Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Arabian Journal of Chemistry (2020) 13, 2287–2308

King Saud University

Arabian Journal of Chemistry


www.ksu.edu.sa
www.sciencedirect.com

REVIEW ARTICLE

Green biosynthesis of superparamagnetic magnetite


Fe3O4 nanoparticles and biomedical applications in
targeted anticancer drug delivery system: A review
Yen Pin Yew a, Kamyar Shameli a,*, Mikio Miyake b,
Nurul Bahiyah Bt Ahmad Khairudin a, Shaza Eva Bt Mohamad a, Takeru Naiki c,
Kar Xin Lee a

a
Department of Environmental Engineering and Green Technology, Malaysia-Japan International Institute of Technology,
Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100 Kuala Lumpur, Malaysia
b
Japan Advanced Institute of Science and Technology (JAIST), Japan
c
Department of Biomedical Engineering, Faculty of Engineering, Okayama University of Science, Japan

Received 21 February 2018; accepted 18 April 2018


Available online 26 April 2018

KEYWORDS Abstract This review discussed about the green biosynthesis of magnetite nanoparticles (Fe3O4-
Green biosynthesis; NPs) and the biomedical applications, which mainly focus on the targeted anticancer drug delivery.
Superparamagnetic; Fe3O4-NPs have been studied and proved that Fe3O4-NPs can be used in various fields of application,
Magnetite nanoparticle; due to ‘‘superparamagnetic” property that Fe3O4-NPs possessed. In targeted drug delivery system,
Anticancer; drug loaded Fe3O4-NPs can accumulate at the tumor site by the aid of external magnetic field. This
Targeted drug delivery can increase the effectiveness of drug release to the tumor site and vanquish cancer cells without harm-
ing healthy cells. In order to apply Fe3O4-NPs in human body, Fe3O4-NPs have to be biocompatible
and biodegradable to minimize the toxicity. So, green biosynthesis plays a crucial role as the biosyn-
thesized Fe3O4-NPs is safe to be consumed by human because the materials used are from biological
routes, such as plant extract and natural polymer. However, biosynthesis using plant extract is the
most popular among them all as plant extract can act as both reducing and stabilizing agents in the
synthesizing process of nanoparticles. This approach is not merely simple, yet economic and less waste
production, which is environmental friendly. Several biomedical applications of Fe3O4-NPs are
included in this review, but anticancer drug delivery study is discussed in detail. The criteria for
Fe3O4-NPs to be used as drug delivery vehicle are discussed so as to study the optimum condition
of Fe3O4-NPs in drug delivery application. Many researches showed the promising results of
* Corresponding author.
E-mail addresses: kamyarshameli@gmail.com (K. Shameli), r-bahiah@utm.my (N.B.B. Ahmad Khairudin), shaza@utm.my (S.E.B. Mohamad),
tnaiki@bme.ous.ac.jp (T. Naiki).
Peer review under responsibility of King Saud University.

Production and hosting by Elsevier

https://doi.org/10.1016/j.arabjc.2018.04.013
1878-5352 Ó 2018 Production and hosting by Elsevier B.V. on behalf of King Saud University.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
2288 Y.P. Yew et al.

Fe3O4-NPs in treating cancer cells via in vitro study. Hence, this review is significant which summarize
the vital points of Fe3O4-NPs in targeted anticancer drug delivery system. Conclusions have been
made according to the literature reviewed and some points of view were proposed for future study.
Ó 2018 Production and hosting by Elsevier B.V. on behalf of King Saud University. This is an open access
article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2288
1.1. Superparamagnetic iron oxide nanoparticles (magnetite nanoparticles). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2288
1.2. Green biosynthesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2289
2. Plant extract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2290
2.1. Plant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2290
2.2. Marine plant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2290
2.3. Leaf. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2290
2.4. Fruit peel . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2292
2.5. Seed . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2292
2.6. Fruit . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2293
2.7. Stolon and root . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2293
2.8. Gum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2293
2.9. Plant waste. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2294
3. Other green materials. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2294
3.1. Glucose . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2294
3.2. Polysaccharides. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2294
3.3. Clay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2296
4. Biomedical applications of Fe3O4-NPs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2297
5. Utility of Fe3O4-based nanoparticles as drug delivery vehicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2298
5.1. Criteria of Fe3O4-based nanoparticles to be used in drug delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2298
5.1.1. Superparamagnetic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2298
5.1.2. Shape of nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2299
5.1.3. Size of nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2299
5.1.4. Surface modification and stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2299
5.1.5. Drug loading and release . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2300
5.2. Targeted anticancer drug delivery. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2300
6. Conclusion and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2303
6.1. Future works . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2304
Declarations of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2304
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2304
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2304

1. Introduction NPs, particularly biomedical application in drug delivery will


be discussed for better understanding of their uses in this mod-
In recent times, researchers have discovered the huge potential ern technology.
behind nanotechnology and ever since it has played a vital role
in this world. Nanotechnology is assuredly gaining in popular- 1.1. Superparamagnetic iron oxide nanoparticles (magnetite
ity owing to the benefits and potential it can provide to human. nanoparticles)
In the past decades, nanotechnology started with material
industry yet its credibility remained low (Hanus and Harris, Iron oxides exist in many forms in nature, such as iron(III)
2013). As time goes by, nanotechnology becomes progressively oxide (FeO), hematite (a-Fe2O3) and maghemite (c-Fe2O3).
influential in various fields of applications, from environmen- In fact, magnetite is the most popular and useful iron oxide
tal to food industry, now even develop in biomedical field which has been employed in various field of applications. Mag-
which shows great potential in the future clinics (Beloqui netite is a kind of mineral and it is one of the most common
et al., 2016). natural occurring iron oxide with chemical formula Fe3O4.
To date, there is still no detailed study of green synthesis of The crystal structure of magnetite shows an inverse spinel pat-
Fe3O4-NPs using biological routes and usage of Fe3O4-NPs in tern with alternating and tetrahedral-octahedral layers. This
biomedical field. Therefore, this review was conducted to high- means that Fe2+ species of Fe3O4 occupy half of the octahe-
light the green biosynthesis of Fe3O4-NPs using plant extract dral lattice sites as a result of greater ferrous crystal field stabi-
and other biological materials. Then, application of Fe3O4- lization energy (CFSE). On the other hand, Fe3+ species
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2289

occupy the other octahedral lattice sites and all the tetrahedral et al., 2017; Narayanan et al., 2011). All these researches give
lattice sites (Blaney, 2007). promising results and provide a platform for Fe3O4-NPs which
Besides, magnetite is also very well known as the strongest their unique features offer tremendous potential for their vast
magnetic mineral on earth (Harrison et al., 2002). This fasci- application.
nating characteristic attracts much attention from researchers
all around the world. Magnetite has a property that it is ferro- 1.2. Green biosynthesis
magnetic at room temperature and the Curie temperature is
850 K (Teja and Koh, 2009). However, the magnetic behavior Generally, green synthesis of nanotechnology means the syn-
of Fe3O4-NPs depends strongly on the synthesis method. thesizing of nanomaterials or nanoparticles without using haz-
Additionally, the size and morphology of magnetite crystal ardous chemicals that produce toxic by-products. In other
play an important role which influence the magnetic properties words, green method is an eco-friendly technique to synthesize
of magnetite (Lin et al., 2006; Song et al., 2012). Hence, opti- nanoparticles where it is not harmful to the environment and
mum parameters of Fe3O4-NPs has to be ascertained for better human health. It is true that conventional methods can fabri-
application. cate nanoparticles in huge quantities with desired morphology
Superparamagnetic nanoparticles is so famous nowadays and size. However, these methods require high cost produc-
because of the properties possessed, where the nanoparticles tion, complicated and outdated procedures (Patra and Baek,
are magnetized up to their saturation magnetization when an 2014). In contrast to the conventional chemical and physical
external magnetic field is applied, yet they will not show any methods, green synthesis has many benefits such as facile, sim-
magnetic interaction once the magnetic field is eliminated ple manufacturing procedure, fast, economic and less waste
(Wahajuddin, 2012). Surprisingly, Fe3O4-NPs exhibit this production.
interesting behavior too. Apart from that, Fe3O4-NPs are bio- Green biosynthesis of nanoparticles employs the bottom-up
compatible, biodegradable and potentially non-toxic to human approach where the metal atoms assemble to form clusters and
(Zhang et al., 2013; Zhao et al., 2009). These characteristics then eventually the nanoparticles. The biological compounds
show a great potential of Fe3O4-NPs in future biomedical present in green materials may act as both reducing and cap-
applications. ping agents that can stabilize the nanoparticles during the syn-
It is well recognized that Fe3O4-NPs are advantageous to thesis process. This can control the size and shape of the
our lives. However, different properties of Fe3O4-NPs con- nanoparticles which can be used in various applications.
tribute to their versatility in different applications. For exam- Fig. 2 shows the simple process of nanoparticles synthesis
ple, the optimal size of nanoparticles is 50 nm in diameter for where the materials needed are metal salt (precursor) and green
efficient endocytosis in drug delivery application (Bamrungsap substrate only. Various parameters such as concentration of
et al., 2012). Therefore, a lot of synthesis methods have been metal salt, concentration of green substrate, time and temper-
reported which can synthesize Fe3O4-NPs with desired proper- ature for reaction and pH of the solution can be altered during
ties. For instance, co-precipitation method (Petcharoen and the nanoparticles synthesis process in order to obtain proper-
Sirivat, 2012; Shen et al., 2014), sol-gel method (Lemine ties that are needed for respective applications.
et al., 2012), hydrothermal synthesis (Haw et al., 2010; Li Green biosynthesized Fe3O4-NPs can possess better charac-
et al., 2014b; Li et al., 2013a), solid state synthesis (Paiva teristics, such as higher biocompatibility and biodegradability,
et al., 2015), flame spray synthesis (Kumfer et al., 2010), ther- compared to physically synthesized Fe3O4-NPs. Hence, they
mal decomposition (Chin et al., 2011), solvothermal (Luo can be utilized in biomedical application due to the special
et al., 2015) and so on. All these physical and chemical meth-
ods arouse numerous issues comprising high production cost,
use of toxic chemicals and yield of hazardous by-products
(Hussain et al., 2016). Thus, synthesizing nanoparticles using
green method is introduced lately to cope with the problems
that caused by conventional approaches.
As discussed, Fe3O4-NPs hold several fascinating charac-
teristics, such as superparamagnetic behavior, biocompatible
and biodegradable, hence numerous studies have been done
in order to maximize the potential usage of Fe3O4-NPs in var-
ious fields of applications. The study of Fe3O4 nanofluid on
thermal conductivity and viscosity with the presence of exter-
nal magnetic source and electric field, has also gaining popular
in heat transfer applications (Sheikholeslami and Sadoughi,
2017; Sheikholeslami and Shehzad, 2017, 2018;
Sheikholeslami and Vajravelu, 2017). Fig. 1 shows the possible
applications of Fe3O4-NPs to be used in the fields of catalyst
(Azarifar et al., 2016; Gawande et al., 2013; Wang et al.,
2015), water remediation (heavy metal ion removal)
(Hardani et al., 2015; Venkateswarlu et al., 2014b;
Venkateswarlu and Yoon, 2015b), lithium ion battery (He
et al., 2016; Liu et al., 2017), magnetic storage media (El
Ghandoor et al., 2012), and last but not least, the biomedical
applications (Kandelousi and Ellahi, 2015; Karimzadeh Fig. 1 Applications of magnetite nanoparticles (Fe3O4-NPs).
2290 Y.P. Yew et al.

2.2. Marine plant

Marine plant, which is also known as seaweed or algae, can be


utilized to synthesize Fe3O4-NPs. Seaweed is important to
marine life because seaweed provides food and habitat where
it can be found in a lot of countries in South-East Asia. It is
well known that seaweed is a food source in our daily life
due to their abundance of lipids, minerals, some vitamins
and particular bioactive substances, such as proteins, poly-
phones and polysaccharides which have the potential in medi-
cal uses. Besides, marine algae can be divided into two groups,
Fig. 2 Nanoparticles synthesis process. which are microalgae and macroalgae. Macroalgae (seaweed)
are plant-like organisms and normally they are used in
nanoparticles synthesis. The phytochemicals presence in sea-
surface coating of green materials, which is not only non-toxic weed can act as metal-reducing agents and capping agents to
and biocompatible yet also allow targeted drug delivery with supply a robust coating on the metal nanoparticles in a single
Fe3O4-NPs localization at particular area. Toxicity towards step. Mahdavi et al. studied on the brown seaweed (Sargassum
human body can be minimized because the green materials muticum) in synthesizing Fe3O4-NPs. The synthesis procedure
used for synthesizing Fe3O4-NPs are safe to be consumed, was very simple by mixing FeCl3 solution to the brown sea-
and thus it would be beneficial in biomedical applications. weed extract, where Fe3O4-NPs were immediately produced
Besides, Fe3O4-NPs can conjugate with drugs, enzymes or pro- with the reduction process. The major components such as sul-
teins which can be directed to targeted tissue, organ or tumor phate, hydroxyl and aldehyde group presence in seaweed may
with the aid of external magnetic field, or can be heated in lead to the reduction of Fe3+ and stabilize the nanoparticles.
alternating magnetic fields for hyperthermia treatment Meanwhile, the pH of the solution decreased during the syn-
(Mahdavi et al., 2013a). thesis process, which indicated the participation of –OH group
in the reduction process. There is also a possibility that the sul-
2. Plant extract phate group reduce the metal ions by oxidation of aldehyde
groups in the molecules to carboxylic acids. The cubic shape
In this review, green biosynthesis of Fe3O4-NPs will be of Fe3O4-NPs were produced in this study with particle size
focused. Fig. 3 shows the green materials that have been used of 18 ± 4 nm. Saturation magnetization value was 22.1 emu/
by researchers in synthesizing Fe3O4-NPs and will be discussed g with negligible coercivity symbolize the superparamagnetic
in detail. There are a lot of successful studies where Fe3O4-NPs properties of Fe3O4-NPs.
can be synthesized using different biological routes, however
plant extract is the most extensively used in green synthesis 2.3. Leaf
because it can be obtained easily, large-scale production, cost
effective and environmental benign (Iravani, 2011). In addi- Most of the researchers used leaves to study the green synthesis
tion, extracts from plants can act as reducing and stabilizing of Fe3O4-NPs. Rajendran et al. studied on the preparation of
agents in the nanoparticles synthesis which might be due to Fe3O4-NPs by using Sesbania grandiflora leaf extract to be
the presence of phytochemicals. Phytochemicals are com- used as a photocatalyst for chemical oxygen demand (COD)
pounds that are produced by plant itself. Table 1 summaries removal. The preparation steps were very simple where the fer-
different parts of plants in synthesizing Fe3O4-NPs, such as rous chloride (FeCl2) was added to the heated leaf extract and
leaves, fruits, fruit peels, roots, seeds and etc. They contain a stirred. The paste form sample was underwent calcination at
notable amount of phytochemicals such as flavonoid, xantho- 500 °C for 2 h to remove all the impurities and eventually
phylls, carotenoids, anthocyanins and phenolic acids which are
believed to be participated in nanoparticle synthesis.

2.1. Plant

Based on the table, different size, shape and magnetic proper-


ties of Fe3O4-NPs can be synthesized using different types of
plants, and these physical characteristics can work effectively
at different uses. Ngernpimai et al. and Phumying et al. studied
on the Fe3O4-NPs synthesized by using Aloe vera but with dif-
ferent conditions. Fe3O4-NPs prepared by Ngernpimai et al.
were passed through the serial centrifugation steps and the size
of spherical nanoparticles decrease with increasing degree of
centrifugation (Ngernpimai et al., 2012). However, with vari-
ous reaction time and temperature controlled by Phumying
et al., a smaller size and irregular shape of nanoparticles were
produced (Phumying et al., 2013). Fig. 3 Materials of green biosynthesis of Fe3O4-NPs.
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles
Table 1 Fe3O4-NPs synthesized using different part of plants.
Part Name Size range/ Morphology Saturation magnetization (Ms) value References
Average size (emu/g)
Plant Soya bean sprouts 8 nm Spherical 37.1 at 300 K Cai et al. (2010)
44.7 at 1.7 K
Aloe vera 93–227 nm Spherical 74.1–75.9 Ngernpimai et al. (2012)
Aloe vera 6–30 nm Agglomerated irregular 56.3–74.1 at 293 K Phumying et al. (2013)
Marine Sargassum muticum 18 ± 4 nm Cubic 22.1 Mahdavi et al. (2013b)
plant
Kappaphycus alvarezii 14.7 ± 1.8 nm Spherical – Yew et al. (2016)
Padina pavonica 10–19.5 nm Spherical – El-Kassas Hala et al. (2016)
Sargassum acinarium 21.6–27.4 nm Spherical – El-Kassas Hala et al. (2016)
Seed Grape Seed Proanthocyanidin 30 nm Irregular shape 56.6 at 298 K Narayanan et al. (2011)
(GSP)
Syzygium cumini 9–20 nm Agglomerated spherical 13.6 at r.t. Venkateswarlu et al. (2014a)
Leaf Carob 4–8 nm Well monodisperse – Awwad and Salem (2012)
Tridax procumbens – Irregular shape - rough surfaces – Senthil and Ramesh (2012)
Artemisia annua 3–10 nm Spherical 20.7 at 300 K Basavegowda et al. (2014a)
Caricaya Papaya 33 nm (from Agglomerated plate like structure with coarsened grains – Latha and Gowri (2014)
XRD) and capsule like
Perilla frutescens 50 nm Spherical 25.2 at 300 K Basavegowda et al. (2014b)
Euphorbia wallichii 10–15 nm Spherical 23.1 at r.t. Atarod et al. (2015)
Green tea 5.7 ± 4.1 nm Spherical 16.7 at 300 K Xiao et al. (2015a)
Zea mays L. (ear leaves) – Aggregated spherical 1.4 Patra et al. (2017)
Sesbania grandiflora 25–60 nm Agglomerated non-spherical – Rajendran and Sengodan (2017)
Rubus glaucus Benth 40–70 nm Aggregated Spherical – Kumar et al. (2016)
Calliandra haematocephala 85.4–87.9 nm Bead-like spherical – Sirdeshpande et al. (2018)
Lagenaria siceraria 30–100 nm Cubic – Kanagasubbulakshmi and
Kadirvelu (2017)
Fruit peel Plantain peel 30–50 nm Spherical 15.8 at r.t. Venkateswarlu et al. (2013)
Punica Granatum D = 40 nm Rod 22.7 Venkateswarlu et al. (2014b)
L = above
200 nm
Rambutan 100–200 nm Agglomerated, spinel – Yuvakkumar and Hong (2014)
Ananas comosus 10–16 nm Agglomerated spherical 21.7 at r.t. Venkateswarlu and Yoon (2015a)
Citrullus lanatus <17 nm Agglomerated spherical 28.4 at r.t. Venkateswarlu and Yoon (2015b)
Citrus aurantium 17–25 nm Slightly elongated All show no sizable hysteresis at r.t. Bano et al. (2016)
Punica granatum Slightly rod-shaped (40–60) Bano et al. (2016)
Malus domestica Spherical Bano et al. (2016)
Citrus limon Spherical Bano et al. (2016)
Fruit Passiflora tripartita 18.2–24.7 nm Spherical 13.2 Kumar et al. (2014a)
Averrhoa carambola 1.9–3.1 nm Spherical 31.3 at r.t. Ahmed et al. (2015)
Lemon 14–17 nm Spherical 31.4–61.8 at r.t Bahadur et al. (2017)
Couroupita guianensis 17 ± 10 nm Spherical 0.1 at r.t Jha (2017)

2291
(continued on next page)
2292 Y.P. Yew et al.

the nanoparticles were stored. The authors found that by rais-


ing the concentration of leaf extract, the rate of reduction and
the reduction of precursor into nanoparticles increased. The
maximum amount that they used was 20% of leaf extract

Khandanlou et al. (2013)


Niraimathee et al. (2016) and it was the optimum condition which analyzed by UV–vis-

Khataee et al. (2017)


Buazar et al. (2016) ible spectroscopy (UV–vis). Besides, the leaves of Andean
Lunge et al. (2014)

Horst et al. (2017)


Khan et al. (2015)
blackberry (ABL) which is also known as Rubus glaucus Benth,
can also be used to synthesize Fe3O4-NPs. Kumar et al. added
sodium hydroxide (NaOH) solution to adjust the pH of the
Saturation magnetization (Ms) value References

mixed solution of leaf extract and iron precursor solution to


pH 10–11. The mix solution was stirred at around 75–80 °C
until black color solution was observed. The resulting
nanoparticles were centrifuged thrice at 5000 rpm and washed
for several times, then nanoparticles were dried and stored.
The synthesized Fe3O4-NPs were found to be spherical in
shape and aggregating in nature with size range from 40 to
70 nm. This article shows that the polyphenolic compounds
3.2 memu/g at 300 K

present in the leaf extract are most probably responsible to


the formation of clusters and aggregation of the nanoparticles
which act as capping and stabilizing agents. These synthesized
55.4 at r.t.

44.2 at r.t.

Fe3O4-NPs has been used as an efficient photocatalyst for the


6.9 at r.t.
(emu/g)

degradation of methylene blue, congo red and methylene


28.8

orange and play a role as mild antioxidant agent.



2.4. Fruit peel

Fruit peel is the skin of a fruit which protect the flesh of fruit
from the environment and also microbes. Fruit peels can be
used as natural fertilizer because most of them are too thick
which cannot be eaten by human. However, researchers always
make use of natural sources and study about fruit peel in syn-
thesizing nanoparticles. Plenty of favorable studies had been
done by using fruit rind extract to synthesize Fe3O4-NPs. Ven-
Agglomerated rough spherical

kateswarlu et al. researched on the fruit peel pulp extract of


Ananas comosus (pineapple) and Citrullus lanatus (water-
Aggregated spherical

melon) in synthesizing Fe3O4-NPs. Both of the experiment


showed that the synthesized Fe3O4-NPs were spherical in
Cuboid/pyramid

shape with an average size of around 17 nm. Fig. 4 showed


Morphology

the TEM image of Fe3O4-NPs synthesized using watermelon


Spherical

Spherical

peel pulp extract which were agglomerated owing to the pres-


Uneven
Cubic

ence of hydroxyl groups from the extract. The synthesized


Fe3O4-NPs also possessed magnetic properties with saturation
Abbreviations: D, diameter; L, length; r.t., room temperature.

magnetization of 21.7 emu/g and 28.4 emu/g for pineapple and


watermelon respectively. The surface of Fe3O4-NPs were fur-
9.9 ± 2.4 nm
40 ± 2.2 nm
Average size

ther functionalized with ligand in order to utilize the synthe-


Size range/

60–80 nm

10–40 nm
18–35 nm
70–80 nm

sized Fe3O4-NPs in heavy metal removal effectively, which


5–25 nm

were cadmium(II) for pineapple and mercury(II) for water-


melon. By comparing to other reported research, the results
showed that surface modified Fe3O4-NPs were one of the best
result in adsorption capacity. The advantages of employing
Acacia mearnsii (biochar)
Coffee waste hydrochar

ferromagnetic property of Fe3O4-NPs are the simple separa-


tion process from large-volume samples by using an external
magnetic field instead of filtration or centrifugation, thus the
Mimosa pudica

isolation process is rapid and easy. Besides, the nanoparticles


Arabic gum
Table 1 (continued)

Tea residue
Rice straw

also demonstrates easy recyclability without significant loss


of heavy metal removal efficiency.
Potato
Name

2.5. Seed
Stolon
Waste
Root

Gum

In addition, seeds from fruit are kind of fruit waste material


Part

and they can act as green solvent, reducing and capping agent
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2293

Fig. 4 TEM image of (a) DHPCT@Fe3O4 MNPs and (b) the particle size histogram. Reproduced with permission (Venkateswarlu and
Yoon, 2015b).

in synthesizing Fe3O4-NPs. There is a study done by Venkates- gap values showed that the Fe3O4-NPs were grouped as
warlu et al. who utilized Syzygium cumini (S. cumini) seed to semiconductor.
produce Fe3O4-NPs. During the green synthesis process, S.
cumini extract acts as reducer because there is carbohydrates 2.7. Stolon and root
and polyphenols which reduce Fe3+ salt to Fe3O4 by simple
reduction reaction. In this study, XRD was performed to study According to recent study, there are a few novel research
the crystallinity and purity of the Fe3O4-NPs. However, where the part of plants have never been used in Fe3O4-NPs
Raman spectroscopy characterization was also done to con- synthesis, such as stolon (potato), root and gum. Buazar
firm the formation of Fe3O4 without presence of any impurity. et al. studied the mechanism of Fe3O4-NPs formation using
The significant peak at around 670 cm 1 indicated the A1g potato. Potato is a tuberous crop and it is rich in carbohy-
modes of Fe3O4. Besides, peaks that can be found at about drates. Starch-rich potato extract plays an important role as
538 (T2g), 306 (Eg) and 194 cm 1 (T2g) were also the character- capping and reducing agents in the formation of Fe3O4-NPs.
istic bands of Fe3O4. Brunauer, Emmett, Teller (BET) surface The reaction started with addition of NaOH and elicited the
area analysis was studied and the result showed that the sur- oxidation of starch in alkaline solution. These oxidations pro-
face area was 3.517 m2/g. The pore size distribution revealed duced electrons that reduced Fe+ ions to Fe0 nanoparticles.
that majority of the mesoporous had a size of approximately Meanwhile, the starch primary hydroxyl groups were oxidized
2 nm. Hence, this green synthesized mesoporous Fe3O4-NPs to carboxyl group. Moreover, the problem of aggregation of
have potential in various applications, such as biomedical, nanoparticles in water was overcome as Fe3O4-NPs dissolved
catalysis and separation field. in potato extract easily. Thus, enhanced dispersion and steric
protection of mediated Fe3O4-NPs through multifunctional
2.6. Fruit starch-rich potato extract would reduce the particle size (40
± 2 nm). Furthermore, Niraimathee et al. researched on the
Bahadur et al. had done a study on using lemon juice to syn- production of Fe3O4-NPs by using Mimosa pudica root
thesize Fe3O4-NPs. Lemon juice was chosen to act as the extract. The sample was analyzed by UV–vis, where the pres-
source of citric acid for controlling size and surface capping ence of iron oxide was confirmed at the sharp absorbance peak
purpose. The modified co-precipitation technique can be used of 294 nm. The magnetic properties of Fe3O4-NPs were
to produce water dispersible Fe3O4-NPs which is the funda- enhanced by controlling the pH of the solution to pH 9 with
mentals for nanoparticles to be used in biomedical applica- the addition of NaOH. The VSM result showed that the Ms
tions. The size of synthesized Fe3O4-NPs can be controlled value of the synthesized Fe3O4-NPs was found to be 55.40
by tuning the amount of reducing agent, where 11 nm and emu/g, which is considered high compared to other studies.
15 nm of Fe3O4-NPs were fabricated in this study. Based on It was observed that the magnetization decreased from the pla-
XRD results, there were 7 significant peaks can be observed, teau value and got to zero while the magnetic field was
which were located at 2h = 30.07°, 35.51°, 43.33°, 53.44°, removed. Thus, this phenomenon indicated that the Fe3O4-
57.18°, 62.88°, and 74.02°. All these peaks corresponded to NPs possessed superparamagnetic behavior because the
the purity of synthesized Fe3O4-NPs. Besides, optical proper- Fe3O4-NPs correlated with the single-crystal domain, where
ties of Fe3O4-NPs was analyzed using UV–vis spectrometer. only one orientation of the magnetic moment was shown.
The optical energy band gap varied from 2.6 eV (15 nm) to
2.8 eV (11 nm) for direct transition and 1.7 eV (15 nm) to 2.8. Gum
1.82 eV (11 nm) for indirect transition. This indicates that
the energy band gap of Fe3O4-NPs depended on the particle On the other hand, study of using gum Arabic (GA) to pro-
size. Thus, the direct and indirect energy band gap values of duce Fe3O4-NPs is also another successful research. Horst
Fe3O4-NPs were in the range of 1.7–2.8 eV, and these band et al. studied about the possible mechanism of the Fe3O4-
2294 Y.P. Yew et al.

NPs formation. There are 2 types of interactions expected to agent and the source of coating agent), except for fructose.
happen between the polysaccharides from the gum and iron Fructose did not show any characteristics of Fe3O4 because
oxide nucleus, which are electrostatic and/or hydrophobic fructose is a non-reducing monosaccharide. Based on TEM
interactions. Another feasibility is the formation of complex, results, the particle size of synthesized Fe3O4-NPs varied from
due to the bridging from biopolymer to the Fe3O4 nucleus. 3.8 to 13.1 nm and the morphology of the nanoparticles were a
In the initial stage of synthesis process, the media is acidic mixture of slightly agglomerated spherical-like, rod-like, and
owing to the iron salt precursor in contact with polymeric moi- dendritic nanostructure. The magnetization measurements
eties. NH4OH is then added to increase the pH and the first were also carried out to study the magnetic properties of syn-
Fe3O4 nucleus is produced. In such conditions, Fe3O4 and thesized Fe3O4-NPs. Fe3O4-NPs that prepared with galactose,
GA show opposite surface charge. Besides, it is high possible mannose and maltose possess superparamagnetic characteris-
that electrostatic interaction occur where FTIR data confirmed tics. However, saturation magnetization value of Fe3O4-NPs
the interaction between carboxylic groups of GA and hydroxyl synthesized with maltose (Ms  40 emu/g) was lower than
groups of Fe3O4 was via hydrogen bonding. As both polymer galactose and mannose (Ms  60 emu/g). The Fe3O4-NPs pre-
and iron oxide have negative charge at the higher pH of Fe3O4, pared by lactose along with its low crystallinity resulted in
steric interactions might occur too which responsible for the weaker magnetization (Ms = 20 emu/g), while for Fe3O4-NPs
GA binding. Steric interactions is crucial to illustrate the stabi- prepared by fructose, the magnetization was very weak even
lization mechanism of Fe3O4-NPs by the polymeric moieties. at high fields. This is explained by the crystallinity of samples
The hydrophilic nature of GA-Fe3O4 can be explained by which analyzed using XRD. The magnetization depends on the
thinking that the GA chains bind to the Fe3O4 in the way that crystallinity of nanoparticles where the higher the crystallinity
charged (mostly negative) functional groups remained surface of Fe3O4-NPs, the higher the magnetization. However, these
exposed, showing electrostatic repulsion between nanoparti- magnetization is smaller than that of bulk particles (92
cles. This situation may take place if not all the functional emu/g). This is due to the presence of surface spin disorder
groups of the biopolymer are interacting with the Fe3O4 sur- and spin canting effects, which happen when the surface to vol-
face groups. ume ratio of particle increases while the particle size decreases.
As a result, the saccharides coated Fe3O4-NPs have a potential
2.9. Plant waste in biomedical applications such as magnetic resonance
imaging.
Furthermore, the applications of Fe3O4-NPs might depend on
the substrate used as well. Rice straw, fruit peels and coffee 3.2. Polysaccharides
waste hydrochar are natural waste which we might think they
are worthless. But, the Fe3O4-NPs synthesized by waste can be On the other hand, Chang et al. did a study on the synthesis of
useful too. Based on studies, Fe3O4-NPs prepared by tea resi- superparamagnetic Fe3O4-NPs using polysaccharides, includ-
due, coffee waste hydrochar and corn Zea mays can be used in ing soluble starch, carboxymethyl cellulose sodium (CMC)
arsenic removal (Lunge et al., 2014), Acid Red 17 (azo dye) and agar. These three polysaccharides acted as stabilizer dur-
removal (Khataee et al., 2017) and drug delivery applications ing the synthesis procedure to enhance the stability, biocom-
(Patra et al., 2017) respectively. All these studies show that patibility and biodegradability. TEM images showed that the
Fe3O4-NPs capped with green substrate have a promising approximately 10 nm spherical Fe3O4-NPs were capped by
potential in various kind of applications in the future. polysaccharide. Polysaccharides could form hybrids with metal
ions owing to their high number of coordinating functional
3. Other green materials groups (hydroxyl and glucoside groups). Hence, most of the
iron ions were associated closely with polysaccharides mole-
Green biosynthesis is not restricted to plant synthesizing cules, where nucleation and initial crystal growth of Fe3O4-
nanoparticles only, yet utilization of other green materials such NPs might then occur preferentially on polysaccharides.
as natural polymer, amino acid, vitamin, enzyme and fungi, Besides, polysaccharides present dynamic supramolecular
assist in Fe3O4-NPs synthesis too. Table 2 shows some of the associations facilitated by inter- and intra-molecular hydrogen
green substrates in synthesizing various size and shape of bonding, which play a role as template for the nanoparticles
Fe3O4-NPs. growth. The size of Fe3O4-NPs synthesized by soluble starch
is less than 10 nm, whilst the other two showed a larger size.
3.1. Glucose This phenomenon might be related to the structure of polysac-
charides; in aqueous state, soluble starch is mainly composed
of branched amylopectin, whereas CMC and agar contain
As predicted, shape, particle size and magnetic properties of more linear-polysaccharide structure. So, soluble starch has
synthesized Fe3O4-NPs are different based on different kinds more interactions with iron ions than CMC and agar, which
of green materials used, just as Table 1. This might due to caused more restriction on the growth of Fe3O4-NPs. It is
the distinct condition used during the synthesis procedure known that the magnetization of Fe3O4 is very sensitive to
and it depends on the properties of green substrates possessed the microstructure. Fe3O4 particles are called single-domain
as well. According to Table 2, glucose is the most popular to be particles when the Fe3O4 particles are smaller than the critical
used in synthesizing Fe3O4-NPs. Demir et al. researched the size. Thus, as the particle size decreases below the critical
effect of 5 different types of saccharides on the characteristics single-domain size, the particles exhibit superparamagnetic
of synthesized Fe3O4-NPs, including mannose, maltose, lac- attributes. Nevertheless, when the magnetizations of particles
tose, galactose and fructose. All the saccharides played a role are random (without any definite direction), each of the parti-
as bifunctional agents (both as the precursor of the reducing
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles
Table 2 Fe3O4-NPs synthesized by other green substrates.
Green substrate Name Size range/ Morphology Saturation magnetization (Ms) References
average size value (emu/g)
Glucose a-D-glucose 12.5 nm Roughly spherical shape 71.3 at 5 K Lu et al. (2010)
60.5 at 300 K
Maltose 12.1 ± 2.1 nm Spherical 43.1 at r.t. Demir et al. (2013)
Sucrose 4–16 nm Spherical 14.8–29.6 at 7 KOe Sun et al. (2009)
a-D-maltose 9.7 ± 1.0 nm Spherical-like, rod-like, and dendritic nanostructure with some 37.4 Demir et al. (2014)
a-D-mannose 13.1 ± 0.3 nm extent of agglomeration 59.1
a-D-galactose 12.4 ± 0.3 nm 58.1
a-D-lactose 3.8 ± 0.21 nm 22.0
D-glucose Twig = 10–20 Coral like – Qin et al. (2011)
nm
L = 10–100 nm
Vitamin Nicotinic acid (N. acid) 0 g N.acid Nanorod 0 g = 55.0 Attallah et al. (2016)
L = 270 nm, 1 g = 30.0
D = 20 nm 2.5 g = 4.0
1g N.acid
L = 300 nm,
D = 30 nm
2.5 g N.acid
L = 350 nm,
D = 40 nm
Ascorbic acid 15 ± 4 nm Irregular – Nene et al. (2016)
Enzyme Urease 19 ± 5 nm 60 °C = nanosphere 52.6 Shi et al. (2014)
Thickness 40 °C (2h) = nanosheets 27.6
< several nm 40 °C (1h), 60 °C (1h) = nanorods 15.8
L > 100 nm
Section = 10 ±
4 nm
Fungi Yeast 16 nm Wormhole-like 22.1 at r.t. Zhou et al. (2009)
Natural Sodium alginate 27.2 nm Uniform and spherical 62.1 Gao et al. (2008)
polymer
Chitosan 22.0 ± 7.8 nm Nearly Spherical 65 at r.t. Shrifian-Esfahni et al. (2015)
Agar 20–30 nm Non spherical 18.7–25.3 at r.t. Hsieh et al. (2010)
aggregated
Polysaccharides Starch Less than 10 nm Spherical 36.2 at 300 K Chang et al. (2011)
Carboxymethyl More than 10 nm Spherical 35.8 at 300 K Chang et al. (2011)
cellulose sodium
Agar More than 10 nm Spherical 20.4 at 300 K Chang et al. (2011)
Pectin 5–18 nm Cubic 53–54 at 300 K Namanga et al. (2013)
Amino acid Arginine Fe/Ar (1:1) = Spherical 51.7 at 300 K Wang et al. (2009)
18–26 nm 39.9 at 300 K
Fe/Ar (1:2) = 9–

2295
15 nm
(continued on next page)
2296 Y.P. Yew et al.

cles suppresses the exchange interaction between the particles.


This lack of hysteresis is very crucial for recognition of a sam-

Hadian-Dehkordi and Hosseini-


ple with superparamagnetic properties. VSM results showed
that all the Fe3O4-NPs prepared by soluble starch, CMC and

Grumezescu et al. (2013)


Shirkhodaie et al. (2016)
Theerdhala et al. (2010) agar exhibited good saturation magnetization, even though

Kalantari et al. (2014)


Belachew et al. (2016)
Belachew et al. (2017)
Fe3O4-NPs synthesized by CMC and agar had slightly larger
hysteresis loop and coercivity.
Park et al. (2009)
Park et al. (2009)

Monfared (2016)
Cao et al. (2014)
Lai et al. (2010)

3.3. Clay
References

Clay is a natural soil or rock material which commonly used in


making pottery and some constructions products, such as
bricks and tiles. However, clay can be used as the supporting
Saturation magnetization (Ms)

materials for nanoparticles synthesis. Kalantari et al. synthe-


sized Fe3O4-NPs by using montmorillonite (MMT) as a solid
support. The shape and size of Fe3O4-NPs can be controlled
by altering the amount of NaOH (1.50–12.50 mL) as reducing
agent in the medium. Fe3O4-NPs were prepared through
12.1–32.4 at r.t.
value (emu/g)

66.7 at 300 K

coprecipitation by addition of base to Fe2+ and Fe3+ salts


 60 at r.t.
49.9 at r.t.

14.7 at r.t.

solution. The chemical reaction equation can be written as


65 at r.t.
85 at r.t.
35–50

Fe2+/Fe3+/MMT + 8OH ? Fe3O4/MMT + 4H2O. The


presence of Fe3O4 can be confirmed by using XRD analysis.

However, XRD analysis also showed that the basal spacing


expanded from 1.47 to 2.85 nm by increasing the weight per-
cent (wt%) of Fe3O4-NPs content in MMT matrix from 1.0
to 12.0 wt%. The 2h° of XRD patterns shifted from 8.75° to
7.46°, where the basal spacing of pristine MMT was 1.24 nm
at 2h°, 8.83°. This indicated that the iron ions might penetrate
into the interlayer space of MMT via ion-exchange and then
Mixture of nanorods and rounded particles

were reduced to Fe3O4-NPs by addition of NaOH. Hence,


the interlayer space might act as microreactor and size con-
troller. Besides, the intensities of XRD peak (2h° = 8.83° to
7.46°) decreased gradually, and the highly ordered parallel
lamellar structure of MMT was disrupted by the formation
of Fe3O4-NPs. Based on TEM results, the Fe3O4-NPs were
in the interlayer space or on the surface of MMT. The size
of Fe3O4-NPs decreased as the amount of NaOH increased.
The charged Fe3O4-NPs are bounded to the surface of MMT
via electrostatic force owing to the high density of ion-
Morphology

exchange sites on MMT. The Fe3O4-NPs aggregate in the


Spherical
Spherical
Spherical
Spherical
Spherical
Spherical

Spherical

Spherical
Irregular

Less than 100 nm Irregular

MMT is in direct correlation with the smaller primary


nanoparticles dimension. Moreover, the magnetic properties
Abbreviations: L, length; D, diameter; r.t., room temperature.

of Fe3O4/MMT NCs increased from 12.1 to 32.4 emu/g as


the Fe3O4 content increased, which indicated more Fe3O4 were
19.5 ± 4.2 nm
4.6 ± 2.6 nm
5.9 ± 1.6 nm

caged in the MMT layers. Besides, LAPONITE is a kind of


average size
Size range/

38 ± 5 nm

nanoclay smectite which has layered structure similar to natu-


2–15 nm

8–13 nm
4.0 nm
5.5 nm

ral hectorite. Ding et al. had synthesized LAPONITEÒFe3O4-


13 nm

10 nm

NPs (LAP-Fe3O4-NPs) via co-precipitation method for in vivo


magnetic resonance imaging of tumors. LAP-Fe3O4-NPs pos-
sessed good colloidal stability, which is about 2-fold increase
L-(+)-Tartaric acid

of T2 relaxivity than naked Fe3O4-NPs. The XRD patterns


Montmorillonite
L-glutamic acid

of LAP, Fe3O4-NPs and LAP-Fe3O4-NPs were shown in


L-methionine

Perlite (soil)

Fig. 5a, where it can be noticed that the intensity of peak


Usnic acid
L-arginine
L-arginine

L-cysteine

decreased as a result of Fe3O4-NPs loading. Fig. 5b is the


L-serine
L-lysine
Table 2 (continued)
Green substrate Name

FTIR spectra of LAP, Fe3O4-NPs and LAP-Fe3O4-NPs. The


peaks at 586–598 cm 1 of Fe3O4-NPs and LAP-Fe3O4-NPs
corresponded to the Fe-O vibration of magnetic core, and
Organic acid

the Si-O stretching vibration band of LAP in LAP and LAP-


Fe3O4-NPs were located at 1016–1019 cm 1. Both of the
Others

results proved that the synthesized nanomaterials are compos-


Clay

ite of LAP and Fe3O4-NPs.


Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2297

Fig. 5 XRD pattern (a) and FTIR spectroscopy (b) of LAP, Fe3O4, and LAP-Fe3O4-NPs. Reproduced with permission (Ding et al.,
2016).

It is well known that silver nanoparticles have excellent


antibacterial properties. However, based on several studies,
Fe3O4-NPs can act as an antibacterial agent too. Patra et al.
studied on the green synthesized Fe3O4-NPs by using corn
ear leaves with the application of antibacterial (Patra et al.,
2017). Antibacterial activity was determined by standard disc
diffusion method using five different foodborne bacteria,
which were Bacillus cereus ATCC 13061, Escherichia coli
ATCC 43890, Listeria monocytogenes ATCC 19115, Staphylo-
coccus aureus ATCC 49444, and Salmonella typhimurium
ATCC 43174. The results revealed that Fe3O4-NPs (25 mg)
and standard antibiotics (kanamycin and rifampicin at concen-
tration 5 mg/disc) did not exhibit any antibacterial activity if
tested separately with five foodborne pathogenic bacteria.
Antibacterial activity can be observed when Fe3O4-NPs mixed
with standard antibiotics kanamycin where all the foodborne
pathogenic bacteria showed inhibition zones in the range of
9.87 and 18.86 nm in diameter. Besides, when Fe3O4-NPs com-
bined with rifampicin, the antibacterial activity can only be
observed against Staphylococcus aureus ATCC 49444, with
inhibition zone of 20.90 mm. This suggests that the Fe3O4-
NPs combined with conventional antibiotics can exert syner-
Fig. 6 Examples of biomedical applications of Fe3O4-NPs.
gistic effect, which reduce the doses of antibiotics and thus
decrease the phenomenon of resistant bacterial and mam-
4. Biomedical applications of Fe3O4-NPs malian cell toxicity.
Another potential biomedical application of Fe3O4-NPs is
hyperthermia treatment. Horst et al. had characterized the
Green synthesized Fe3O4-NPs are capable to be used in vari-
Gum Arabic synthesized superparamagnetic Fe3O4-NPs with
ous applications as reported in literature. However, biomedical
magnetocalorimetric assays to study the potential of their for-
applications of Fe3O4-NPs are more concerned by experts
mulations for magnetic hyperthermia therapy under radiofre-
nowadays as human health is threaten due to several issues,
quency fields. Magnetocalorimetric assays were carried out in
such as pollutions, processed food industry, weather change
a wide range of frequency and amplitude. Specific absorption
and unbalance lifestyle. These issues may cause plenty of seri-
rate (SAR) was 218 W/g Fe, which was identified at field fre-
ous diseases, and one of the most popular diseases is cancer.
quency of 260 kHz and the amplitude of 52 kA/m. This results
Fig. 6 summarize the potential biomedical applications of
revealed the feasibility of the Fe3O4-NPs to be applied in
Fe3O4-NPs, including antibacterial, tissue engineering and
tumor ablation treatments. By using the linear response theory
hyperthermia. They also play important roles as magnetic res-
and restricting field parameters to the accepted biomedical
onance imaging (MRI) contrast agent (Li et al., 2014a; Sun
window, it is found that the estimated maximum useful value
et al., 2016), photothermal therapy of tumors (Li et al.,
is 74 w/h Fe at amplitude of 12 kA/m and field frequency of
2015), magnetofection agent and can be used for magnetic
417 kHz. On top of that, the fascinating physicochemical prop-
bioseparation and DNA molecule detection.
erties of these Fe3O4-NPs such as small size, polydispersity and
2298 Y.P. Yew et al.

stability in aqueous colloid suspensions transformed the cells, and the level of gene expression was highly dependent
Fe3O4-NPs to an efficient device for hyperthermia treatment on the dendrimer generation, plasmid DNA concentration
(Horst et al., 2017). and the amine to phosphate group ratio. The best system
Tissue engineering is also one of the important applications was found out to be the dendriplex-coated Fe3O4-NPs formed
in biomedical field in repairing, replacing or regenerating parts by generation 6 dendrimers at an amine to phosphate group
of or whole tissues. According to the research done by Gil ratio of 10. The analyzed results revealed that the nanohybrids
et al. (2015), they had constructed cell sheets using Fe3O4- possess the potential as an effective gene delivery nanomateri-
NPs with the presence of magnetic force. It is observed that als (Xiao et al., 2015b).
the magnetically labelled cells moved towards the magnet Moreover, Fe3O4-NPs can be employed in deoxyribonu-
and gathered on the bottom of the nonadherent plate in situ, cleic acid (DNA) molecule detection application. Sun et al.
which then constructing a sheet-like structure, in the presence fabricated a chemiluminescence (CL) biosensor for ultrasensi-
of external magnetic field without using artificial polymer scaf- tive determination of DNA (Sun et al., 2017). Core-shell Fe3-
folds. It is reported that the cell sheet constructs were not O4@SiO2 – graphene oxide (Fe3O4@SiO2@GO) polymers
adhered to the culture plate, which can be easily removed from were prepared in this study. The principle of this CL biosensor
the surface of culture plate without utilizing any detachment was the adsorption recognition function between Fe3O4@-
procedure. Besides, nanospheres showed better internalization SiO2@GO polymers and DNA. The results of the adsorption
efficiency, and the labelled cells exhibit strong transportation capacity of Fe3O4@SiO2@GO achieved the maximum value
reaction with external magnetic fields, compared to nanorods. of 3.24  10 9 mol/g. The binding process of the polymers
The results of this research confirm the evolution of magnetic- and DNA comply with the Langmuir isotherm equation and
responsive nano-biomaterials which applicable in tissue engi- pseudo second order sorption kinetics. The selectivity and sen-
neering or cellular therapies. sitivity of DNA detection was notably enhanced by applying
Fe3O4-NPs are well known with its superparamagnetic the CL technique, where the reactions of complementary base
properties where they are suitable for magnetic bioseparation, pair between Fe3O4@SiO2@GO-DNA and complementary
especially in cell separation. Based on the research reported by nucleotide chains were studied. Based on the promising results
Lu et al. (2014), they had synthesized polyethylenimine (PEI)- obtained, the Fe3O4@SiO2@GO-DNA-CL biosensor is appli-
coated Fe3O4-NPs for the separation and enrichment of lung cable in diagnosing human genetic diseases and provide advis-
cancer cell from sputum samples, and then cytopathology able treatment.
analysis was performed. Exfoliative cytopathology analysis Lastly, the potential biomedical application of Fe3O4-NPs
gave a result which the percentage of positive cells increased via targeted drug delivery system will be discussed. There are
from 6.3% in untreated sputum samples to 38.5% in sputum a few types of drugs can be integrated with Fe3O4-NPs, such
samples treated with the PEI-coated Fe3O4-NPs. This outcome as anticancer and anti-inflammatory. One of the studies
presents the promising application of PEI-coated Fe3O4-NPs showed that indomethacin (a poorly water-soluble non-
in enrichment of lung cancer cells from sputum for cytopathol- steroidal anti-inflammatory drug) conjugated with Fe3O4-
ogy analysis. NPs incorporated into electrospun nanofiber composites of
Furthermore, Fe3O4-NPs can be utilized as magnetofection two cellulose derivatives. The results showed that the compos-
agent. In this case, magnetofection is developed where the ite nanofiber exhibit superparamagnetism at room tempera-
nucleic acid drugs combine with superparamagnetic iron oxide ture, and the presence of Fe3O4-NPs in the nanofiber did not
nanoparticles to form magnetoplexes. The production of mag- affect the drug release process, which was found to be mainly
netoplexes can be quickly accumulated on the targeted sites controlled by the polymeric carrier matrix properties (Wang
with the aid of additional magnetic field and as a conse- et al., 2012). Therefore, the magnetic drug loaded nanofibers
quences, the transfection efficiency can be enhanced. Liu have a potential in medicine applications, particularly targeted
et al. reported their research where DMSPION-G6/DNA/ drug delivery in digestive system.
PEI ternary magnetoplexes was prepared for in vitro gene
delivery (Liu et al., 2011). The results showed that the 5. Utility of Fe3O4-based nanoparticles as drug delivery vehicles
DMSPION-G6/DNA/PEI ternary magnetoplexes exhibited
enhanced transfection efficiencies in three cell lines, including Lately, human health is threatened with miscellaneous diseases
COS-7, 293 T and HeLa cells. By utilizing DMSPION-G6/ and new drugs have to be invented to solve this crucial issue.
DNA/PEI ternary magnetoplexes, the incubation time needed Hence, Fe3O4 is gaining popularity in drug delivery system
was shorten and DNA dose required decreased when magnetic due to the excellent magnetic properties possessed which is also
field was employed. This revealed that high-level transgene known as superparamagnetism. Besides, in order to utilize the
performance was accomplished, which time and dose issues Fe3O4-NPs as drug delivery vehicle, they have to exhibit a few
were resolved when magnetofection was used. Another result significant properties which cannot be neglected and will be
from Prussian blue staining analysis showed that addition of discussed in this section.
magnetic field could accumulate the magnetoplexes rapidly
to the surface of target cells and then improved the magneto- 5.1. Criteria of Fe3O4-based nanoparticles to be used in drug
plexes uptake by the cells. Xiao et al. reported that Fe3O4- delivery
NPs can be functionalized with plasmid DNA to develop
nanohybrid systems for nucleic acid therapy. Nanohybrids
were produced by merging the dendrimers complexes, plasmid
5.1.1. Superparamagnetic
DNA (dendriplexes) and poly(styrene) sulfonate-coated
Fe3O4-NPs via electrostatic interactions. The outcome of the The basic requirements for Fe3O4-based nanoparticles to be
study showed that the nanohybrids can transfect NIH 3T3 used in drug delivery application are presented in Fig. 7,
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2299

most of the nanoparticles synthesized are spherical in shape


for the in vitro study in treating various kinds of cancerous
cells. All the studies show significant cytotoxicity and effective-
ness in inhibiting cancer cells growth, which reveal the promis-
ing potential of nanoparticles to be used in drug delivery
application.

5.1.3. Size of nanoparticles


The dimensions of nanomaterial act an important role in
Fig. 7 Criteria of nanocarrier in drug delivery application. determination of total cell uptake in drug delivery system.
Hence, optimum size of nanodrug carrier has to be figured
out in order to maximize the cells uptake rate and intracellular
comprising magnetic properties, shape, size and surface char- concentration in mammalian cells. In general, a 50 nm diame-
acteristics, which are the important matters in this section. ter of nanomaterial is expected to be the optimal one, but not
First, Fe3O4-based nanoparticles are well known as magnetic towards all kinds of cells. A larger nanoparticle, such as larger
nanoparticles and they can be guided easily to a specific site than 50 nm, could bind with high affinity to a huge number of
by the aid of magnetic field that improve its local concentra- receptors and might limit the binding of additional nanoparti-
tion. This is one of the advantages which can enhance the drug cles. In contrast, a 40–50 nm nanoparticle is able to assemble
delivery efficacy as well as reduce the side effects of chemother- and bind sufficient receptors to produce membrane wrapping
apeutic drugs. In addition, superparamagnetic behavior is a favourably. However, the effectiveness of size of nanomaterial
must for magnetic nanoparticles to exert its maximum effect in cell uptake depends on the types of cell as well, because each
in the application of drug delivery. Actually, superparamag- cell type owns a distinct phenotype. On the other hand, the size
netic nanoparticles possess zero net magnetic moment without of nanoparticles dictates the half-life in the blood circulation.
the presence of external magnetic field. This can be a great ben- For example, a particle that has a size larger than 200 nm will
efit in tumor targeting as the propensity of self-aggregation is concentrate in spleen or it will be taken up by phagocytic cells
minimized outside the targeted site (Chertok et al., 2008). of the body. While a particle with a size smaller than 10 nm, it
Thus, the anticancer drug can be delivered perfectly to the will be removed by renal clearance. In literature, particles lie in
desired region without damaging healthy tissues. the size range of 10–100 nm are believed to be the optimum.
They have a longer circulation times in the body as they can
5.1.2. Shape of nanoparticles evade the reticuloendothelial system in the body as well as pen-
Furthermore, shape of Fe3O4-based nanoparticles is one of the etrate through very small capillaries. For the superparamag-
important features that has to be taken into consideration in netic nanoparticles, small size nanoparticles are capable to
drug delivery. The issue that researchers concern the most is improve permeability and retention effect, which can lead to
to prolong the nanoparticles in targeted site as well as perform maximal accumulation of nanoparticles at the targeted site.
great cellular cytotoxicity. Besides, the blood circulation time, However, superparamagnetic nanoparticles with a size smaller
cellular uptake and biodistribution may change based on the than 2 nm are not advisable for medical use. The reason is that
shape of nanoparticles. Plenty of studies had been done to the nanoparticles in this range of size have the potential to dif-
research on the nanoparticles shape particularly for anticancer fuse through cell membrane readily and thus causes intracellu-
drug delivery. Filomicelles is the one which found to own lar organelles damage. This situation not merely can exhibit
higher anticancer drug encapsulation capacity and apoptotic toxic effects, but endanger human life. Different shapes and
efficiency in comparison with spherical micelles. One of the sizes of nanoparticles have to be avoided because as they move
studies tested an in vivo antitumor activity of various shapes through narrower capillaries, agglomeration might occur
of micelle, and the results showed that filamentous micelles which may lead to clogging in blood system. Hence, the size
exhibit the highest DOX loading capacity and encapsulation of nanoparticles has to be controlled during the preparation
efficiency. Besides, the filamentous DOX-loaded micelles stage so that they can be utilized effectively as a drug carrier.
reveal the highest safety to human body and the greatest ther-
apeutic effects to artificial solid tumors (Chen et al., 2012). 5.1.4. Surface modification and stability
There are also many other shapes of nanoparticles that had Besides, surface properties is also one of the important factors
been synthesized in drug delivery study, such as rod shape, which can influence the performance of nanoparticles in drug
worm shape and bead shape. In literature, non-spherical and delivery. Generally, nanoparticles used in delivery in vivo
rod-shaped nanoparticles possess a longer blood circulation ought to possess good antifouling property which can defy
time compared with the spherical nanoparticles. This might nonspecific adsorption of protein or other biological macro-
due to the rod-shaped particles cause a lower phagocytic activ- molecules. This can thus prolong the blood circulation time
ity of macrophages than spherical ones (Wahajuddin, 2012). by undergoing adequate surface functionalization of nanopar-
However, spherical nanoparticles exhibit significant advan- ticles (Ma et al., 2017). In fact, most of the synthesized Fe3O4-
tages than rods, according to the research with sub-100 nm NPs undergo surface modification before loading with any
nanoparticles. The spherical nanoparticles can provide an even drug because this is essential for them to play the role as a drug
surface coating and conjugation of ligands in surface modifica- carrier. There are many materials can be used to modify the
tion, which mean more drug can be loaded on the surface of surface of nanoparticles by coating, particularly polymer. This
nanoparticles for better drug release at the targeted site and coating process can increase the colloidal stability of nanopar-
hence show a greater cellular toxicity. Based on literature, ticles and improve the dispersity. In comparison to the
2300 Y.P. Yew et al.

uncoated Fe3O4-NPs, bare Fe3O4-NPs can be oxidized easily not perform well. On the other hand, highly stable conjugation
under ambient conditions (Ali et al., 2016). Besides, bare between drug molecules and surface of Fe3O4-NPs could elicit
Fe3O4-NPs tend to agglomerate owing to their high specific failure of drug release at the targeted site. Hence, researches
surface area versus respective volume and strong inherent mag- should be done to overcome these kinds of problems in order
netic dipole interactions, eliciting rapid total clearance by the to eradicate the tumor effectively. Furthermore, Fe3O4-NPs
reticuloendothelial system (RES) (Hu et al., 2018). Moreover, should be designed in a way that not only release the
surface-engineered nanoparticles can provide a surface for chemotherapeutic drug in eliminating cancer cells, but should
linkage between drug molecules and targeting ligands. Blood also study the non-toxicity, biodegradability and sterility as
circulation time can also be increased by preventing the clear- they will be used in drug delivery system.
ance through RES and thus makes the nanoparticles biocom-
patible which exhibit lower toxicity towards human body. In 5.2. Targeted anticancer drug delivery
general, the stability of Fe3O4-NPs in the biomedical applica-
tion can be improved by undergoing surface coating. However, A targeted drug delivery system is illustrated in Fig. 8. The
most of the coating approaches showed some drawback drug loaded Fe3O4-NPs is consumed by human through par-
because they are complicated, time consuming and some even enteral drug administration. It is shown that the drug loaded
require high energy (high pressure and high temperature) (Li Fe3O4-NPs are injected into the blood capillary and located
et al., 2017). Hence, proper manner of coating need to be done at the targeted site (cancer cells/tumor) by the aid of external
to maintain the desirable properties of Fe3O4-NPs in drug magnetic field. This can help to accumulate the drug and
delivery application. Fe3O4-NPs with a positive surface charge release the drug at the desired site, and thus increase the effi-
possess a better properties as compared to neutral and negative cacy in treating cancer cells without harming neighbour
charge. It is reported that the cell membrane owns a slight neg- healthy cells.
ative charge and cell uptake is driven by electrostatic attrac- Table 3 shows the examples of Fe3O4-based nanoparticles
tions. Hence, positively charged nanoparticles are better as an anticancer drug vehicle in treating different kinds of can-
because they can be taken up at a faster rate. However, as cer cell line using various anticancer drugs in these recent
aforementioned, the intake of nanoparticles depends on cell years. Every study modified the surface of Fe3O4-NPs with dis-
type. tinct materials, such as chitosan, polymer and silica. All the
results show the potential and promising application of
5.1.5. Drug loading and release Fe3O4-NPs in anticancer drug delivery system, which cancer-
Drug loading should be done in the way that the functionality ous cells were eradicated after treating with drug loaded
of drug is not affected. In the meantime, drug loaded nanopar- Fe3O4-NPs.
ticles should release the drug at the targeted site at an appro- Besides in vitro, in vivo drug delivery study is also crucial
priate rate without harming the healthy cells. There are few because it is essential to understand how the nanocarriers func-
ways to load the drug on nanoparticles, such as conjugating tion in the body to eradicate cancer cells. There are plenty of
the drug molecules on the surface of nanoparticles or encapsu- researches have been done which adopt mouse as subcuta-
lating the drug molecules together with the coating material. neous tumor model. Lu et al. had prepared a pH-sensitive dual
For conjugation of drug on the surface of nanoparticles, the targeting magnetic nanocarrier for chemo-phototherapy in
linking process can be divided into two groups, which are con- cancer treatment (Lu et al., 2018). They synthesized magnetic
jugation via cleavable covalent linkage and via physical inter- graphene oxide (MGO) by depositing Fe3O4-NPs on graphene
actions. Covalent linkage incorporate the combination of drug oxide (GO) via chemical co-precipitation method. MGO was
molecule with functional groups present on the surface of then modified with polyethylene glycol (PEG) and cetuximab
nanoparticles, which have been coated with polymer. Linker (CET) to acquire MGO-PEG-CET. An anticancer drug dox-
can also be used to attach the drug molecule to the nanoparti- orubicin (DOX) was then loaded to MGO-PEG-CET to
cles. This method can enhance drug loading capacity and also
preserve the functionality of the drug, and thus efficacy. Phys-
ical interactions such as electrostatic interactions, hydrophobic
and hydrophilic interactions can lead to conjugation of drug
molecules on the surface of nanoparticles. This phenomenon
happens when there is different of charges. For example, the
Fe3O4-NPs coated with cationic polymer can interact electro-
statically with negatively charged DNA. Besides, lipophilic
drugs can link with the Fe3O4-NPs readily if the Fe3O4-NPs
coated with hydrophobic polymers, and this can enhance the
drug release as the coating degrades. As a drug delivery sys-
tem, Fe3O4-NPs should be able to release their drug payload
at optimal condition. However, there are a few drawbacks
on the drug release which cannot be neglected. Most of the
drug payload released rapidly upon injection into the in vivo
situation because the drug is loaded on the surface of Fe3O4-
NPs, such as burst effect. As a result, low entrapment effi-
ciency causes only small amount of drug reached the targeted Fig. 8 Targeted drug delivery system using drug loaded Fe3O4-
site and the effectiveness of drug in killing cancer cells could NPs.
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles
Table 3 Examples of anticancer drug loaded Fe3O4-NPs used in drug delivery.
Ref. Anticancer drug Surface modification Drug carrier (shape and size) Type of Cell Line Results
Cancer
Malekzadeh Quercetin 1. Poly citric acid (PCA) Regular spherical shape in the Cervical HeLa Significant cytotoxicity was clearly showed in both HeLa and MDA-
et al. (2017) 2. Poly(ethylene glycol) (PEG) range of 10–15 nm Breast MDA- MB-231 cells for quercetin loaded nanocarrier, yet nanocarrier did
3. Folic acid MB-231 not show any cytotoxicity against cancerous cell lines
Barahuie Phytic acid 1. Chitosan Roughly spherical shape with Colon HT-29 Have good anticancer potential against colon cancer, do not show
et al. (2017) mean size of 8 ± 3 nm any cytotoxicity to normal fibroblast cells
Venugopal Doxorubicin 1. Gold coated Irregular shape with size varied Brain Rat C6 Doxorubicin loaded NPs killed tumor cells, and the efficacy increase
et al. (2016) 2. Gellan gum between 75 and 150 nm glioma with the presence of magnetic fields
Taghavi et al. Deferasirox 1. (3-aminopropyl) Uniform spherical shape which Breast MCF-7 The nanocarrier showed excellent cytotoxicity against human
(2016) trimethoxysilane (APTMS) had a size around 44 nm Cervical HeLa leukemia cell line compared to other cell line. Drug loaded NPs
Colon HT-29 showed higher apoptosis-inducing effect in cancer cell lines than free
Leukemia K-562 drugs in vitro
Nerve Neuro-2a

Shahabadi Cytarabine 1. Tetraethoxysilane (TEOS) Almost spherical with uniform Leukemia HL-60 Drug loaded NPs had better anticancer effect where the study showed
et al. (2016) average particle size of 23 nm. KG-1 double antiproliferative effects on cancerous cell lines compared to
Lymph Raji free drug
Pham et al. Curcumin 1. cetyl trimethylammonium Spherical in shape with average Lung A549 Increase concentration of drug loaded NPs increased the percentage
(2016) bromide (CTAB) size of 13–17 nm of inhibition. Free curcumin inhibited cancer cells better than drug
2. Chitosan loaded NPs due to the slow release rate of curcumin from NPs
Rehana et al. Paclitaxel 1. L-arginine Spherical in shape with particle Lung A549 L-arginine coated NPs showed enhanced cytotoxicity effect against
(2015) size of 26 nm cancer cells and lead to apoptosis, compared to other coated NPs.
IC50 value of drug loaded L-arginine coated NPs was lower than free
drug which showed the effectiveness in inhibiting cancer cells growth
Ghosh et al. Diosgenin 1. Citric acid Monodispersed between 19 and Breast MCF-7 Diosgenin loaded NPs exhibited better antiproliferative activity
(2015) 21 nm (51.08 ± 0.37%) against MCF7 compared to free diosgenin (33.31
± 0.37%). The drug loaded NPs possessed good migrating inhibiting
and apoptosis inducing properties against breast cancer
Kumar et al. Quercetin 1. Dextran Monodispersed prism like shape Breast MCF-7 Quercetin loaded NPs induce apoptosis in MCF-7 cells
(2014b) with a size of 20 nm
Voicu et al. Epirubicin (Epi) – Highly homogeneous and have Colon HCT-8 Lower concentration of Fe3O4@Epi (1.95 mg/mL) was needed to
(2014) a mean diameter of 4 nm obtain tumor cell viability less than 50%, compared to free drug
which needed more amount (7.81 mg/mL) to get similar percent of
viable tumor cells
Fludarabine (Flu) Flu showed delayed tumor cells inhibitory effect where 31.25 mg/mL
of Fe3O4@Flu was needed to reduce cell viability for 24 h incubation,
yet less amount of Fe3O4@Flu (1.95 mg/mL) was sufficient to reduce
cell viability after 72 h of incubation
Javid et al. Doxorubicin (DOX) 1. polyethylene glycol (PEG) Dispersed and spherical with Ovary A2780 PTX loaded NPs showed lower cell viability for both cancer cell lines,
(2014) Paclitaxel (PTX) 2. (3-aminopropyl) particle size of 27 ± 0.7 nm OVCAR- compared to DOX loaded NPs with the same concentration. The
triethoxysilane (APTES) (DOX) and 30 ± 0.45 nm 3 result revealed significant antineoplastic effect as compared to free
(PTX) drugs
Li et al. Doxorubicin 1. Graphene oxide Nanohybrid with a lateral size Cervical HeLa The nanohybrid can be up taken by HeLa cells easily and hence
(2014c) 2. Pluronic F127 of 110 nm showed the cytotoxicity effect towards HeLa cells

2301
(continued on next page)
2302
Table 3 (continued)
Ref. Anticancer drug Surface modification Drug carrier (shape and size) Type of Cell Line Results
Cancer
Sharma et al. Doxorubicin 1. Sodium hexametaphosphate Roughly spherical in shape with Bone MG63 The anticancer drug from NPs showed sustained release profile in
(2014) hydrochloride (SHMP) size around 10 nm acidic environment, which is suitable to be used as drug carrier to
delivery anticancer drug to low pH tumor site
Kubovcikova Taxol 1. Poly(D,L-lactide-coglycolide) Mostly spherical with diameter Skin B16 Around 90% of growth inhibition was achieved in 3 days by treating
et al. (2013) (PLGA) of nanoparticles less than 300 melanoma cancerous cells with drug loaded NPs
2. Pluronic F68 nm
Chen et al. Methotrexate 1. Poly(lactide) (PLA) Spherical morphology with an Breast MCF-7 NPs showed low cytotoxicity, but drug loaded NPs showed high
(2013) 2. Polyethylene glycol average size of 10 nm and shell cytotoxicity against cancer cells, indicating the effectiveness in
(PEG) thickness of around 3 nm antitumor activity
Lv et al. Evodiamine 1. copolymer methoxy poly Spherical morphology with an Cervical HeLa The drug loaded nanocarrier showed antitumor activity at higher
(2013) (ethylene glycol)–poly(D,L- average size of 45 nm concentration (15–20 mg/mL) and exhibit a more sustained and
lactide-co-glycolide) (MPEG– controlled drug release in the intracellular compartments after
PLGA) cellular internalization
Rose et al. Epirubicin hydroxide 1. polyvinyl pyrrolidone (PVP) Spherical in shape with the Breast MCF-7 Drug loaded NPs showed the highest growth inhibition in breast
(2013) particle size of 8–10 nm Leukemia THP-1 cancer cells (81%). PVP coated NPs showed better anticancer activity
Prostate PC-3 in breast cancer cell lines than the uncoated NPs
Lung A549
Fazilati Doxorubicin 1. Folic acid Almost spherical shape with Ovary CP70 Free drug showed a lower cytotoxicity against C30 (49.2%) and CP70
(2014) particle size of 43 nm C30 (46.6%). Drug loaded NPs have a better effect towards C30 cells,
where C30 and CP70 cells reached 91% and 81.8% apoptosis
respectively after treating with drug loaded NPs
Ding et al. 10- 1. MPEG-PLGA copolymer Nearly spherical in shape with Cervical HeLa The nanoplatform has excellent in vitro antitumor efficacy compared
(2013) hydroxycamptothecin an average diameter less than Lung A549 to free drug via apoptosis activation. Cells at targeted site were killed
(HCPT) 100 nm Liver Hep G2 with the aid of external magnetic field, where the drug was directly
delivered without affecting the growth of cells at control area
Dorniani 6-mercaptopurine 1. Chitosan Generally spherical with an Leukemia WEHI-3 Drug loaded NPs did not show toxic to normal mouse fibroblast cell
et al. (2013) average diameter of 19 nm line, but showed cytotoxicity effect against cancer cell. Solvent used in
preparing drug loaded NPs has an effect in drug release study, where
drug solution prepared in dimethyl sulfoxide did not show burst
effect, but hot ethanol did
Li et al. 5-fluorouracil 1. poly(styrene-alt-maleic acid) Truncated octahedral Bladder MBT-2 Large scale cancer cells were killed after hyperthermia treatment with
(2013b) sodium nanostructure with an edge cancer cell-specific targeting NPs, which the NPs were prepared by
Salt (PSMA) length 22 nm conjugating anticancer drug (5-Fu) and anti-human epidermal
2. Poly-A15 polynucleotides growth factor receptor 2 (anti-HER2) antibody
Abbreviations: NPs, nanoparticles.

Y.P. Yew et al.


Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2303

Fig. 9 The anti-tumor efficacy in vivo with tumor-bearing BALB/C mice. BALB/c mice were subcutaneously implanted with CT-26 cells
and were given different treatment by intravenous injection of normal saline (control), DOX, MGO-PEG-CET/DOX, MGO-PEG-CET/
DOX + magnet, and MGO-PEG-CET/DOX + magnet + laser (30 mg/kg DOX). (a) The gross observation of tumor-bearing BALB/c
mice on day 0 and 14, the gross view of incised tumor and the H&E staining of the incised tumor on day 14 (bar = 200 mm); The relative
tumor volume (b) and body weight (c) were recorded. *p < 0.05 compared with control, DOX, and MGO-PEG-CET/DOX, # p < 0.05 as
compared with MGO-PEG-CET/DOX + magnet. Reproduced with permission (Lu et al., 2018).

become MGO-PEG-CET/DOX for anticancer study. The anti- tothermal therapy to control the growth of tumor. MGO-PE
tumor efficacy of MGO-PEG-CET/DOX was investigated G-CET/DOX + magnet + laser treatment could inhibit the
in vivo in xenograft tumor model in mice. The experiment tumor growth and shrank the size of tumor. Fig. 9c showed
was carried out using BALB/c with subcutaneous CT-26 the weight of the mice observed throughout the 14 days. How-
tumors of 60–100 mm3, which were subjected to treatment ever, the mice in control group were noticed to have a better
with normal saline (control) and DOX in different ways. The weight gain compared to other groups that underwent DOX
images of the tumor-bearing mice were taken on day 0 and treatment. This could be attributed to the common adverse
14 which the tumor size differences were recorded. The tumor effect from chemotherapy, but the appetite and behavior of
removed from the mice on day 14 revealed the anti-tumor the mice were not changed much throughout the period for
effects with each treatment, but to a different degree all of the mice under treatment.
(Fig. 9a). The tumor tissue on day 14 underwent H&E staining
and the results showed that necrosis of the cancer cells was 6. Conclusion and future perspectives
most substantial in MGO-PEG-CET/DOX + magnet and
MGO-PEG-CET/DOX + magnet + laser group. However, Pharmaceutical field begins to develop in recent decades and
the cells were continue growing for control, DOX and has introduced a huge number of novel drug delivery system.
MGO-PEG-CET/DOX groups. The tumor volumes were Most of them are still in incipient stage, including Fe3O4-
recorded every day until day 14 and a graph of relative tumor NPs. Plenty of factors make Fe3O4-NPs the potential nan-
volume after normalizing the tumor volume at each time point odrug carrier in drug delivery system. The usage of external
with the tumor volume at day 0 was presented (Fig. 9b). It was magnetic field which guides the Fe3O4-NPs to the specific
observed that MGO-PEG-CET/DOX + magnet and MGO- region shows the promising applications of Fe3O4-NPs in vari-
PEG-CET/DOX + magnet + laser groups revealed substan- ety of biomedical related field, particularly targeted drug deliv-
tial tumor suppression throughout the observation period ery. However, there is still no Fe3O4-based nanoparticles drug
(*p < 0.05), as compared to the control. DOX and MGO- delivery product on the market. Many intensive researches are
PEG-CET/DOX groups also showed the tumor volume reduc- yet to be done to commercialize these nanoparticles as a pro-
tions, but both of the groups did not give notable difference in duct in medical domain. Before these Fe3O4-NPs to be
tumor volume from control throughout the experiment. This launched as a product, several limitations need to be over-
tells the significance of dual targeting with external magnetic come. The methodologies in the preparation of Fe3O4-NPs
guidance, but still the MGO-PEG-CET/DOX + magnet treat- need to be improved and the characterization is the most cru-
ment fail to suppress tumor growth after day 8 with a rapid cial part. The results will show the properties of the Fe3O4-NPs
increase of tumor volume. Hence, laser light was used as pho-
2304 Y.P. Yew et al.

possessed which subsequently determine the potential applica- Malaysia-Japan International Institute of Technology, Univer-
tion Fe3O4-NPs. siti Teknologi Malaysia.
Important features that have to be taken into consideration
when selecting Fe3O4-NPs for drug delivery are the saturation References
magnetization, size, shape, surface charge, colloidal stability,
drug loading capacity and release behavior, biocompatibility Ahmed, M.J.K., Ahmaruzzaman, M., Bordoloi, M.H., 2015. Novel
and toxicity. However, the fettle of the Fe3O4-NPs in body Averrhoa carambola extract stabilized magnetite nanoparticles: a
after drug delivery is important. It is safe if they are able to green synthesis route for the removal of chlorazol black E from
eliminate from body system. But, if the Fe3O4 core is exposed, wastewater. RSC Adv. 5, 74645–74655.
it can cause several problems which correlated with neurolog- Ali, A., Hira Zafar, M.Z., ul Haq, I., Phull, A.R., Ali, J.S., Hussain,
A., 2016. Synthesis, characterization, applications, and challenges
ical disorders. Hence, the selection of Fe3O4-NPs for targeted
of iron oxide nanoparticles. Nanotechnol. Sci. Appl. 9, 49–67.
drug delivery should be chosen carefully based on the mecha-
Atarod, M., Nasrollahzadeh, M., Sajadi, S.M., 2015. Green synthesis
nism of conjugation between polymer, drug molecules and of a Cu/reduced graphene oxide/Fe3O4 nanocomposite using
Fe3O4-NPs, else the burst effect would produce toxic chemicals Euphorbia wallichii leaf extract and its application as a recyclable
that is harmful to body system. and heterogeneous catalyst for the reduction of 4-nitrophenol and
rhodamine B. RSC Adv. 5, 91532–91543.
6.1. Future works Attallah, O.A., Girgis, E., Abdel-Mottaleb, M.M., 2016. Synthesis of
non-aggregated nicotinic acid coated magnetite nanorods via
hydrothermal technique. J. Magn. Magn. Mater. 399, 58–63.
In vivo test using Fe3O4-NPs should be carried out and inves-
Awwad, A.M., Salem, N.M., 2012. A green and facile approach for
tigated thoroughly before clinical practice. This procedure can synthesis of magnetite nanoparticles. Nanosci. Nanotech. 2, 208–
study the effectiveness of Fe3O4-NPs in body system and might 213.
provide useful information to improve the desirable character- Azarifar, D., Badalkhani, O., Abbasi, Y., 2016. Silica-modified
istics of Fe3O4-NPs in targeted drug delivery study. The link- magnetite Fe3O4 nanoparticles grafted with sulfamic acid func-
age of drug loaded Fe3O4-NPs and cells should also be tional groups: an efficient heterogeneous catalyst for the synthesis
studied to understand the mechanism of cell uptake. Hence, of 3,4-dihydropyrimidin-2(1H)-one and tetrahydrobenzo[b]pyran
in this case, molecular docking simulation can be done to study derivatives. J. Sulfur Chem. 37, 656–673.
the interaction between drug carrier and cells. Besides, in order Bahadur, A., Saeed, A., Shoaib, M., Iqbal, S., Bashir, M.I., Waqas,
M., Hussain, M.N., Abbas, N., 2017. Eco-friendly synthesis of
to evaluate the efficacy of nanomedicine, preclinical research is
magnetite (Fe3O4) nanoparticles with tunable size: dielectric,
required to generate data sets that depicts nanomedicine
magnetic, thermal and optical studies. Mater. Chem. Phys. 198,
behavior, such as tumoral accumulation, intratumoral distri- 229–235.
bution, tumoral retention of the system and the additional Bamrungsap, S., Zhao, Z., Chen, T., Wang, L., Li, C., Fu, T.,
contribution of the peripheral pharmacokinetics of the nano- Tan, W., 2012. Nanotechnology in therapeutics: a focus on
medicine (Hare et al., 2017). Some key parameters that affect nanoparticles as a drug delivery system. Nanomedicine 7, 1253–
the nanomedicine efficacy must be confirmed in preclinical 1271.
testing. There are several researches are essential to this study, Bano, S., Nazir, S., Nazir, A., Munir, S., Mahmood, T., Afzal, M.,
for instance, characterization of the intra-tumoral carrier Ansari, F.L., Mazhar, K., 2016. Microwave-assisted green synthe-
retention, identification of the treatment efficacy in tumors sis of superparamagnetic nanoparticles using fruit peel extracts:
surface engineering, T2 relaxometry, and photodynamic treatment
that reaches less EPR-rich sizes and evaluation the efficacy
potential. Int. J. Nanomed. 11, 3833–3848.
of nanomedicine with appropriate dose and schedule. The data
Barahuie, F., Dorniani, D., Saifullah, B., Gothai, S., Hussein, M.Z.,
obtained from these researches can understand how the Pandurangan, A.K., Arulselvan, P., Norhaizan, M.E., 2017.
plasma, off-target tissue and tumor pharmacokinetics of the Sustained release of anticancer agent phytic acid from its chi-
nanomedicine are influenced by repeat dosing. tosan-coated magnetic nanoparticles for drug-delivery system. Int.
Numerous researches have been done in this study to over- J. Nanomed. 12, 2361–2372.
come the problems of targeted drug delivery in treating cancer Basavegowda, N., Magar, K.B.S., Mishra, K., Lee, Y.R., 2014a.
disease. All the experimental results so far provide a promising Green fabrication of ferromagnetic Fe3O4 nanoparticles and their
outcome and show the potential of Fe3O4-NPs in targeted novel catalytic applications for the synthesis of biologically
drug delivery. It is believed that Fe3O4-based nanoparticles interesting benzoxazinone and benzthioxazinone derivatives. New
J. Chem. 38, 5415–5420.
in clinical treatment is not far off anymore, where they can
Basavegowda, N., Mishra, K., Lee, Y.R., 2014b. Sonochemically
eventually be used in curing diseases rather than just research
synthesized ferromagnetic Fe3O4 nanoparticles as a recyclable
in the near future. catalyst for the preparation of pyrrolo[3, 4-c]quinoline-1,3-dione
derivatives. RSC Adv. 4, 61660–61666.
Declarations of interest Belachew, N., Devi, D.R., Basavaiah, K., 2016. Facile green synthesis
of L-methionine capped magnetite nanoparticles for adsorption of
pollutant Rhodamine B. J. Mol. Liq. 224, 713–720.
None.
Belachew, N., Rama Devi, D., Basavaiah, K., 2017. Green synthesis
and characterisation of L-Serine capped magnetite nanoparticles
Acknowledgements for removal of Rhodamine B from contaminated water. J. Exp.
Nanosci. 12, 114–128.
The authors would like to extend their gratitude and appreci- Beloqui, A., Solinı́s, M.Á., Rodrı́guez-Gascón, A., Almeida, A.J.,
ation to the members of Chemical Energy Conversions and Préat, V., 2016. Nanostructured lipid carriers: promising drug
Applications (ChECA) Research Laboratory, Department of delivery systems for future clinics. Nanomedicine. NBM 12, 143–
Environmental Engineering and Green Technology, 161.
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2305

Blaney, L., 2007. Magnetite (Fe3O4): Properties, synthesis, and Gil, S., Correia, C.R., Mano, J.F., 2015. Magnetically labeled cells
applications. with surface-modified Fe3O4 spherical and rod-shaped magnetic
Buazar, F., Baghlani-Nejazd, M.H., Badri, M., Kashisaz, M., nanoparticles for tissue engineering applications. Adv. Healthc.
Khaledi-Nasab, A., Kroushawi, F., 2016. Facile one-pot phytosyn- Mater. 4, 883–891.
thesis of magnetic nanoparticles using potato extract and their Grumezescu, A.M., Cotar, A.I., Andronescu, E., Ficai, A., Ghitulica,
catalytic activity. Starch-Starke 68, 796–804. C.D., Grumezescu, V., Vasile, B.S., Chifiriuc, M.C., 2013. In vitro
Cai, Y., Shen, Y., Xie, A., Li, S., Wang, X., 2010. Green synthesis of activity of the new water-dispersible Fe3O4@usnic acid nanostruc-
soya bean sprouts-mediated superparamagnetic Fe3O4 nanoparti- ture against planktonic and sessile bacterial cells. J. Nanopart. Res.
cles. J. Magn. Magn. Mater. 322, 2938–2943. 15, 1–10.
Cao, H., Li, J., Shen, Y., Li, S., Huang, F., Xie, A., 2014. Green Hadian-Dehkordi, L., Hosseini-Monfared, H., 2016. Enantioselective
synthesis and surface properties of Fe3O4@SA core–shell aerobic oxidation of olefins by magnetite nanoparticles at room
nanocomposites. Appl. Surf. Sci. 301, 244–249. temperature: a chiral carboxylic acid strategy. Green Chem. 18,
Chang, P.R., Yu, J., Ma, X., Anderson, D.P., 2011. Polysaccharides 497–507.
as stabilizers for the synthesis of magnetic nanoparticles. Carbo- Hanus, M.J., Harris, A.T., 2013. Nanotechnology innovations for the
hyd. Polym. 83, 640–644. construction industry. Prog. Mater. Sci. 58, 1056–1102.
Chen, F., Zhao, T., Chen, Q., Han, L., Fang, S., Chen, Z., 2013. Hardani, K., Buazar, F., Ghanemi, K., Kashisaz, M., Baghlani-
Synthesis and release behavior of methotrexate from Fe3O4/PLA– Nezhad, M., Khaledi-Naseb, A., Badri, M., 2015. Removal of toxic
PEG core/shell nanoparticles with high saturation magnetization. mercury (II) from water via Fe3O4/hydroxyapatite nanoadsorbent:
Mater. Lett. 108, 179–182. an efficient, economic and rapid approach. Am. Assoc. Sci.
Chen, T., Guo, X., Liu, X., Shi, S., Wang, J., Shi, C., Qian, Z., Zhou, Technol. J. Nanosci. 1, 11–18.
S., 2012. A strategy in the design of micellar shape for cancer Hare, J.I., Lammers, T., Ashford, M.B., Puri, S., Storm, G., Barry, S.
therapy. Adv. Healthc. Mater. 1, 214–224. T., 2017. Challenges and strategies in anti-cancer nanomedicine
Chertok, B., Moffat, B.A., David, A.E., Yu, F., Bergemann, C., development: an industry perspective. Adv. Drug Deliv. Rev. 108,
Ross, B.D., Yang, V.C., 2008. Iron oxide nanoparticles as a drug 25–38.
delivery vehicle for MRI monitored magnetic targeting of brain Harrison, R.J., Dunin-Borkowski, R.E., Putnis, A., 2002. Direct
tumors. Biomaterials 29, 487–496. imaging of nanoscale magnetic interactions in minerals. Proc. Natl.
Chin, S.F., Pang, S.C., Tan, C.H., 2011. Green synthesis of magnetite Acad. Sci. USA 99, 16556–16561.
nanoparticles (via thermal decomposition method) with control- Haw, C.Y., Mohamed, F., Chia, C.H., Radiman, S., Zakaria, S.,
lable size and shape. J. Mater. Environ. Sci. 2, 299–302. Huang, N.M., Lim, H.N., 2010. Hydrothermal synthesis of
Demir, A., Baykal, A., Sözeri, H., 2014. Green synthesis of Fe3O4 magnetite nanoparticles as MRI contrast agents. Ceram. Int. 36,
nanoparticles by one-pot saccharide-assisted hydrothermal 1417–1422.
method. Turk. J. Chem. 38, 825–836. He, D., Li, L., Bai, F., Zha, C., Shen, L., Kung, H.H., Bao, N., 2016.
Demir, A., Topkaya, R., Baykal, A., 2013. Green synthesis of One-pot synthesis of pomegranate-structured Fe3O4/carbon nano-
superparamagnetic Fe3O4 nanoparticles with maltose: Its magnetic spheres-doped graphene aerogel for high-rate lithium ion batteries.
investigation. Polyhedron 65, 282–287. Chem. Eur. J. 22, 4454–4459.
Ding, G.-B., Liu, H.-Y., Wang, Y., Lü, Y.-Y., Wu, Y., Guo, Y., Xu, Horst, M.F., Coral, D.F., van Raap, M.B.F., Alvarez, M., Lassalle,
L., 2013. Fabrication of a magnetite nanoparticle-loaded polymeric V., 2017. Hybrid nanomaterials based on gum Arabic and
nanoplatform for magnetically guided drug delivery. Chem. Res. magnetite for hyperthermia treatments. Mater. Sci. Eng., C 74,
Chin. Univ. 29, 103–109. 443–450.
Ding, L., Hu, Y., Luo, Y., Zhu, J., Wu, Y., Yu, Z., Cao, X., Peng, C., Hsieh, S., Huang, B., Hsieh, S., Wu, C., Wu, C., Lin, P., Huang, Y.,
Shi, X., Guo, R., 2016. LAPONITEÒ-stabilized iron oxide Chang, C., 2010. Green fabrication of agar-conjugated Fe3O4
nanoparticles for in vivo MR imaging of tumors. Biomater. Sci. magnetic nanoparticles. Nanotechnology 21, 1–6.
4, 474–482. Hu, Y., Mignani, S., Majoral, J.-P., Shen, M., Shi, X., 2018.
Dorniani, D., bin Hussein, M.Z., Kura, A.U., Fakurazi, S., Shaari, Construction of iron oxide nanoparticle-based hybrid platforms for
A.H., Ahmad, Z., 2013. Preparation and characterization of 6- tumor imaging and therapy. Chem. Soc. Rev. 47, 1874–1900.
mercaptopurine-coated magnetite nanoparticles as a drug delivery Hussain, I., Singh, N., Singh, A., Singh, H., Singh, S., 2016. Green
system. Drug Des. Devel. Ther. 7, 1015–1026. synthesis of nanoparticles and its potential application. Biotechnol.
El-Kassas Hala, Y., Aly-Eldeen Mohamed, A., Gharib Samiha, M., Lett. 38, 545–560.
2016. Green synthesis of iron oxide (Fe3O4) nanoparticles using Iravani, S., 2011. Green synthesis of metal nanoparticles using plants.
two selected brown seaweeds: Characterization and application for Green Chem. 13, 2638–2650.
lead bioremediation. Acta Oceanol. Sin. 35, 89–98. Javid, A., Ahmadian, S., Saboury, A.A., Kalantar, S.M., Rezaei-
El Ghandoor, H., Zidan, H., Khalil, M.M., Ismail, M., 2012. Zarchi, S., Shahzad, S., 2014. Biocompatible APTES–PEG mod-
Synthesis and some physical properties of magnetite (Fe3O4) ified magnetite nanoparticles: effective carriers of antineoplastic
nanoparticles. Int. J. Electrochem. Sci. 7, 5734–5745. agents to ovarian cancer. Appl. Biochem. Biotechnol. 173, 36–54.
Fazilati, M., 2014. Folate decorated magnetite nanoparticles: Syn- Jha, P.K., 2017. Green synthesis of magnetic Fe3O4 nanoparticles
thesis and targeted therapy against ovarian cancer. Cell Biol. Int. using Couroupita guianensis Aubl. fruit extract for their antibac-
38, 154–163. terial and cytotoxicity activities. Artif. Cells Nanomed. Biotechnol.,
Gao, S., Shi, Y., Zhang, S., Jiang, K., Yang, S., Li, Z., Takayama- 1–10
Muromachi, E., 2008. Biopolymer-assisted green synthesis of iron Kalantari, K., Ahmad, M.B., Shameli, K., Hussein, M.Z.B., Khan-
oxide nanoparticles and their magnetic properties. J. Phys. Chem. danlou, R., Khanehzaei, H., 2014. Size-controlled synthesis of
C 112, 10398–10401. Fe3O4 magnetic nanoparticles in the layers of montmorillonite. J.
Gawande, M.B., Branco, P.S., Varma, R.S., 2013. Nano-magnetite Nanomater. 2014, 1–9.
(Fe3O4) as a support for recyclable catalysts in the development of Kanagasubbulakshmi, S., Kadirvelu, K., 2017. Green synthesis of
sustainable methodologies. Chem. Soc. Rev. 42, 3371–3393. Iron oxide nanoparticles using Lagenaria siceraria and evaluation
Ghosh, S., More, P., Derle, A., Kitture, R., Kale, T., Gorain, M., of its Antimicrobial activity. Defence Life Sci. J. 2, 422–427.
Avasthi, A., Markad, P., Kundu, G.C., Kale, S., 2015. Diosgenin Kandelousi, M.S., Ellahi, R., 2015. Simulation of ferrofluid flow for
functionalized iron oxide nanoparticles as novel nanomaterial magnetic drug targeting using the lattice Boltzmann method. Z.
against breast cancer. J. Nanosci. Nanotechno. 15, 9464–9472. Naturforsch. A 70, 115–124.
2306 Y.P. Yew et al.

Karimzadeh, I., Aghazadeh, M., Doroudi, T., Ganjali, M.R., mia combined with 5-fluorouracil chemotherapy. Biomaterials 34,
Kolivand, P.H., 2017. Superparamagnetic iron oxide (Fe3O4) 7873–7883.
nanoparticles coated with PEG/PEI for biomedical applications: Li, Y., Liu, J., Dong, H., Liu, G., Hu, H., 2014c. Engineering of a
a facile and scalable preparation route based on the cathodic Pluronic F127 functionalized magnetite/graphene nanohybrid for
electrochemical deposition method. Adv. Phys. Chem. 2017, 1–7. chemophototherapy. Nanotechnology 25, 065602.
Khan, M., Mangrich, A., Schultz, J., Grasel, F., Mattoso, N., Mosca, Lin, C.-R., Chu, Y.-M., Wang, S.-C., 2006. Magnetic properties of
D., 2015. Green chemistry preparation of superparamagnetic magnetite nanoparticles prepared by mechanochemical reaction.
nanoparticles containing Fe3O4 cores in biochar. J. Anal. Appl. Mater. Lett. 60, 447–450.
Pyrol. 116, 42–48. Liu, M., Jin, H., Uchaker, E., Xie, Z., Wang, Y., Cao, G., Hou, S.,
Khandanlou, R., Ahmad, M.B., Shameli, K., Kalantari, K., 2013. Li, J., 2017. One-pot synthesis of in-situ carbon-coated Fe3O4 as a
Synthesis and characterization of rice straw/Fe3O4 nanocomposites long-life lithium-ion battery anode. Nanotechnology 28, 1–9.
by a quick precipitation method. Molecules 18, 6597–6607. Liu, W.-M., Xue, Y.-N., Peng, N., He, W.-T., Zhuo, R.-X., Huang,
Khataee, A., Kayan, B., Kalderis, D., Karimi, A., Akay, S., S.-W., 2011. Dendrimer modified magnetic iron oxide nanoparticle/
Konsolakis, M., 2017. Ultrasound-assisted removal of Acid Red DNA/PEI ternary magnetoplexes: a novel strategy for magneto-
17 using nanosized Fe3O4-loaded coffee waste hydrochar. Ultrason. fection. J. Mater. Chem. 21, 13306–13315.
Sonochem. 35, 72–80. Lu, W., Ling, M., Jia, M., Huang, P., Li, C., Yan, B., 2014. Facile
Kubovcikova, M., Koneracka, M., Zavisova, V., Muckova, M., synthesis and characterization of polyethylenimine-coated Fe3O4
Timko, M., Schmidtova, L.U., Bartos, P., Kopcansky, P., 2013. superparamagnetic nanoparticles for cancer cell separation. Mol.
Biodistribution and in vivo anticancer effects of taxol loaded Med. Report. 9, 1080–1084.
magnetic nanospheres. IEEE Trans. Magn. 49, 353–358. Lu, W., Shen, Y., Xie, A., Zhang, W., 2010. Green synthesis and
Kumar, B., Smita, K., Cumbal, L., Debut, A., 2014a. Biogenic characterization of superparamagnetic Fe3O4 nanoparticles. J.
synthesis of iron oxide nanoparticles for 2-arylbenzimidazole Magn. Magn. Mater. 322, 1828–1833.
fabrication. J. Saudi Chem. Soc. 18, 364–369. Lu, Y.-J., Lin, P.-Y., Huang, P.-H., Kuo, C.-Y., Shalumon, K.,
Kumar, B., Smita, K., Cumbal, L., Debut, A., Galeas, S., Guerrero, Chen, M.-Y., Chen, J.-P., 2018. Magnetic graphene oxide for dual
V.H., 2016. Phytosynthesis and photocatalytic activity of magnetite targeted delivery of doxorubicin and photothermal therapy.
(Fe3O4) nanoparticles using the Andean blackberry leaf. Mater. Nanomaterials 8, 193.
Chem. Phys. 179, 310–315. Lunge, S., Singh, S., Sinha, A., 2014. Magnetic iron oxide (Fe3O4)
Kumar, S.R., Priyatharshni, S., Babu, V., Mangalaraj, D., Viswa- nanoparticles from tea waste for arsenic removal. J. Magn. Magn.
nathan, C., Kannan, S., Ponpandian, N., 2014b. Quercetin Mater. 356, 21–31.
conjugated superparamagnetic magnetite nanoparticles for in-vitro Luo, Y., Yang, J., Yan, Y., Li, J., Shen, M., Zhang, G., Mignani, S.,
analysis of breast cancer cell lines for chemotherapy applications. J. Shi, X., 2015. RGD-functionalized ultrasmall iron oxide nanopar-
Colloid Interface Sci. 436, 234–242. ticles for targeted T1-weighted MR imaging of gliomas. Nanoscale
Kumfer, B.M., Shinoda, K., Jeyadevan, B., Kennedy, I.M., 2010. 7, 14538–14546.
Gas-phase flame synthesis and properties of magnetic iron oxide Lv, Y., Ding, G., Zhai, J., Guo, Y., Nie, G., Xu, L., 2013. A
nanoparticles with reduced oxidation state. J. Aerosol Sci. 41, 257– superparamagnetic Fe3O4-loaded polymeric nanocarrier for tar-
265. geted delivery of evodiamine with enhanced antitumor efficacy.
Lai, Y., Yin, W., Liu, J., Xi, R., Zhan, J., 2010. One-pot green Colloids Surf. B Biointerfaces 110, 411–418.
synthesis and bioapplication of L-arginine-capped superparamag- Ma, D., Chen, J., Luo, Y., Wang, H., Shi, X., 2017. Zwitterion-
netic Fe3O4 nanoparticles. Nanoscale Res. Lett. 5, 302–307. coated ultrasmall iron oxide nanoparticles for enhanced T1-
Latha, N., Gowri, M., 2014. Biosynthesis and characterisation of weighted magnetic resonance imaging applications. J. Mater.
Fe3O4 nanoparticles using Caricaya papaya leaves extract. Int. J. Chem. B 5, 7267–7273.
Sci. Res. 3, 1551–1556. Mahdavi, M., Ahmad, M.B., Haron, M.J., Namvar, F., Nadi, B.,
Lemine, O., Omri, K., Zhang, B., El Mir, L., Sajieddine, M., Rahman, M.Z.A., Amin, J., 2013a. Synthesis, surface modification
Alyamani, A., Bououdina, M., 2012. Sol–gel synthesis of 8nm and characterisation of biocompatible magnetic iron oxide
magnetite (Fe3O4) nanoparticles and their magnetic properties. nanoparticles for biomedical applications. Molecules 18, 7533–
Superlattices Microstruct. 52, 793–799. 7548.
Li, J., He, Y., Sun, W., Luo, Y., Cai, H., Pan, Y., Shen, M., Xia, J., Mahdavi, M., Namvar, F., Ahmad, M.B., Mohamad, R., 2013b.
Shi, X., 2014a. Hyaluronic acid-modified hydrothermally synthe- Green biosynthesis and characterization of magnetic iron oxide
sized iron oxide nanoparticles for targeted tumor MR imaging. (Fe3O4) nanoparticles using seaweed (Sargassum muticum) aque-
Biomaterials 35, 3666–3677. ous extract. Molecules 18, 5954–5964.
Li, J., Hu, Y., Yang, J., Wei, P., Sun, W., Shen, M., Zhang, G., Shi, Malekzadeh, A.M., Ramazani, A., Rezaei, S.J.T., Niknejad, H.,
X., 2015. Hyaluronic acid-modified Fe3O4@Au core/shell nanos- 2017. Design and construction of multifunctional hyperbranched
tars for multimodal imaging and photothermal therapy of tumors. polymers coated magnetite nanoparticles for both targeting mag-
Biomaterials 38, 10–21. netic resonance imaging and cancer therapy. J. Colloid Interface
Li, J., Shi, X., Shen, M., 2014b. Hydrothermal synthesis and Sci. 490, 64–73.
functionalization of iron oxide nanoparticles for MR imaging Namanga, J., Foba, J., Ndinteh, D.T., Yufanyi, D.M., Krause, R.W.
applications. Part. Part. Syst. Char. 31, 1223–1237. M., 2013. Synthesis and magnetic properties of a superparamag-
Li, J., Wang, S., Shi, X., Shen, M., 2017. Aqueous-phase synthesis of netic nanocomposite pectin-magnetite nanocomposite. J. Nano-
iron oxide nanoparticles and composites for cancer diagnosis and mater. 2013, 1–8.
therapy. Adv. Colloid Interface Sci. 249, 374–385. Narayanan, S., Sathy, B.N., Mony, U., Koyakutty, M., Nair, S.V.,
Li, J., Zheng, L., Cai, H., Sun, W., Shen, M., Zhang, G., Shi, X., Menon, D., 2011. Biocompatible magnetite/gold nanohybrid con-
2013a. Polyethyleneimine-mediated synthesis of folic acid-targeted trast agents via green chemistry for MRI and CT bioimaging. ACS
iron oxide nanoparticles for in vivo tumor MR imaging. Bioma- Appl. Mater. Interfaces 4, 251–260.
terials 34, 8382–8392. Nene, A.G., Takahashi, M., Wakita, K., Umeno, M., 2016. Size
Li, T.-J., Huang, C.-C., Ruan, P.-W., Chuang, K.-Y., Huang, K.-J., controlled synthesis of Fe3O4 nanoparticles by ascorbic acid
Shieh, D.-B., Yeh, C.-S., 2013b. In vivo anti-cancer efficacy of mediated reduction of Fe(acac)3 without using capping agent. J.
magnetite nanocrystal-based system using locoregional hyperther- Nano Res. 40, 8–19.
Green biosynthesis of superparamagnetic magnetite Fe3O4 nanoparticles 2307

Ngernpimai, S., Thomas, C., Maensiri, S., Siri, S., 2012. Stability and Sheikholeslami, M., Vajravelu, K., 2017. Forced convection heat
cytotoxicity of well-dispersed magnetite nanoparticles prepared by transfer in Fe3O4-ethylene glycol nanofluid under the influence of
hydrothermal method. Adv. Mat. Res. 506, 122–125. Coulomb force. J. Mol. Liq. 233, 203–210.
Niraimathee, V., Subha, V., Ravindran, R.E., Renganathan, S., 2016. Shen, L., Qiao, Y., Guo, Y., Meng, S., Yang, G., Wu, M., Zhao, J.,
Green synthesis of iron oxide nanoparticles from Mimosa pudica 2014. Facile co-precipitation synthesis of shape-controlled mag-
root extract. Int. J. Environ. Sustain. Dev. 15, 227–240. netite nanoparticles. Ceram. Int. 40, 1519–1524.
Paiva, D., Andrade, A., Pereira, M., Fabris, J., Domingues, R., Shi, H., Tan, L., Du, Q., Chen, X., Li, L., Liu, T., Fu, C., Liu, H.,
Alvarenga, M., 2015. Novel protocol for the solid-state synthesis Meng, X., 2014. Green synthesis of Fe3O4 nanoparticles with
of magnetite for medical practices. Hyperfine Interact. 232, 19– controlled morphologies using urease and their application in dye
27. adsorption. Dalton Trans. 43, 12474–12479.
Park, J.Y., Choi, E.S., Baek, M.J., Lee, G.H., 2009. Colloidal Shirkhodaie, M., Hossein Beyki, M., Shemirani, F., 2016. Biogenic
stability of amino acid coated magnetite nanoparticles in physio- synthesis of magnetic perlite@iron oxide composite: application as
logical fluid. Mater. Lett. 63, 379–381. a green support for dye removal. Desalin. Water Treat. 57, 11859–
Patra, J.K., Ali, M.S., Oh, I.-G., Baek, K.-H., 2017. Proteasome 11871.
inhibitory, antioxidant, and synergistic antibacterial and antican- Shrifian-Esfahni, A., Salehi, M.T., Nasr-Esfahni, M., Ekramian, E.,
didal activity of green biosynthesized magnetic Fe3O4 nanoparticles 2015. Chitosan-modified superparamgnetic iron oxide nanoparti-
using the aqueous extract of corn (Zea mays L.) ear leaves. Artif. cles: design, fabrication, characterization and antibacterial activity.
Cells Nanomed. Biotechnol. 45, 349–356. Chemik 69, 19–32.
Patra, J.K., Baek, K.-H., 2014. Green nanobiotechnology: factors Sirdeshpande, K.D., Sridhar, A., Cholkar, K.M., Selvaraj, R., 2018.
affecting synthesis and characterization techniques. J. Nanomater. Structural characterization of mesoporous magnetite nanoparticles
2014, 1–12. synthesized using the leaf extract of Calliandra haematocephala and
Petcharoen, K., Sirivat, A., 2012. Synthesis and characterization of their photocatalytic degradation of malachite green dye. Appl.
magnetite nanoparticles via the chemical co-precipitation method. Nanosci., 1–9
Mater. Sci. Eng. B 177, 421–427. Song, M., Zhang, Y., Hu, S., Song, L., Dong, J., Chen, Z., Gu, N.,
Pham, X.N., Nguyen, T.P., Pham, T.N., Tran, T.T.N., Tran, T.V.T., 2012. Influence of morphology and surface exchange reaction on
2016. Synthesis and characterization of chitosan-coated magnetite magnetic properties of monodisperse magnetite nanoparticles.
nanoparticles and their application in curcumin drug delivery. Adv. Colloids Surf. A Physicochem. Eng. Asp. 408, 114–121.
Nat. Sci.: Nanosci. Nanotechnol. 7, 1–9. Sun, W., Mignani, S., Shen, M., Shi, X., 2016. Dendrimer-based
Phumying, S., Labuayai, S., Thomas, C., Amornkitbamrung, V., magnetic iron oxide nanoparticles: their synthesis and biomedical
Swatsitang, E., Maensiri, S., 2013. Aloe vera plant-extracted applications. Drug Discov. Today 21, 1873–1885.
solution hydrothermal synthesis and magnetic properties of mag- Sun, X., Zheng, C., Zhang, F., Yang, Y., Wu, G., Yu, A., Guan, N.,
netite (Fe3O4) nanoparticles. Appl. Phys. A 111, 1187–1193. 2009. Size-controlled synthesis of magnetite (Fe3O4) nanoparticles
Qin, Z., Jiao, X., Chen, D., 2011. Preparation of coral-like magnetite coated with glucose and gluconic acid from a single Fe(III)
through a glucose-assisted solvothermal synthesis. CrystEngComm precursor by a sucrose bifunctional hydrothermal method. J. Phys.
13, 4646–4651. Chem. C 113, 16002–16008.
Rajendran, S.P., Sengodan, K., 2017. Synthesis and characterization Sun, Y., Li, J., Wang, Y., Ding, C., Lin, Y., Sun, W., Luo, C., 2017.
of zinc oxide and iron oxide nanoparticles using Sesbania grandi- A chemiluminescence biosensor based on the adsorption recogni-
flora leaf extract as reducing agent. J. Nanosci. 2017, 1–7. tion function between Fe3O4@SiO2@GO polymers and DNA for
Rehana, D., Haleel, A.K., Rahiman, A.K., 2015. Hydroxy, car- ultrasensitive detection of DNA. Spectrochim. Acta. A Mol.
boxylic and amino acid functionalized superparamagnetic iron Biomol. Spectrosc. 178, 1–7.
oxide nanoparticles: synthesis, characterization and in vitro anti- Taghavi, F., Saljooghi, A.S., Gholizadeh, M., Ramezani, M., 2016.
cancer studies. J. Chem. Sci. 127, 1155–1166. Deferasirox-coated iron oxide nanoparticles as a potential cyto-
Rose, P.A., Praseetha, P., Bhagat, M., Alexander, P., Abdeen, S., toxic agent. Med. Chem. Commun. 7, 2290–2298.
Chavali, M., 2013. Drug embedded PVP coated magnetic nanopar- Teja, A.S., Koh, P.-Y., 2009. Synthesis, properties, and applications
ticles for targeted killing of breast cancer cells. Technol. Cancer of magnetic iron oxide nanoparticles. Prog. Cryst. Growth Char-
Res. Treat. 12, 463–472. act. Mater. 55, 22–45.
Senthil, M., Ramesh, C., 2012. Biogenic synthesis of Fe3O4 nanopar- Theerdhala, S., Bahadur, D., Vitta, S., Perkas, N., Zhong, Z.,
ticles using Tridax procumbens leaf extract and its antibacterial Gedanken, A., 2010. Sonochemical stabilization of ultrafine
activity on Pseudomonas aeruginosa. Dig. J. Nanomater. Biostruct. colloidal biocompatible magnetite nanoparticles using amino acid,
7, 1655–1660. L-arginine, for possible bio applications. Ultrason. Sonochem. 17,
Shahabadi, N., Falsafi, M., Mansouri, K., 2016. Improving antipro- 730–737.
liferative effect of the anticancer drug cytarabine on human Venkateswarlu, S., Kumar, B.N., Prasad, C., Venkateswarlu, P.,
promyelocytic leukemia cells by coating on Fe3O4@SiO2 nanopar- Jyothi, N., 2014a. Bio-inspired green synthesis of Fe3O4 spherical
ticles. Colloids Surf. B Biointerfaces 141, 213–222. magnetic nanoparticles using Syzygium cumini seed extract.
Sharma, P., Rana, S., Barick, K.C., Kumar, C., Salunke, H.G., Physica B Condens. Matter 449, 67–71.
Hassan, P.A., 2014. Biocompatible phosphate anchored Fe3O4 Venkateswarlu, S., Kumar, B.N., Prathima, B., SubbaRao, Y.,
nanocarriers for drug delivery and hyperthermia. New J. Chem. 38, Jyothi, N.V.V., 2014b. A novel green synthesis of Fe3O4 magnetic
5500–5508. nanorods using Punica Granatum rind extract and its application
Sheikholeslami, M., Sadoughi, M.K., 2017. Numerical modeling for for removal of Pb(II) from aqueous environment. Arab. J. Chem.
Fe3O4-water nanofluid flow in porous medium considering MFD Venkateswarlu, S., Rao, Y.S., Balaji, T., Prathima, B., Jyothi, N.,
viscosity. J. Mol. Liq. 242, 255–264. 2013. Biogenic synthesis of Fe3O4 magnetic nanoparticles using
Sheikholeslami, M., Shehzad, S., 2017. CVFEM for influence of plantain peel extract. Mater. Lett. 100, 241–244.
external magnetic source on Fe3O4-H2O nanofluid behavior in a Venkateswarlu, S., Yoon, M., 2015a. Rapid removal of cadmium ions
permeable cavity considering shape effect. Int. J. Heat Mass using green-synthesized Fe3O4 nanoparticles capped with diethyl-4-
Transf. 115, 180–191. (4amino-5-mercapto-4H-1,2,4-triazol-3-yl) phenyl phosphonate.
Sheikholeslami, M., Shehzad, S., 2018. Numerical analysis of Fe3O4– RSC Adv. 5, 65444–65453.
H2O nanofluid flow in permeable media under the effect of external Venkateswarlu, S., Yoon, M., 2015b. Surfactant-free green synthesis
magnetic source. Int. J. Heat Mass Transf. 118, 182–192. of Fe3O4 nanoparticles capped with 3,4-dihydroxyphenethylcar-
2308 Y.P. Yew et al.

bamodithioate: stable recyclable magnetic nanoparticles for the Xiao, L., Mertens, M., Wortmann, L., Kremer, S., Valldor, M.,
rapid and efficient removal of Hg(II) ions from water. Dalton Lammers, T., Kiessling, F., Mathur, S., 2015a. Enhanced in vitro
Trans. 44, 18427–18437. and in vivo cellular imaging with green tea coated water-soluble
Venugopal, I., Pernal, S., Duproz, A., Bentley, J., Engelhard, H., iron oxide nanocrystals. ACS Appl. Mater. Interfaces 7, 6530–
Linninger, A., 2016. Magnetic field-enhanced cellular uptake of 6540.
doxorubicin loaded magnetic nanoparticles for tumor treatment. Xiao, S., Castro, R., Rodrigues, J., Shi, X., Tomás, H., 2015b.
Mater. Res. Express 3, 1–13. PAMAM dendrimer/pDNA functionalized-magnetic iron oxide
Voicu, G., Crica, L.E., Fufa, O., Moraru, L.I., Popescu, R.C., Purcel, nanoparticles for gene delivery. J. Biomed. Nanotechnol. 11, 1370–
G., Stoilescu, M.C., Grumezescu, A.M., Bleotu, C., Holban, A.M., 1384.
2014. Magnetite nanostructures functionalized with cytostatic Yew, Y.P., Shameli, K., Miyake, M., Kuwano, N., Khairudin, N.B.
drugs exhibit great anti-tumoral properties without application of B.A., Mohamad, S.E.B., Lee, K.X., 2016. Green synthesis of
high amplitude alternating magnetic fields. Rom. J. Morphol. magnetite (Fe3O4) nanoparticles using seaweed (Kappaphycus
Embryol. 55, 357–362. alvarezii) extract. Nanoscale Res. Lett. 11, 1–7.
Wahajuddin, S.A., 2012. Superparamagnetic iron oxide nanoparti- Yuvakkumar, R., Hong, S., 2014. Green synthesis of spinel magnetite
cles: magnetic nanoplatforms as drug carriers. Int. J. Nanomed. 7, iron oxide nanoparticles. Adv. Mat. Res. 1051, 39–42.
3445–3471. Zhang, L., Dong, W.-F., Sun, H.-B., 2013. Multifunctional super-
Wang, L., Wang, M., Topham, P.D., Huang, Y., 2012. Fabrication of paramagnetic iron oxide nanoparticles: design, synthesis and
magnetic drug-loaded polymeric composite nanofibres and their biomedical photonic applications. Nanoscale 5, 7664–7684.
drug release characteristics. RSC Adv. 2, 2433–2438. Zhao, H., Saatchi, K., Häfeli, U.O., 2009. Preparation of biodegrad-
Wang, S., Zhang, Z., Liu, B., 2015. Catalytic conversion of fructose able magnetic microspheres with poly(lactic acid)-coated mag-
and 5-hydroxymethylfurfural into 2,5-furandicarboxylic acid over a netite. J. Magn. Magn. Mater. 321, 1356–1363.
recyclable Fe3O4–CoOx magnetite nanocatalyst. ACS Sustain. Zhou, W., He, W., Zhong, S., Wang, Y., Zhao, H., Li, Z., Yan, S.,
Chem. Eng. 3, 406–412. 2009. Biosynthesis and magnetic properties of mesoporous Fe3O4
Wang, Z., Zhu, H., Wang, X., Yang, F., Yang, X., 2009. One-pot composites. J. Magn. Magn. Mater. 321, 1025–1028.
green synthesis of biocompatible arginine-stabilized magnetic
nanoparticles. Nanotechnology 20, 1–10.

You might also like