being killed by heat with the help of magnetic hyperther-
nanoparticles. mia. The main challenges in this field is the generation
K. Rajendran et al. [30] have synthesised the hematite of therapeutic temperature that is selective in the whole
nanoparticles from ferric chloride by eco-friendly biologi- tumour region. So, they brought this idea to improve mag-
cal method using the culture supernatant of B. cereus SVK1 netic hyperthermia of breast cancer by using innovative
at physiological temperature and pH. They formed the nanoparticles. The researchers also reported that a high
hematite nanoparticles of 15–30 nm diameter, and they stud- heating potential was functionalised with cell internalisa-
ied anticancer activity against HepG2 cell. Their study sug- tion and a chemotherapeutic agent to increase cell death.
gested that this process was efficient and cost-effective com- Their report also highlighted that the therapeutic effects
pared to the conventional chemical method for preparing of breast cancer magnetic hyperthermia could be strongly
hematite nanoparticle. Their report showed the possibility of enhanced by the combination of MF66 functionalised with
the use of less-toxic hematite nanoparticles for cancer treat- N6L and doxorubicin (DOX) and magnetic hyperthermia.
ment. The researchers have analysed the cytotoxicity effect They approached the study on combining two ways of
of iron oxide nanoparticles on human HepG2 liver cancer tumour cell killing such as magnetic hyperthermia and
cell and tried various concentrations of hematite nanoparti- chemotherapy on injecting nanoparticles intratumourally as
cles like 50 ng/mL, 100 ng/mL, 250 ng/mL, 500 ng/mL and clinical practice. A convenient carrier system for suspending
1000 ng/mL (nanograms). Their main intention was identifi- superparamagnetic iron oxide nanoparticles in an aqueous
cation of cytotoxicity manifestation towards HekG2 cancer medium of PEG-PE-based polymeric micelles was reported
cells at low concentration. M. Calero et al. [31] have made by R. M. Sawant et al. [34]. The stability of superpara-
another study on incubating the cells with dimercaptosuc- magnetic iron oxide nanoparticles was also made between
cinic acid-coated superparamagnetic iron oxide nanoparti- polymeric micelles with respect to superparamagnetic iron
cles for different time intervals, from 0.5 to 72 h. They found oxide nanoparticles with and without polymeric micelles.
that their nanoparticles showed efficient internalisation and They found that superparamagnetic iron oxide nanoparticle-
relatively slow clearance [31]. Time-dependent uptake stud- loaded PEG-PE micelles provided dramatically improved
ies revealed that the maximum accumulation was observed T2 relaxation rates. These superparamagnetic iron oxide
for dimercaptosuccinic acid-coated superparamagnetic iron nanoparticle measles could be easily modified with anti-
oxide nanoparticles after 24 h of incubation, and then cancer, nuclear some-specific map 2C5 with the preserva-
they were slowly removed from cells. The superparamag- tion of the antibody-specific activity to make them can-
netic iron oxide nanoparticles were verified by them using cer cell-specific. Mob 2C5-superparamagnetic iron oxide
energy-dependent endocytosis and localised in endosomes. nanoparticle immunomicelles specifically recognise and
The macropinocytosis uptake and clathrin-mediated inter- bind cancer cells in vitro, bring a higher quantity of super-
nalisation depending on the particle size were observed by paramagnetic iron oxide nanoparticles to these cells and
them using transmission electron microscopy. They reported have a potential to serve as an MRI contrast agent with
that the MCF-7 cells accumulated these nanoparticles with- improved T2 for better tumour imaging. In addition to this,
out any significant effect on cell morphology, cytoskele- the specific accumulation of mAb 2C5-superparamagnetic
ton organisation, cell cycle distribution, reactive oxygen iron oxide nanoparticle immunomicelles into the cancer
species generation and cell viability. They made a confir- cells made these micelles good candidates to be used in
mation with similar behaviour to untreated control cells. conjunction with AC field-assisted hyperthermic cancer
Thus, their findings indicated that dimercaptosuccinic acid therapy.
(DMSA)-coated superparamagnetic iron oxide nanoparti- P. C. Nagajyothi et al. [35] have tested the hematite
cles possessed excellent properties in terms of efficiency nanoparticles on the cell viability and the percentage of cell
and biocompatibility to target breast cancer cells. Also, A. growth inhibition, which were calculated. The concentration
Marcu et al. [32] tested their antitumour effect on MCF-7 is dependent on cell growth inhibition with the hematite
cells and reported that iron oxide nanoparticles may provide nanoparticle and exposure on P soraleacorylifolia seeds.
better and more targeted drug accumulation in the tumour They found that these nanoparticles showed efficient
cells. The lower concentrations are more efficient in dis- anticancer activities. Finally, they revealed that an aqueous
turbing tumour cells than commercial ones. They confirmed extract of Psoralea corylifolia seeds plays a reducing agent
lowering the therapeutic dose can consequently reduce the role and became to have a significant anticancer activity
side effects of chemotherapy. against renal tumour cells at lower concentrations. Also,
The next aspects of the role of iron oxide nanoparti- DNA fragmentation on A549 cell line with L-arginine-
cles in tumour cells have been studied by S. Kossatz et coated iron oxide nanoparticles and control cells was
al. [33]. They mentioned the tumour cells were effectively examined by D. Rehana et al. [36], and they speculated
J Supercond Nov Magn

the drug paclitaxel-loaded L-arginine-coated iron oxide of toxicity. Nanoparticles with various surface modifica-
nanoparticles could be used as an effective drug carrier tions and different sizes induced slight, but possibly mean-
for the destruction of cancer cells. B. Sangeetha and A.K. ingful, changes in cell cytotoxicity and genotoxicity, which
Kumaraguru [37] have studied Sargassum myriocystum leaf would be significantly valuable in further studies of biocon-
extract to be used for synthesis of iron oxide nanoparticles. jugation and cell interaction for drug delivery, cell culture
The 2.8 nm size of biosynthesised iron oxide was achieved and cancer-targeting applications. Furthermore, in geno-
which was used for cancer treatment, and they were done toxicity analysis, the negatively charged (citrate-modified)
for in vitro and in vivo analysis. The flutamide with iron superparamagnetic iron oxide nanoparticles presented the
oxide nanoparticles increased the life span of the tumour- highest toxicity at low concentrations (100 ppm). Above
bearing mice. They reported flutamide nanoparticles at the 100 ppm, the tendency towards genotoxicity was similar
dose of 10 mg/kg body weight by decreasing the nutritional regardless of the size and surface charge but the change
fluid volume controlled the growth of tumour and increased was not significant. They observed a detailed dependence
the life span of Dalton’s lymphoma ascite-bearing mice. of cell cytotoxicity and genotoxicity on the surface modi-
Thus, flutamide nanoparticles at the dose of 10 mg/kg body fication and size of nanoparticles. It is obvious that small
weight had antitumour activity against Dalton’s lymphoma modifications in these nanoparticles induced slight, but
ascite-bearing mice. possibly meaningful, changes in cell cytotoxicity and geno-
S. C. Hong et al. [38] have widely utilised the super- toxicity; this information would be significantly valuable
paramagnetic iron oxide nanoparticles for the diagnosis in studies of bioconjugation and cell interaction for drug
and therapy of specific diseases, as magnetic resonance delivery, cell culture and cancer-targeting applications or
imaging contrast agents and drug delivery carriers, due further advanced precise control-required bioengineering.
to their easy transportation to targeted areas by an exter- They observed that superparamagnetic iron oxide nanopar-
nal magnetic field for such biomedical applications, and ticles affected the cell viability and DNA stability of L-929
superparamagnetic iron oxide nanoparticles must have mul- fibroblastic cells in a dose-dependent manner.
tifunctional characteristics, including optimised size and M. Kumar et al. [39] have reported a novel nanoparticle-
modified surface. However, the biofunctionality and bio- based delivery system that can transport therapeutic car-
compatibility of superparamagnetic iron oxide nanoparti- gos to the intracellular sites without the need for a cell
cles with various surface functional groups of different transduction or penetration domain. They have used iron
sizes have yet to be elucidated clearly. Therefore, it is oxide nanoparticles to deliver an oncopeptide, NuBCP-9,
important to carefully monitor the cytotoxicity and geno- targeting the BCL-2 BH3 domain. Citric acid/2-bromo-2-
toxicity of superparamagnetic iron oxide nanoparticles that methylpropanoic acid-capped superparamagnetic iron oxide
are surfaced-modified with various functional groups of nanoparticles were used to immobilise and deliver the
different sizes. In their study, they evaluated superparam- NuBCP-9 peptide to the cancer cells without any notice-
agnetic iron oxide nanoparticles with diameters of approxi- able off-target effects. Their results have demonstrated that
mately 10 nm and 100–150 nm, containing different surface NuBCP-9-superparamagnetic iron oxide nanoparticles effi-
functional groups. Superparamagnetic iron oxide nanopar- ciently penetrate into cancer cells and bind to their intracel-
ticles were covered with O groups, the so-called “bare lular target protein BCL-2. Moreover, significant inhibition
superparamagnetic iron oxide nanoparticles”. Following of proliferation and substantial induction of cell death were
this, they were modified with three different functional observed when cancer cells were treated with NuBCP-
groups: hydroxyl, carboxylic and amine groups by coat- 9-superparamagnetic iron oxide nanoparticles at different
ing their surfaces with tetraethyl orthosilicate (TEOS), (3- time intervals. Importantly, the IC50 values for killing of
aminopropyl)trimethoxysilane (APTMS), TEOS-APTMS breast cancer cells with NuBCP-9-superparamagnetic iron
or citrate, which imparted different surface charges and oxide nanoparticles were much lower compared to those of
sizes to the particles. The effects of superparamagnetic iron cells treated with the NuBCP-9 peptide linked with a CPP
oxide nanoparticles coated with these functional groups on (Arg-8; NuBCP-9-R8). Molecular and biochemical anal-
mitochondrial activity, intracellular accumulation of reac- yses further supported that NuBCP-9-superparamagnetic
tive oxygen species, membrane integrity and DNA stability iron oxide nanoparticles killed breast cancer cells by
in fibroblasts (L-929) were determined by water-soluble apoptosis-mediated mechanisms. Their data also demon-
tetrazolium, 2,7-dichlorodihydrofluorescein, lactate dehy- strated that administration of NuBCP-9 superparamagnetic
drogenase and comet assays, respectively. The toxicological iron oxide nanoparticles in mice bearing Ehrlich ascites
observations of their study suggested that the functional tumours (EATs) was associated with loss of tumourigenic-
groups and sizes of superparamagnetic iron oxide nanoparti- ity and extensive apoptosis in tumour tissues. These find-
cles were critical determinants of cellular responses, degrees ings showed that a non-CPP-tagged peptide could be suc-
of cytotoxicity and genotoxicity and potential mechanisms cessfully delivered to undruggable intracellular oncotargets
J Supercond Nov Magn

using superparamagnetic iron oxide nanoparticles. Finally, HER2-positive tumours in photo-acoustic imaging studies.
they suggested that this induced effective anticancer activity This data indicated that SNP20 was a potential photo-
in vitro and in vivo. The effects of the cellular interaction acoustic contrast agent for imaging of HER2-expressing
of MCF-7 cells treated with lower concentrations to higher tumours. L. Guo et al. [42] have developed superparam-
concentrations of rhodamine B-conjugated and NuBCP-9 agnetic iron oxide nanoparticles by drug delivery system.
are shown in Fig. 4. The dosage required less frequent dos- In their system, the amino group and carboxyl-modified
ing (twice/week) as compared to the daily injections that dual-function superparamagnetic iron oxide nanoparticles
were necessary for the antitumour activity of the D-amino have been synthesised as carriers. The cyclic arginine-
acid NuBCP-9-r8 peptide in vivo. Also, T. K. Jain et al. glycine-aspartic acid (RGD) peptide (c(RGDyK)) was cou-
[40] reported that the folic acid-Pluronic-coated iron oxide pled to the carrier for special av B3 integrin targeting. They
nanoparticles they used have a dose-dependent antiprolifer- used DOX as an anticancer agent and the novel triplex
ative effect on breast cancer at the earlier stage of cancer “hand” reagent Tris-surinch amino triacetate which could
cell line. be coupled to one to two amino groups. They synthesised
K. Kanazaki et al. [41] have suggested that photo-acoustic c(RGDyK)-coupled magnetic nanoparticles by this method
imaging may promise imaging modality for biomedical for a specific delivery of large amounts of DOX to integrin
information with high sensitivity and resolution. Iron oxide av B3 -expressing tumour cells. They have analysed tumour
nanoparticles have low toxicity and biodegradable prop- cell structure drug-released target cell uptake and cytotoxic-
erties. Also, they mentioned that iron oxide nanoparticle ity effect. Their results implied that the cell cytotoxicity due
delivery was restricted by its modest leakage and retention to the c(RGDyK) and DSP carrier was minimal. The inhi-
in tumours. This study was designed in the basis of iron bition ratio of RDSP of U87MG cells was higher than that
oxide nanoparticles sizes 20 nm, 50 nm and 100 nm con- of DDSP under a mole-per-mole basis. They reported that
jugated with anti-HER2 moieties for HER2-targeted photo- the inhibition ratio for RDSP reached 66.8% for U87MG
acoustic tumour imaging. The binding affinity, cellular cells while that of DDSP was 37.3% with the DOX concen-
uptake and in vivo biodistribution were examined by them. tration of 6.25 μg/mL. The IC50 value of RDSP was also
They proposed this 20-nm anti-HER2 scFv-conjugated iron lower than that of DDSP correspondingly. This behaviour
oxide nanoparticle (SNP20) as a novel photo-acoustic con- demonstrated that the special c(RGDyK)-integrin avb3 tar-
trast agent. SNP20 demonstrated high affinity and specific geting enhanced the uptake of RDSP to integrin avb3.
binding to HER2-expressing cells. It selectively visualised They suggested that this behaviour raised the drug delivery

Fig. 4 Cellular interaction studies. a Cellular uptake studies of MCF-7. b Co-localisation studies of NuBCP-9 in SPIONs [39]
J Supercond Nov Magn

effectively and concluded that RDSP not only showed the has developed a method to detect and diagnose cancers,
superparamagnetic property high trapping efficient for mag- for diagnosis of cancer relies on medical imaging but,
netic targeting and the high drug load ratio of RDSP which ultimately, ought to take tissue out for testing by optical
could also carried a large amount of DOX to target tumour imaging approach which technique is recently used on
cells. Their study reported that the specific method can also cancer diagnosis. The nanoparticle research has been
be applicable for targeted delivery of therapeutic agents successful in detecting multiple forms of breast cancer in
in tumours with high efficiency and low carrier toxicity. mice. Also, the current research has made cancer diagnosis
H. Mackova et al. [43] reported P(DMAAm-AA) γ -Fe2 O3 without the need to take tissue samples via biopsy. The
nanoparticles have higher antitumour and antimetastatic magnetic iron oxide nanoparticle is important for cancer
activities than cisplatin probably due to an enhanced oxida- diagnosis and treatments compared to semiconducting iron
tive stress in tumour cells. A. H. Atta et al. [44] have made oxide nanoparticles.
precursor complexes with resulted cytotoxic and antitumour
activity of iron oxide against breast carcinoma cell line with
priority for Fe2 O3 . 4 Iron Oxide Nanoparticle Treatments
M. Singh et al. [45] have focused on optimisation on Human Bone Marrow Cells and Molecular
method to synthesise iron oxide nanoparticles grafted with Cell Binding
PEG silane. The coating of nanoparticle surface with 3-
amino propyl triethoxy silane and carboxylated PEG ligand 4.1 Bone Marrow Cells
has provided stability in solution and size control. They
mentioned these opened nanoparticles surface to conjugate The iron oxide nanoparticles have different cell populations
with various biomolecules containing amine or carboxylic obtained from bone marrow cells (MSCs, BMCs and
group at its terminal end. Their report said that the the indigenous mobilisation of bone marrow cells) and
increased stability of these functionalised nanosystems was have specific effects on MSCs or BMCs or G-CSF. The
due to the formation of the hydrogen bonding between iron oxide nanoparticle treatment induces functional and
PEG molecules on the surface of nanoparticles. Therefore, morphological improvement which may have more effect
their study showed that the surface-modified maghemite on bone marrow cell treatments studied by L. Urdzikova
metal nanoparticles have been used in biomedical field et al. [49] in which the bone marrow cell treatment
due to their properties and biocompatibility. Thus, they and nanoparticle impacts lead to focus specific area of
suggested that the clinical use of maghemite nanoparticles research. K. Turnovcova et al. [50] have reported human
could come to an end by integration of maghemite mesenchymal stromal cells (hMSCs) are a promising tool
nanoparticles into a hybrid multifunctional structure with for future clinical application, but their use requires rapid
its improved ability to engage specific molecular binding. cell expansion in media suitable for clinical use. Therefore,
The researchers A. Masotti et al. [46] have made an attempt they tested the influence of several culture media on
on polyethylenimine (PEI)-based iron oxide. They found colony formation, population doubling (PD) time, cell
that the compound PEI-25 is a good contrast agent for cycle and surface marker expression. The choice of serum
MRI bolus tracking experiments. Actually, they reported influences hMSC expansion and cell properties; α-MEM
small differences about PEI-coated iron oxide compounds supplemented with hABS seems to be a promising candidate
were less efficient than the commercially available contrast for clinical use.
agent Endorem, and the versatility of these polymers and S.-Y.-R. Paik et al. [51] analysed the physicochemi-
derivatives resided in their ability to complex and deliver cal properties and cytotoxicity of iron oxide nanoparticles
DNA in vitro and in vivo; also, they were monitored by in bone marrow cells. They were compared with iron
non-invasive optical imaging techniques and conjugated oxide samples and surface-modified iron oxide nanoparti-
with dye. B. Bajaj et al. [47] have reported amino cles. Their results showed that the iron samples were not
acid-based diethylene trimming pentaacetic acid (DTPA)- cytotoxic to the bone marrow cells and did not affect the
functioned Fe3 O4 nanoparticles synthesised by thermal expression of cell surface, and that the effects of phago-
decomposition could be useful to radiolabelling and cancer cytosis were stronger in iron oxide nanoparticles than in
therapy. The recent development of superparamagnetic iron surface-modified iron oxide nanoparticles. B. Novotna et al.
oxide nanoparticles targeting drug delivery systems is more [52] tested human bone marrow mesenchymal stromal cells
effective on cancer therapy treatments, and it is reviewed (hBMSCs) from two donors: hBMSC-1 and hBMSC-2. The
by G. Kandasamy and D. Maity [48]. In the biomedical measurements were performed at two intervals: after 3 days
applications, natural products and green sythesised iron for cell growth without nanoparticles. The dose 15.4 g
oxide nanoparticles have been used for the treatment of Fe/mg/mL of superparamagnetic iron oxide nanoparticles
anticancer effects. A research on iron oxide nanoparticles was selected as being sufficient for in vivo cell tracking
J Supercond Nov Magn

using MRI. Concerning cell viability and cell death, only the P. Jendelova et al. [54, 55] studied animals with a corti-
hBMSC-2 seemed to be sensitive to the action of superpara- cal photochemical lesion or with a balloon-induced spinal
magnetic iron oxide nanoparticles. The TEM micrograph cord compression lesion and the fate of implanted rat bone
results of the cell viability of unlabelled and labelled hBM- marrow stromal cells and mouse embryonic stem cells
SCs are shown in Fig. 5. On the same way, G. M. van labelled with superparamagnetic iron oxide nanoparticles.
Buul et al. [53] studied cell-based therapy using a tempt- They reported that MSCs labelled with iron oxide nanoparti-
ing approach for cartilage repair and articular cartilage cles are more viable and migrate into injured cells. This pro-
which have very limited intrinsic regenerative capacity. The cedure is used to track implanted cells in experimental ani-
cell tracking driven a major step towards unraveling and mals. They found positive results in animals and suggested
improving the repair process of these therapies. They report that human trials were successful. The effectiveness of
superparamagnetic iron oxides for labelling human bone Resovist-labelled bone marrow stem cells was evaluated by
marrow-derived mesenchymal stem cells regarding effectiv- J. Guo et al. [56] in vivo, including their cerebral transplan-
ity, cell viability, long-term metabolic cell activity, chondro- tation in a model of Parkinson’s disease (PD) in rats using
genic differentiation and hBMSC secretion. They studied MRI, the effect and fate of transplanted cells in various dis-
limited intrinsic regenerative capacity of articular cartilage, ease models and the possibility of longer observation time.
making cell-based therapy a tempting approach for cartilage The iron oxide nanoparticles are more beneficial in devel-
repair. Cell tracking can be a major step towards unrav- oping new strategies of cell-based gene therapy. M. Radu et
eling and improving the repair process of these therapies. al. [57] investigated the effects of numerous anthropogenic
They studied superparamagnetic iron oxides for labelling and natural sources and reported the crystalline size of α-
hBMSCs concerning effectivity, cell viability, long-term Fe2 O3 nanoparticles is very effective and has a potential to
metabolic cell activity, chondrogenic differentiation and damage lung cells. The concentration of hematite (α-Fe2 O3 )
hBMSC secretion profile. nanoparticles during lipid peroxidation was 12.5 μg/mL. An

Fig. 5 The TEM micrographs of


the labelled and unlabelled
hBMSC-2 with
superparamagnetic iron oxide
nanoparticles. N nucleus, M
mitochondrion [52]
J Supercond Nov Magn

antioxidative system in MRC-5 lung fibroblast cell’s expo- more biological research interests. L. Mohammed et al. [61]
sure was tested for 1–3 days. Exposure to α-Fe2 O3 nanopar- have reviewed that the maghemite nanoparticles possessed
ticles increased lipid peroxidation by different percentages, all potential applications in both the environmental and
such as 81%, 189% and 110% after 1 day, 2 days and medical fields. Also, they mentioned that magnetic nanopar-
3 days, respectively, and they reported MRC-5 antioxidant ticles had not yet fully reached their optimal safety than
defense mechanisms not efficiently counteract the oxidative hematite and efficiency owing to the challenges faced when
stress induced by exposure to hematite nanoparticles and used in vivo. They were convinced by overcoming the short-
possible damaging effects could occur in human lung cells. comings through their article of improvement of magnetic
D. Horak et al. [58, 59] developed surface-modified iron targeted carriers for preclinical trials.
oxide nanoparticles by precipitation of Fe(II) and Fe(III) S. S. Eamegdool et al. [62] studied the comparison
salts with ammonium hydroxide, and they compared their between iron oxide nanoparticle labelling conditions and
results with those using two methods: in the first method, D- neural stem cell function, and they found that human
mannose solution (in situ coating) was used, and the second fetal neural precursor cells can be efficiently loaded with
method involved oxidation of sodium hypochlorite followed iron oxide nanoparticles at the optimum conditions. H.
by the addition of D-mannose solution (post-synthesis coat- L. Karlsson et al. [63] have made comparisons with the
ing). Also, rat bone marrow stromal cells (rBMSCs) were toxicity of nano- and micrometre particles of different
labelled with uncoated and D-mannose-modified iron oxide metal oxides. They tested the particles to cause cell death,
nanoparticles and with Endorem. The labelling efficiency mitochondrial damage and DNA damage, and oxidative
and the viability of both rat and human mesenchymal DNA lesions were evaluated after exposure of the human
stem cells labelled with Endorem, poly(L-lysine) (PLL)- cell line A549. This study showed that nanoparticles of
modified Endorem, uncoated γ -Fe2 O3 , D-mannose and metal oxide were much more toxic compared to micrometre
poly(N, N-dimethylacrylamide) (PDMAAm)-coated iron particles of metal oxide. One key mechanism may be the
oxide nanoparticles were compared. The coated magnetic ability of metal oxide to damage the mitochondria. In
iron oxide nanoparticles labelled cells better than did contrast, the micrometre particles of TiO2 caused more
Endorem. High relaxation rates and in vitro magnetic res- DNA damage compared to the nanoparticles, which is likely
onance imaging of cells labelled with coated nanoparticles explained by the crystal structures. The iron oxides showed
showed visible contrast compared with unlabelled cells or low toxicity and no clear difference between the different
cells labelled with Endorem. They reported surface modifi- particle sizes. In conclusion, nanoparticles are not always
cation of iron oxide nanoparticles with D-mannose, PLL and more toxic than micrometre particles, but the high toxicity
PDMAAm has a strong impact on cell viability. A. Tomi- of metal oxide nanoparticles shows that the nanolevel gives
taka et al. [60] tested the composite of gelatin iron oxide rise to the specific concern. M. Darroudi et al. [64] have
nanoparticles and co-cultured it with human bone marrow- reported a dose-dependent toxicity with the non-toxic effect
derived mesenchymal stem cells. They reported gelatin- of concentration below 62.5 μg/mL on studied in vitro
based iron oxide composites internalised and degraded cytotoxicity on neuro2A cells.
intracellularly on bone marrow-derived mesenchymal stem S. J. H. Soenen et al. [65] have found out that high
cells. The potential cytotoxic effects of superparamagnetic intracellular iron oxide nanoparticle concentrations are
iron oxide nanoparticles on bone marrow cells become more more effective to affect cell physiology in a concentration-
predictable in comparison of iron oxide nanoparticles with dependent manner. High levels of iron oxide cores affect
the other examined cells. Superparamagnetic iron oxide the actin cytoskeleton, formation and maturation of focal
nanoparticles are not toxic to the bone marrow used at low adhesion complexes and can affect protein expression
concentrations. The superparamagnetic iron oxide exposure levels. J. A. Frank et al. [66] made the validation of
has an impact in vivo on the viability and key functions of intracytoplasmic iron oxide in labelled cells by using simple
local bone marrow stromal cells (BMSCs) as investigated, Prussian blue staining. In this study, the lower uptake is
and they appear suitable for contrast enhancement in bone below the threshold of detection with Prussian blue stain.
marrow, suggesting that this necessitates future research. The HeLa cells were incubated with ferumoxide-PLL and
MION-46L-PLL under the different iron concentrations,
4.2 Cell Labelling and they observed incubation time with cells caused the
large numbers of intracytoplasmic Prussian blue-positive
The present review work was based on cell labelling and particles in cells labelled with ferumoxides versus those
cell binding with iron oxide nanoparticles. The physical labelled with MION-46L-PLL; then, the cells were co-
properties and different magnetic behaviours of iron oxide incubated at minimum 6 h to12 h, washed and evaluated
like semiconductors (hematite) and superparamagnetic iron by cell labelling using Prussian blue stain. They tested
oxide (maghemite and magnetite) nanoparticle generate the combination of FDA-approved dextran-coated iron
J Supercond Nov Magn

oxide MR contrast agents with tannic acids (TA) which iron oxide nanoparticles. M. Ma et al. [70] reported
resulted in the formation of ferumoxide-TA and MION- amino-superparamagnetic iron oxide nanoparticles can be
46L-TA complexes that allow a universal non-specific used as an MRI contrast agent and bi-functional labelling
method of magnetically labelling stem cells. The FDA- probe for neural stem cells. The table shows a brief
approved reagent dextran is used to mouse or human; in review of iron oxide nanoparticles on different parameters
this method of labelling, cells may facilitate the introduction such as preparation method, particle size, incubation time
of MR monitoring of the biodistribution of magnetically and specific testing of biological applications. It can be
labelled cells in clinical settings. M. Babic et al. [67] concluded from this review that the targeting transfer of iron
tested the PDMAAm-coated γ -Fe2 O3 nanoparticles were oxide nanoparticles with DOX plays a significant role in
obtained by the solution radical polymerisation of DMAAm enhancing the performance of cell labelling and cytotoxic
in the presence of maghemite nanoparticles obtained by activity. It was found that in the future nanodrug delivery
the chemical co-precipitation of Fe(II) and Fe(III) salts systems, the iron oxide nanoparticles can be optimised for
with ammonium hydroxide and subsequent oxidation with applications and are effective for cell labelling treatment.
sodium hypochlorite. The presence of PDMAAm coating
on the maghemite surface was confirmed by both elemental 4.3 Special Targeted Drug Delivery
analysis and FTIR spectroscopy. Cellular monitoring is
particularly important for evaluating cell-based therapies. In The specific biological treatments (human placenta, insulin
the study of C. Heyn et al. [68], MRI of superparamagnetic and retinal loci) with iron oxide nanoparticles are discussed
iron oxide-labelled cells has become a valuable tool for here shortly. An investigation was made by J. J. Faust
studying the in vivo trafficking of transplanted cells. et al. [71]; they developed the size-dependent effects of
Cellular detection with MRI is generally considered to be α-Fe2 O3 nanoparticles through an in vitro model of the
orders of magnitude less sensitive compared to positron human placenta. They predicted that the diameter of the
emission tomography, single photon emission-computed nanoparticle affected the viability of the epithelium. They
tomography or optical fluorescence microscopy techniques. found that exposure of different diameters would affect
They reported inconsistency with the observed increase the epithelium in different ways. Their study showed that
in the experimentally determined Kfsl value relative to the large diameters of 50 nm and 78 nm of α-Fe2 O3
theoretical prediction diffusion is expected to play some disrupted the junctional integrity of the epithelium and
role in the determination of fractional signal loss, given apoptosis. The α-Fe2 O3 nanoparticles of a smaller diameter
the comparable dimensions of free water self-diffusion of 15 nm have a little effect on the epithelium. They
distance and FIESTA signal refocusing bands. S. J. reported that gross changes as observed due to exposure
H. Soenen et al. [69] made in vitro labelling cells of large diameters were not observed for less-diameter
with iron oxide nanoparticles for a frequent practice in nanoparticles. The report of their study concluded that
biomedical research. The potential cytotoxicity of these the fetal/maternal interface was responsible for selecting
iron oxide nanoparticles remains an issue of debate. In shuttling nutrients to the placenta, which acted as a barrier
their present study, four different NP types (dextran- to the mother’s immune components, and permitting waste
coated Endorem, carboxydextran-coated Resovist, lipid- and gas exchange, junctional disruption or apoptosis of
coated magnetoliposomes (MLs) and citrate-coated very these cells in pregnancy complications, or resulted in
small iron oxide particles) were tested on a variety of fetal malnutrition. S. Correia Carreira et al. [72] studied
cell types, being C17.2 neural progenitor cells, PC12 and demonstrated the transport and uptake of iron oxide
rat pheochromocytoma cells and human blood outgrowth nanoparticles occurs in vitro in the BeWo b30 placental
endothelial cells. The MRI up to 500 cells/mL labelled with barrier model, and they found the effect of size on transport
very small iron oxide particles is required to efficiently and toxicity was less pronounced in this study archived by
visualise in an agar phantom in contrast to only 50 cells/mL 25 nm and 50 nm. Their report concluded the characteristics
for MLs and 200 cells/mL for Endorem and Resovist. driving toxicity and the placental barrier model were
Their results highlighted the importance of in-depth strongly recommended to allow more accurate extrapolation
cytotoxic evaluation of cell labelling studies as at non-toxic to specific biological applications. A. Kebede et al. [73]
concentrations, few of the particles appear to be less suitable studied chitosan and iron oxide nanocomposites loaded with
for the MR visualisation of labelled cells. The effects insulin, which resulted in reduction of blood glucose level
of iron oxide nanoparticles on cell homeostasis require a for different diabetic levels and achieved a 51% reduction
multidisciplinary approach where many parameters should in blood glucose level. The next discussion goes to specific
be analysed as iron oxide nanoparticles can lead to a iron oxide nanoparticles targeted to retinal loci by A. Yanai
wide variety of toxicological belongings and used as a et al. [74]; their reports produced therapeutic possibilities
model system for the further toxicological analysis of achievable with MSCs in diseased neurological tissues.
Table 1 Comparative review on iron oxide phases and its applications

S. no. Nanoparticles Preparation methods Particle size Duration Applications References

1 Leaf extract + iron oxide Co-precipitation 10–30 nm 24 h P. mirabilis and E.coli S. Arokiyaraj et al. [13]
2 Iron oxide Wet chemical method 239 nm 12 h S. aureus C. Narendhar et al. [15]
J Supercond Nov Magn

3 PVA + iron oxide Precipitation 4–9 nm 12–24 h S. aureus N. Tran et al. [16]
4 Hematite Direct heating 85–77 am 10–15 min A. hydrophila and E.coli M. Mohamed Rafi et al. [8]
5 Magnetite Co-precipitation 10–20 nm 45 min Antimicrobial activity E.coli M. Arakha et al. [9]
6 Hematite Penners and Koopal method 120 nm 24 h Antibacterial activity W. Zhang et al. [10]
7 GC + superparamagnetic Co-precipitation 8–9 nm 24 h E. coli B. S. Inbaraj et al. [11]
iron oxide
8 Hematite Chemical precipitation 26–98 nm 25–45 min S. aureus W. Zhang et al. [12]
9 Iron oxide Laser ablation technique 5–10 nm 9h S. aureus E. Fazio et al. [14]
10 Hematite Hydrothermal method 180 nm 2 days Hek293 cells H. Yan and B. Zhang [29]
11 Hematite Biosynthesis 15–30 nm 48 h HepG2 cells K. Rajendran et al. [30]
12 DMSA + superpramagnetic Thermal decomposition 15 nm 72 h MCF-7 cells M. Calero et al. [31]
iron oxide
13 Maghemite Laser pyrolysis 8–10 nm 4h Violamycine B1 and MCF-7 A. Marcu et al. [32]
14 Superparamagnetic iron oxide Co-precipitation 3–11 nm 120 h Tumour cell MF66-DOX S. Kossatz et al. [33]
15 Superparamagnetic iron oxide Lipid film rehydration 25–50 nm 2 weeks MCF-7 M. Rishikesh et al. [34]
16 Hematite Green synthesis 39 nm 48 h Anticancer MCF-7 and HBL-100 P. C. Nagajyothi et al. [35]
17 Superparamagnetic iron oxide Chemical co-precipitation 26–31 nm 48 h A549 cancer cell line D. Rehana et al. [36]
18 Iron oxide Precipitation 6–12 nm 12 h Antitumour B. Sangeetha and A.K. Kumaraguru [37]
19 Superparamagnetic iron oxide Co-precipitation 10–150 nm 20 min L-929 fibroblastic cells S. C. Hong et al. [38]
20 Superparamagnetic iron oxide Thermal decomposition 7–10 nm 16–96 h MCF-7 and HepG2 M. Kumar et al. [39]
21 OA + iron oxide Ultracentrifugation 2–11 nm 2–48 h MCF-7 and PC3 cells T. K. Jain et al. [40]
22 Maghemite Ultrasonication 20–100 nm 24 h Anti-HER2-PA tumour K. Kanazaki et al. [41]
23 C(RGDyK)-sperparamagnetic Co-precipitation 60–70 nm 75 h Tumour cells L. Guo et al. [42]
iron oxide
24 Maghemite Co-precipitation 54–86 nm 28 days Antitumour and H. Mackova et al. [43]
antimetastatic
25 Iron oxide Co-precipitation 20–60 nm 24 h MCF-7 cells A. H. Atta et al. [44]
26 Iron oxide Co-precipitation 86 nm 12 h Tumours M. Singh et al. [45]
27 Iron oxide Ultrafiltration process 150–250 nm 2 min MRI A. Masotti et al. [46]
28 Iron oxide Thermal decomposition 22 nm 24 h Radio-labelling and B. Bajaj et al. [47]
cancer therapy
29 Iron oxide Sonication 200–2000 nm 72 h Bone marrow cells S.-Y.-R. Paik et al. [51]
Table 1 (continued)

S. no. Nanoparticles Preparation methods Particle size Duration Applications References

30 Maghemite Commercially 145–260 nm 72 h Bone marrow cells B. Novotna et al. [52]


available (CA)
31 Superparamagnetic iron oxide CA 73–273 μm 5–44 days hBMSCs G. M. van Buul et al. [53]
32 Iron oxide CA 20–30 nm 2–47 days MSCs Jendelova et al.
33 Superparamagnetic iron oxide CA 60 nm 1–4 weeks MSCs and ESCs P. Jendelova et al. [54, 55]
34 Superparamagnetic iron oxide CA 4–60 nm 1–12 weeks PD-BMSCs J. Guo et al. [56]
35 Hematite Laser ablation technique 40–60 nm 24–72 h MRC-5 cells M. Radu et al. [57]
36 D -Mannose + iron oxide Co-precipitation 6–7 nm 2–3 days rMSCs Horak et al.
37 Maghemite Co-precipitation 10 nm 1–5 weeks rMSCs D. Horak et al. [58, 59]
38 Iron oxide Co-precipitation 1–5 nm 7–44 days rMSCs and MRI A. Tomitaka et al. [60]
39 Superparamagnetic iron oxide Massart method 10–15 nm 1–7 days hNPCs S. S. Eamegdool et al. [62]
40 Iron oxide Sonication 30–60 nm 4–24 h Human alveolar epithelial cell line H. L. Karlsson et al. [63]
41 Superparamagnetic iron oxide Co-precipitation 30–40 nm 24 h Neuro2A cells M. Darroudi et al. [64]
42 Iron oxide CA 8–150 nm 72 h Cell viability S. J. H. Soenen et al. [65]
43 Superparamagnetic iron oxide CA 530–575 nm 2–48 h MSCs + HeLa cells J. A. Frank et al. [66]
44 Superparamagnetic iron oxide Co-precipitation 50–170 nm rMSCs + hMSCs M. Babic et al. [67]
45 Superparamagnetic iron oxide CA 100 μm 5 days THP-1 cell types C. Heyn et al. [68]
46 Iron oxide CA 3–14 nm 1–7 days C17.2 and PC12 cells S. J. H. Soenen et al. [69]
47 Superparamagnetic iron oxide Co-precipitation 10–60 nm 60 min Neural stem cells M. Ma et al. [70]
48 Hematite Penners and Koopal method 15–78 nm 12–72 h Human placenta J. J. Faust et al. [71]

49 Iron oxide – 25–50 nm 4–48 h BeWo b30 placental cell barrier S. Correia Carreira et al. [72]
50 Iron oxide Laser ablation – 24 h Insulin A. Kebede et al. [73]
51 PEG + magnetite Co-precipitation 70–140 nm 30 min Reduction of cytochrome c M. Anindita et al. [79]
52 Superparamagnetic iron oxide 200 nm 4 weeks MSCs and retinal loci A. Yanai et al. [74]
53 Iron oxide 2h Staphylococcus aureus M. P. Kumar et al. [17]
54 Iron oxide 22–26 mm Bacillus cereus and Klebsiella pneumoniae A. S. Ahmed et al. [18]
55 Cynometra ramiflora with iron oxide E. coli and S. epidermidis G. Silvia et al. [19]
56 Gallic acid + magnetite Post-synthesis methods 5–8 nm Bacterial and fungal species S. S. Tawab et al. [20]
J Supercond Nov Magn
J Supercond Nov Magn

They suggested that iron oxide-targeted cell delivery for a binding is due to the hybrid and multifunctional structure of
limited area of tissue leads to adverse collateral damage by iron oxide nanoparticles and its improved ability to employ
direct injection of cells. specific PEG silane molecular binding. The review study
The iron oxide nanoparticles doped with different projected that the iron oxide materials are eco-friendly and
dopants were used to enhance the chemical and physical less toxic for biological studies. Recently, cancer diagno-
properties. The dopants such as amino covalent binding sis and therapies advanced into the next level of research.
[29, 38, 63], agarose [8], arginine [35], citric acid [31], On that, the energy relay approaches are implemented in
c(RGDyK) [35], DMSA [24], DMAA [60], dextran [8, lifetime imaging and provide a low toxicity. The risk of ther-
72], D-mannose [51], ethylene glycol [71], glycol chitosan mal accumulation and tissue damage as well as damages
[11], gelatin [8, 53], L-arginine [28], PVP [22, 75], of healthy tissues is also reduced compared to that using
polysaccharide [8], PVA [16], PEI [39, 70], PEG [27, 38, 68, ordinary intensity imaging methods of cancer chemother-
71], PVC [76], peptide [32], polymer [60, 73], silane [69], apy, rendering the use of hyperthermia/thermal treatment
saccharide [74], leaf extracts [13, 28, 30] and tannins [77] less a concern [78]. These technologies have high transfor-
for different biological applications are listed in Table 1. mational diagnoses for a wide range of clinical application
The systematic studies on the relationships between the and biomedical research studies.
nanoparticle size and structures of iron oxide or different
morphologies play an important role in the early diagnosis
and post treatments. References

1. Cornell, R.M., Schwertmann, U.: VCH. Weinheim (1996)


2. Vayssieres, L., Sathe, C., Butorin, S.M., Smith, D.K., Nordgren,
5 Conclusion J., Guo, J.H.: Adv. Mater. 17, 2320–2323 (2005)
3. Bhushan, B., Luo, D., Schricker, S.R., Sigmund, W., Zauscher, S.:
The property of iron oxide in nanorange shows the sig- Springer, Berlin 12, 355–388 (2014)
nificant performance as biological applications, which is 4. Mohapatra, M., Anand, S.: Int. J. Eng. Sci. Technol. 8, 127–146
(2010)
evidently confirmed from this review analysis. It helps to 5. Sharifi-Rad, J., Hoseini-Alfatemi, S.M., Sharifi-Rad, M., da Silva,
improve iron oxide property by adding dopants and surfac- J.A.T.: 3 Biotechnol. 5, 677–684 (2015)
tant, which may be a supporting aid that makes a big impact 6. Zorofchian Moghadamtousi, S., Abdul Kadir, H., Hassandarvish,
on specified target-based biomedical applications. The co- P., Tajik, H., Abubakar, S., Zandi, K.: Biomed. Res. Int. 2014, 1–
12 (2014)
precipitation method is the more appropriate than the other 7. Hajipour, M.J., Fromm, K.M., Ashkarran, A.A., de Aberasturi,
preparation processes to prepare iron oxide nanoparticle. D.J., de Larramendi, I.R., Rojo, T., Serpooshan, V., Parak, W.J.,
The response of iron oxide in antimicrobial activities was Mahmoudi, M.: Trends Biotechnol. 30(10), 1–13 (2012)
8. Mohamed Rafi, M., Syed Zameer Ahmed, K., Prem Nazeer, K.,
identified to be more effective on certain types of bacteria:
Siva Kumar, D., Tamil Selvan, M.: Appl. Nanosci. 5(4), 515–520
Bacillus subtilis, Bacillus cereus, Aeromonas hydrophila, (2015)
Escherichia coli and Staphylococcus aureus. The antimicro- 9. Arakha, M., Pal, S., Samantarrai, D., Panigrahi, T.K., Mallick, B.C.,
bial efficiency against the bacterial pathogens due to iron Pramanik, K., Mallick, B., Jha, S.: Sci. Rep. 5(14813), 1–12 (2015)
10. Zhang, W., Rittmann, B., Chen, Y.: Environ. Sci. Technol. 45,
oxide nanoparticles is more effective for Staphylococcus 2172–2178 (2011)
aureus and less for Aeromonas hydrophila. Similarly, the 11. Inbaraj, B.S., Tsai, T.-Y., Chen, B.-H.: Sci. Technol. Adv. Mater.
role of iron oxide in anticancer activity emphasised that the 13(1), 1–9 (2012)
level of toxicity was due to the dosage level of iron oxide 12. Zhang, W., Hughes, J., Chena, Y.: Appl. Environ. Microbiol.
78(11), 3905–3915 (2012)
nanoparticles. The iron oxide nanoparticle, in this review, 13. Arokiyaraj, S., Saravanan, M., Udaya Prakash, N.K., Valan Arasu,
was found to play a role in analysis and cytotoxicity level in M., Vijayakumar, B., Vincent, S.: Mater. Res. Bull. 48, 3323–
cancer treatment, which is due to the iron oxide synthesised 3327 (2013)
14. Fazio, E., Santoro, M., Lentini, G., Franco, D., Guglielmino,
from different methods and analyses. The iron oxide parti-
S.P.P., Neri, F.: Colloids Surf. A Physicochem. Eng. Asp. 490,
cle size and incubation time play a main role on targeted 98–103 (2016)
drug delivery systems. The magnetic (maghemite and mag- 15. Chandrasekar, N., Mohan Kumar, K.M., Balasubramnian, K.S.,
netite) properties of iron oxide nanoparticles are appropriate Karunamurthy, K., Varadharajan, R.: Dig. J. Nanomater.
Biostruct. 8(2), 765–775 (2013)
to anticancer, antitumour and cell labelling. The semicon- 16. Tran, N., Mir, A., Mallik, D., Sinha, A., Nayar, S., Webster, T.J.:
ducting behaviour of iron oxide (hematite) nanoparticle is Int. J. Nanomedicine 5, 277–283 (2010)
apt and effective on antibacterial and antimicrobial stud- 17. Manna, P.K., Nickel, R., Wroczynskyj, Y., Yathindranath, V., Li,
ies from this review analysis. The predominant cancer and J., Liu, S., Thliveris, J.A., Klonisch, T., Miller, D.W., van Lierop,
J.: Langmuir 34(8), 2748–2757 (2018)
tumour cell controlled by iron oxide nanoparticle are iden- 18. Ahmed Ansari, S., Oves, M., Rukhsana, S., Khan, A., Ahmad, S.I.,
tified from this review study as Hek293, HepG2, MCF-7 Jafri, M.A., Kashif Zaidi, S., Husein Alqahtani M.: Pol. J. Chem.
and rMSCS cells. Another typical role in molecular cell Technol. 19(4), 110–115 (2017)
J Supercond Nov Magn

19. Groiss, S., Selvaraj, R., Varadavenkatesan, T., Vinayagam, R.: J. 46. Masotti, A., Pitta, A., Ortaggi, G., Corti, M., Innocenti, C.,
Mol. Struct. 1128, 572–578 (2017) Lascialfari, A., Marinone, M., Marzola, P., Daducci, A., Sbarbati,
20. Shah, S.T., Yehye, W.A., Saad, O., Simarani, K., Chowdhury, A., Micotti, E., Orsini, F., Poletti, G., Sangregorio, C.: Magn.
Z.Z., Alhadi, A.A., Al-Ani, L.A.: Nanomaterials 7(10), 306/1– Reson. Mater. Phys. 8(22), 77–87 (2009)
306/17 (2017) 47. Bajaj, B., Malhotra, B.D., Choi, S.: Thin Solid Films 519, 1219–
21. Karki, H.P., Ojha, D.P., Joshi, M.K., Kim, H.J.: Appl. Surf. Sci. 1223 (2010)
435, 599–608 (2018) 48. Kandasamy, G., Maity, D.: Int. J. Pharm. 2, 191–218 (2015)
22. Abdulhady, Y.A.M., El-Shazly, M.M., El-Kased, R.F.: J. Environ. 49. Urdzikova, L., Jendelova, P., Glogarova, K., Burian, M., Hajek,
Sci. Health A Tox. Hazard. Subst. Environ. Eng. 53(3), 205–212 M., Sykova, E.: J. Neurotrauma 23(9), 1379–1391 (2006)
(2018)
50. Turnovcova, K., Ruzickova, K., Vanecek, V., Sykova, E.,
23. Bhushan, M., Kumar, Y., Periyasamy, L., Viswanath, A.K.: Appl.
Jendelova, P.: Cytotherapy 7, 874–885 (2009)
Nanosci. 8(1–2), 137–153 (2018)
24. Malekigorji, M., Curtis, A.D.M., Hoskins, C.: J Nanomed Res. 51. Paik, S.-Y.-R., Kim, J.-S., Shin, S.J., Ko, S.: Int. J. Mol. Sci. 16,
1(1), 0004 (2014) 22243–22257 (2015)
25. Nguyen, X.P., Tran, D.L., Ha, P.T., Pham, H.N., Mai, T.T., Pham, 52. Novotna, B., Jendelova, P., Kapcalova, M., Rossner, P. Jr.,
H.L., Le, V.H., Do, H.M., Phan, T.B.H., Pham, T.H.G., Nguyen, Turnovcova, K., Bagryantseva, Y., Babic, M., Horak, D., Sykova,
D.T., Hoang, T.M.N., Lam, K., Nguyen, T.Q.: Adv. Nat. Sci.: E.: Toxicol. Lett. 210, 53–63 (2012)
Nanosci. Nanotechnol. 3(033001), 1–13 (2012) 53. van Buul, G.M., Kotek, G., Wielopolski, P.A., Farrell, E., Bos,
26. Huang, J.S.-H., Juang, R.-S.: J. Nanopart. Res. 13, 4411–4430 P.K., Weinans, H., Grohnert, A.U., Jahr, H., Verhaar, J.A.N.,
(2011) Krestin, G.P., van Osch, G.J.V.M., Bernsen, M.R.: PLoS ONE 2,
27. Chen, H., Zhen, Z., Todd, T., Chu, P.K., Xie, J.: Mater. Sci. Eng. 1–9 (2011)
R 74, 35–69 (2013) 54. Jendelova, P., Herynek, V., Urdzikova, L., Glogarova, K.,
28. Bakhtiary, Z., Saei, A.A., Hajipour, M.J., Raoufi, M., Vermesh, Kroupova, J., Andersson, B., Bryja, V., Burian, M., Hajek, M.,
O., Mahmoudi, M.: Nanomedicine: NBM 12, 287–307 (2016) Sykova, E.: J. Neurosci. Res. 76, 232–243 (2004)
29. Yan, H., Zhang, B.: Mater. Lett. 65, 815–817 (2011) 55. Jendelova, P., Herynek, V., Urdzikova, L., Glogarova, K.,
30. Rajendran, K., Karunagaran, V., Mahanty, B., Sen, S.: Int. J. Biol. Kroupova, J., Andersson, B., Bryja, V., Burian, M., Hajek, M.,
Macromol. 74, 376–381 (2015) Sykova, E.: Magn. Reson. Med. 50, 767–776 (2003)
31. Calero, M., Chiappi, M., Lazaro-Carrillo, A., Rodrı́guez. MJ, 56. Guo, J., Shen, J.-K., Wang, L., Xiao, L., Zhang, R.-J., Luo, W.-F.,
Chichón, F.J., Crosbie-Staunton, K., Prina-Mello, A., Volkov, Y., Gong, Z.-G., Sun, J., Xu, H., Sirois, P., Li, K.: Appl. Biochem.
Villanueva, A., Carrascosa, J.L.: J. Nanobiotechnol. 13(16), 1–15 Biotechnol. 163, 636–648 (2011)
(2015) 57. Radu, M., Munteanu, M.C., Petrache, S., Serban, A.I., Dinu, D.,
32. Marcu, A., Pop, S., Dumitrache, F., Mocanu, M., Niculite, C.M., Hermenean, A., Sima, C., Dinischiotu, A.: Acta Biochim. Pol. 57,
Gherghiceanu, M., Lungu, C., Fleaca, C.P., Ianchis, R., Barbut, A., 1–6 (2010)
Grigoriu, C., Morjan, I.: Appl. Surf. Sci. 281, 60–65 (2013)
58. Horak, D., Babic, M., Jendelova, P., Herynek, V., Trchova, M.,
33. Kossatz, S., Grandke, J., Couleaud, P., Latorre, A., Aires, A.,
Pientka, Z., Pollert, E., Hajek, M., Sykova, E.: Bioconjug. Chem.
Crosbie-Staunton, K., Ludwig, R., Dahring, H., Ettelt, V., Lazaro-
18, 635–644 (2007)
Carrillo, A., Calero, M., Sader, M., Courty, J., Volkov, Y.,
59. Horak, D., Babic, M., Jendelova, P., Herynek, V., Trchova, M.,
Prina-Mello, A., Villanueva, A., Somoza, A., Cortajarena, A.L.,
Likavcanova, K., Kapcalova, M., Hajek, M., Sykova, E.: J. Magn.
Miranda, R., Hilger, I.: Breast Cancer Res. 17(66), 1–17 (2015)
34. Sawant, R.M., Sawant, R.R., Gultepe, E., Nagesha, D., Magn. Mater. 321, 1539–1547 (2009)
Papahadjopoulos-Sternberg, B., Sridhar, S., Torchilin, V.P.: J. 60. Tomitaka, A., Jo, J.-I., Aoki, I., Yasuhiko, T.: Inflamm. Regen.
Nanopart Res. 11, 1777–1785 (2009) 34(1), 45–55 (2014)
35. Nagajyothi, P.C., Pandurangan, M., Kim, D.H., Sreekanth, T.V.M., 61. Mohammed, L., Gomaa, H.G., Ragab, D., Zhu, J.: Particuology
Shim, J.: J. Clust. Sci. 1, 245–257 (2017) 30, 1–14 (2017)
36. Rehana, D., Haleel, A.K., Rahiman, A.K.: J. Chem. Sci. 7, 1155– 62. Eamegdool, S.S., Weible, M.W. ., Pham, B.T.T., Hawkett, B.S.,
1166 (2015) Grieve, S.M., Chan-ling, T.: Biomaterials 35, 5549–5564 (2014)
37. Boulieu Sangeetha, N., Kumaraguru, A.K.: Int. J. Pharm. Pharm. 63. Karlsson, H.L., Gustafsson, J., Cronholm, P., Moller, L.: Toxicol.
Sci. 7(2), 469–476 (2015) Lett. 188, 112–118 (2009)
38. Hong, S.C., Lee, J.H., Lee, J., Kim, H.Y., Park, J.Y., Cho, J., Lee, 64. Darroudi, M., Hakimi, M., Goodarzi, E., Oskuee, R.K.: Ceram.
J., Han, D.W.: Int. J. Nanomed. 6, 3219–3231 (2011) Int. 40(9), 14641–14645 (2014)
39. Kumar, M., Singh, G., Sharma, S., Gupta, D., Bansal, V., Arora, 65. Soenen, S.J.H., Himmelreich, U., Nuytten, N., De Cuyper, M.:
V., Bhat, M., Srivastava, S.K., Sapra, S., Kharbanda, S., Dinda, Biomaterials 32, 195–205 (2011)
A.K., Singh, H.: Nanoscale 6, 14473–14483 (2014) 66. Frank, J.A., Miller, B.R., Arbab, A.S., Zywicke, H.A., Jordan,
40. Jain, T.K., Morales, M.A., Sahoo, S.K., Leslie-Pelecky, D.L., E.K., Lewis, B.K., Bryant, L.H., Bulte, J.W.M.: Radiology
Labhasetwar, V.: Mol. Pharm. 2(3), 194–205 (2005) 228(2), 480–487 (2003)
41. Kanazaki, K., Sano, K., Makino, A., Shimizu, Y., Yamauchi, F.,
67. Babic, M., Horak, D., Jendelova, P., Glogarova, K., Herynek, V.,
Ogawa, S., Ding, N., Yano, T., Temma, T., Ono, M., Saji, H.: Biol.
Trchova, M., Likavcanova, K., Lesny, P., Pollert, E., Hajek, M.,
Med. 11(8), 2051–2060 (2015)
Sykova, E.: Bioconjug. Chem. 20(2), 283–294 (2009)
42. Guo, L., Ding, W., Zheng, L.-M.: J. Nanopart. Res. 15, 1720
(2013) 68. Heyn, C., Bowen, C.V., Rutt, B.K., Foster, P.J.: Magn. Reson.
43. Mackova, H., Horak, D., Donchenko, G.V., Andriyaka, V.I., Med. 53, 312–320 (2005)
Palyvoda, O.M., Chernishov, V.I., Chekhun, V.F., Todor, I.N., 69. Soenen, S.J.H., Nuytten, N., De Meyer, S.F., De Smedt, S.C., De
Kuzmenko, O.I.: J. Magn. Magn. Mater. 380, 125–131 (2015) Cuyper, M.: Small 6(7), 832–842 (2010)
44. Atta, A.H., El-ghamry, M.A., Hamzaoui, A., Refat, M.S.: J. Mol. 70. Ma, M., Zhan, Y., Shen, Y., Xia, X., Zhang, S., Liu, Z.: J.
Struct. 1086, 246–254 (2015) Nanopart. Res. 13, 3249–3257 (2011)
45. Singh, M., Sviridenkova, N., Timur, N., Savchenko, A., Shetinin, 71. Faust, J.J., Zhang, W., Chen, Y., Capco, D.G.: Cell Biol. Toxicol.
I., Majouga, A.: J. Clust. Sci. 4, 1383–1393 (2016) 30, 31–53 (2014)
J Supercond Nov Magn

72. Correia Carreira, S., Walker, L., Paul, K., Saunders, M.: The 75. Qi, M., Zhang, K., Li, S.: New J. Chem. 40(5), 4480–4491 (2016)
toxicity, transport and uptake of nanoparticles in the in vitro 76. Mukhopadhyay, A., Joshi, N., Chattopadhyay K., De, G.: ACS
BeWo b30 placental cell barrier model used within nanoTEST. Appl. Mater. Interfaces 4(1), 142–149 (2012)
Nanotoxicology 9(S1), 66–78 (2015) 77. Barrera, C., Herrera, A.P., Bezares, N.: J. Colloid Interface Sci. 1,
73. Kebede, A., Singh, A.K., Rai, P.K., Giri, N.K., Rai, A.K., Watal, 40–50 (2012)
G., Gholap, A.V.: Lasers Med. Sci. 28, 579–587 (2013) 78. Fan, Y., Wang, P., Lu, Y., Wang, R., Zhou, L., Zheng, X., Li, X.,
74. Yanai, A., Hafeli, U.O., Metcalfe, A.L., Soema, P., Addo, L., Piper, J.A., Zhang, F.: Nat. Nanotechnol. 1–6 (2018)
Gregory-Evans, C.Y., Po, K., Shan, X., Moritz, O.L., Gregory- 79. Anindita, M., Nidhi, J., Krishnanan-da, C.: ACS Appl. Mater.
Evans, K.: Cell Transplant. 21, 1137–1148 (2012) Interfaces 4(1), 142–149 (2012)

You might also like