ArticlePDF AvailableLiterature Review

Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles

Authors:

Abstract and Figures

In this review, we have presented the latest results and highlights on biomedical applications of the group of noble metal nanoparticles and their compounds, such as gold, silver, platinum, and the group of magnetic nanoparticles, such as cobalt, nickel, iron, and their compounds. Their most important related compounds are also discussed in biomedical applications for treating various diseases, typically as cancers . At present, both physical and chemical methods have been proved very successful to synthesize, shape, control, and produce metal- and oxide-based homogeneous particle systems, e.g. nanoparticles and microparticles. Therefore, we have mainly focused on functional magnetic nanoparticles for nanomedicine because of their high bioadaptability to the organs inside human body. Here, bioconjugation techniques are very crucial to link nanoparticles with conventional drugs, nanodrugs, biomolecules or polymers for biomedical applications. Biofunctionalization of engineered nanoparticles for biomedicine is shown respective to in vitro and in vivo analysis protocols that typically include drug delivery, hyperthermia therapy, magnetic resonance imaging (MRI), and recent outstanding progress in sweep imaging technique with Fourier transformation (SWIFT) MRI. The latter can be especially applied using magnetic nanoparticles, such as Co-, Fe-, Ni-based nanoparticles, α-Fe2O3, and Fe3O4 oxide nanoparticles for analysis and treatment of malignancies. Therefore, this review focuses on recent results of scientists, and related research on diagnosis and treatment methods of common and dangerous diseases by biomedical engineered nanoparticles. Importantly, nanosysems (nanoparticles) or microsystems (microparticles) or hybrid micronano systems are shortly introduced into nanomedicine. Here, Fe oxide nanoparticles ultimately enable potential and applicable technologies for tumor-targeted imaging and therapy. Finally, we have shown the latest aspects of the most important Fe-based particle systems, such as Fe, α-Fe2O3, Fe3O4, Fe–FexOy oxide core–shell nanoparticles, and CoFe2O4–MnFe2O4 core–shell nanoparticles for nanomedicine in the efficient treatment of large tumors at low cost in near future.
Content may be subject to copyright.
Copyright © 2015 American Scientific Publishers
All rights reserved
Printed in the United States of America
Review
Journal of
Nanoscience and Nanotechnology
Vol. 15, 10091–10107, 2015
www.aspbs.com/jnn
Biomedical Applications of Advanced Multifunctional
Magnetic Nanoparticles
Nguyen Viet Long123, Yong Yang1, Toshiharu Teranishi3,
Cao Minh Thi4, Yanqin Cao1, and Masayuki Nogami5
1State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics,
Chinese Academy of Sciences, 1295, Dingxi Road, Shanghai 200050, China
2Posts and Telecommunications Institute of Technology, km 10 Nguyen Trai, Hanoi 100000, Vietnam
3Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
4Ho Chi Minh City University of Technology, 144/24 Dien Bien Phu, Ward 25, BinhThach, Ho Chi Minh City 700000, Vietnam
5Toyota Physical and Chemical Research Institute, 41-1 Yokomichi Nagakute, 480-1192, Japan
In this review, we have presented the latest results and highlights on biomedical applications of
a class of noble metal nanoparticles, such as gold, silver and platinum, and a class of magnetic
nanoparticles, such as cobalt, nickel and iron. Their most important related compounds are also
discussed for biomedical applications for treating various diseases, typically as cancers. At present,
both physical and chemical methods have been proved very successful to synthesize, shape,
control, and produce metal- and oxide-based homogeneous particle systems, e.g., nanoparticles
and microparticles. Therefore, we have mainly focused on functional magnetic nanoparticles for
nanomedicine because of their high bioadaptability to the organs inside human body. Here, bio-
conjugation techniques are very crucial to link nanoparticles with conventional drugs, nanodrugs,
biomolecules or polymers for biomedical applications. Biofunctionalization of engineered nanopar-
ticles for biomedicine is shown respective to in vitro and in vivo analysis protocols that typically
include drug delivery, hyperthermia therapy, magnetic resonance imaging (MRI), and recent out-
standing progress in sweep imaging technique with Fourier transformation (SWIFT) MRI. The latter
can be especially applied using magnetic nanoparticles, such as Co-, Fe-, Ni-based nanoparticles,
-Fe2O3, and Fe3O4oxide nanoparticles for analysis and treatment of malignancies. Therefore, this
review focuses on recent results of scientists, and related research on diagnosis and treatment
methods of common and dangerous diseases by biomedical engineered nanoparticles. Importantly,
nanosysems (nanoparticles) or microsystems (microparticles) or hybrid micronano systems are
shortly introduced into nanomedicine. Here, Fe oxide nanoparticles ultimately enable potential and
applicable technologies for tumor-targeted imaging and therapy. Finally, we have shown the latest
aspects of the most important Fe-based particle systems, such as Fe, -Fe2O3,Fe
3O4, Fe–FexOy
oxide core–shell nanoparticles, and CoFe2O4–MnFe2O4core–shell nanoparticles for nanomedicine
in the efficient treatment of large tumors at low cost in near future.
Keywords: Fe Nanoparticles, Fe Oxide Nanoparticles, Au Nanoparticles, Ag Nanoparticles,
Pt Nanoparticles, Drug Delivery, Hyperthermia, Magnetic Resonance Imaging, Drug,
Cancers, Tumors, Cancer Therapy.
CONTENTS
1. Introduction . .......................................10092
2. Nanoparticles . ......................................10095
2.1. Metal Nanoparticles . .............................10097
2.2. Magnetic Nanoparticles ...........................10098
2.3. Alloy and Core–Shell Nanoparticles . . . ..............10098
Author to whom correspondence should be addressed.
3. Synthesis..........................................10099
4. BiomedicalApplication ...............................10099
5. Drug Delivery ......................................10100
6. Hyperthermia.......................................10101
7. MagneticResonance Imaging ..........................10102
8. Conclusions........................................10103
Acknowledgments . ..................................10104
Referencesand Notes ................................10104
J. Nanosci. Nanotechnol. 2015, Vol. 15, No. 12 1533-4880/2015/15/10091/017 doi:10.1166/jnn.2015.11691 10091
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
1. INTRODUCTION
At present, biology and medicine, e.g., traditional ther-
apy, are considered as the traditional methods of treat-
ing various diseases through conventional materials.
Nguyen Viet Long He is Young researcher at Shanghai Institute of Ceramics, Chinese
Academy of Sciences, China. His interest focuses on new functional magnetic metal and
oxide nanoparticles with bio-conjugates for biomedical applications in Nanomedicine. He
was a Researcher in Kyushu University, Fukuoka, Japan. He received B.Sc. degree of
Physics in Solid State Physics at Department of Physics, Hanoi National University of
Education, Hanoi, Vietnam. He received M.Eng. degree in Semiconductor, and Ph.D.
degree in Optical and Photonic materials at International Training Institute for Materials
Science, Hanoi University of Science and Technology, Hanoi. He was a Lecturer in Gen-
eral Physics. Then, he worked as a Researcher in Department of Materials Science and
Engineering, Nagoya Institute of Technology, Nagoya, Japan. His main research directions
of Pt and Pd based catalysts are focused on fuel cells, and energy conversion with Pro-
fessor Masayuki Nogami. He worked as researcher or visiting Professor in Kyushu University, Fukuoka, Japan. He
became a member of Laboratory for Nanotechnology, Vietnam National University at Ho Chi Minh City.
Yong Yang He works at the Shanghai Institute of Ceramics, Chinese Academy of Sci-
ences. He received B.Eng. and M.Eng., Department of Materials Science and Engineering,
from Shanxi University of Science and Technology, China. He also received Dr. Eng.,
Shanghai Institute of Ceramics, Chinese Academy of Sciences, China. He worked as JSPS
postdoctoral research fellow and project Associate Professor in Department of Materials
Science and Engineering, Nagoya Institute of Technology, Nagoya, Japan. At present, he
was officially appointed as Professor in Shanghai Institute of Ceramics, Chinese Academy
of Sciences. His main interest in scientific areas of research are focused on the prepa-
ration and self-assembly of metal nanomaterials, catalytic properties and optical proper-
ties such as Surface Plasmon Resonance (SPR) for potential applications SPR sensors,
Surface-enhanced Raman spectroscopy (SERS), and nonlinear optical properties, surface
modification of materials and optical film. He is the author of more than 60 scientific publications in international
peer-reviewed journals.
Toshiharu Teranishi He is a professor at Institute for Chemical Research, Kyoto Univer-
sity. He received his Ph.D. from the University of Tokyo under the direction of Professor
N. Toshima in 1994. He spent seven and a half years at Japan Advanced Institute of Sci-
ence and Technology as an Assistant Professor and an Associate Professor. In 2004, he
moved to University of Tsukuba as a Full Professor, and moved to Kyoto University in
2011. His current research interests include precise structural control of inorganic nano-
materials and structure-specific functions for high-performance devices and photo-energy
conversion.
Cao Minh Thi He studied in Mathematics and Physics, and Solid State Physics, in Ho
Chi Minh University of Technology, 144/24 Dien Bien Phu, Ward 25, Binh Thanh, Ho Chi
Minh City. He received B.sc. in Physics, Hanoi National University of Education (HNUE)
in 1960. Then, he studied in Mathematics and Physics at Lomonosov Moscow State Uni-
versity (1966), and received Ph.D. in Mathematics and Physics at Lomonosov Moscow
State University (1970). He was also the Lecturer in Physics at HNUE, Vietnam (1960),
Lecturer of Faculty of Physics, HNUE in the years of 1972–1975. He was appointed by
Vietnamese Leaders of the Vietnam country as vice President of Ho Chi Minh City Univer-
sity of Education or Saigon National Pedagogical University (1975), President of Ho Chi
Minh City University of Education (Saigon College) (1981), Associate professor (1984),
Deputy Director or Leader of Sector of Education and Training in the entire Ho Chi Minh
City (1989). Now He is Vice-president of Vietnam Physical Society, President of Ho Chi Minh City Physical Society.
Recently, nanomedicine science has appeared through the
application of nanomaterials and nanotechnology com-
bined with above traditional methods.1–5 To obtain bio-
logical applications, particle systems must be combined
10092 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
Yanqin Cao She studies at the Shanghai Institute of Ceramics (SIC), Chinese Academy of
Sciences (CAS). She received B.Eng., Department of Materials Science and Engineering,
Wuhan University of Science and Technology, China. Her research focuses on the con-
trollable synthesis of noble metal (Pt, Pd, Au, and Ag) nanostructures and their Catalytic
characterization and Surface enhanced Raman spectroscopy (SERS).
Masayuki Nogami He is Senior researcher at Toyota Physical and Chemical Research
Institute. He worked at Nagoya Institute of Technology (NIT), Nagoya, Japan. He received
B.S. in Ceramics, then M.S. in Ceramics at NIT, and Dr. Eng. at Osaka University. He is
Professor Emeritus at NIT, President of The Japanese Sol–Gel Society, Senior Researcher
at Nagoya Industrial Science Research Institute, Senior Researcher at National Industrial
Research Institute, Osaka, Visiting Research Associate at Rensselaer Polytechnic Institute,
USA, and Associate Professor at Aichi Institute of Technology. He has over 400 pub-
lications, 20 books, and 20 patents. He was co-editor to Journal Sol–Gel Science and
Technology (Springer-USA), Director Chairman of Glass Division in The Ceramics Soci-
ety, Japan, Visiting Professor at National Engineering College for Industrial Ceramics,
France. He was Senior Researcher at Laboratory of Science of Ceramic Processes and of
Surface Treatments, National Research Council (CNRs), French. In 2012–2013, he was an International Scientist at
Shanghai Institute of Ceramics, Chinese Academy of Science, China.
with numerous bioconjugates.2For example, researchers
need to prepare conjugates and labeled molecules for use
in nanomedicine. Fundamentally, the special antibodies
for cancers can be used.2Today, particle systems can be
prepared in the various sized ranges, as small as macro-
molecules (10 nm), and larger up to approximately the
diameter of cells, such as red blood cells (10 m).2So far,
various engineered nanoparticles have been very poten-
tially used for clinical and biomedical applications, such as
polymeric nanoparticles, carbon nanotubes (CNTs), silica
(SiO2, silver (Ag) nanoparticles, gold (Au) nanoparticles,
quantum dots (QDs), magnetic nanoparticles, superpara-
magnetic iron (Fe) oxide nanoparticles (SIONs), carbon
nanoparticles, and various nanoparticles and nanomaterials
(Figs. 1–3).1–6 They are very important particle structures,
such as micro-, nano-, and micronano-systems as the core
platforms for drug delivery systems (DDSs). These specific
formulations can be bioconjugated, typically with targeting
biomolecules, nanodrugs or traditional drugs, for therapeu-
tic and diagnostic approaches. Thus, DDSs are used to
enhance biocompatibility, bioavailability and pharmacoki-
netics of therapeutics or nanotherapeutics.1–5 According to
the improved research directions in Japan, nanomedicine
has been focused on diagnostic devices, ultrafine imaging
systems, invasive and noninvasive medical devices, espe-
cially on early point-of-care diagnosis for the detection and
treatment of several diseases, such as malignant/benign
tumors at any beginning stages of brain, muscle, lung,
kidney, liver etc. Therefore, nanobiotechnology provides
an integration of detection, diagnosis and therapy as well
as modern molecular imaging and living-cell detection.11
In the past, through an interesting research, therapeutic
DDSs using gas-filled microspheres and a therapeutic were
created, and various methods for employing such micro-
spheres in therapeutic drug delivery applications were also
provided.7In one typical case, the DDSs comprising gas-
filled liposomes having encapsulated a drug were preferred
in drug delivery applications. To provide therapeutic deliv-
ery to organs such as the liver and to allow differentia-
tion of tumor from normal tissue, smaller microspheres of
around 30–100 nm were preferred. These systems (from
several nm to tens m in size) greatly offer drug delivery
applications in common diseases in humans.7Alterna-
tively, they were designed to replace invasive adminis-
trations. Therefore, those DDSs were suitably used to
improve bioavailability, biocompatibility, and pharmacoki-
netics of therapeutics.78For example, albumin described
as a plasma protein was used for DDS approach. Thus,
a nanoDDS is created involving various nanocarriers. In
addition, the nanoDDSs offer most advantages of con-
venience, flexibility, and efficacy in delivering drugs in
diseased tissue, increase the solubility of drugs in aque-
ous media, overcome various kinds of biological bar-
riers, such as blood brain barrier (BBB), and increase
chemical stability of the drug to the diseased areas.910
Most solid tumors possess unique pathophysiological
characteristics that are not observed in normal tissues or
organs, such as extensive angiogenesis and hence hyper-
vasculature, defective vascular architecture, impaired lym-
phatic drainage/recovery system, and greatly increased
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10093
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
Biomedical functionalization
(a) Polymeric micelles (b) Dendrimer
Multifunctional
dendritic carrier
(c) Liposome carrier
(d) Functional
core-shell quantum dot
(f) Drug is bioconjugated with
polymer-protected Au nanoparticle
(e) SiO2nanoparticle
with anticancer (CPT)
–600 –400 –200 0 200 400 600
–1.0
–0.8
–0.6
–0.4
–0.2
0.0
0.2
0.4
0.6
0.8
1.0
HC
MR Sample 3
Sample 2
Sample 1
M(emu/g)
H(Oe)
–15000 –10000 –5000 0 5000 10000 15000
–3
–2
–1
0
1
2
3Sample 3
Sample 2
Sample 1
M(emu/g)
H(Oe)
(I) Types of Nanoparticles and Microparticles
Pt nanoparticles
(size range: 10 nm)
(size range: 10
μ
m)
α-Fe2O3
microparticles
(VI) α-Fe2O3 particles (Micronano system)
(IV) Alloy/Coreshell system
Pt-Pd
Pd (shell)
Pt (core)
(V)
(II) Nanosystem (III) Microsystem
Biomedical Functional Particles
Functional nanoparticles
Functional microparticles
Functional micronano particles
Hydrophilic
Shell
Hydrophobic
Core
Large pores of SiO2
(size range: 25 nm)
Figure 1. (I)–(IV), and (VI) Typical functional engineered nanoparticles for nanosystems in potential biomedical applications. Reprinted with permis-
sion from [6], (a) N. V. Long, et al., Curr. Phys. Chem. 4, 1 (2014), © 2014. All Rights Reserved. (II) Pt nanoparticles. Reprinted with permission
from [63], N. V. Long, et al., New J. Chem. 36 (2012), © 2012. All Rights Reserved. (III) -Fe2O3microparticles, Reprinted with permission from [79, 80],
N. V. Long, et al., RSC Adv. 4(2014), © 2014. All Rights Reserved, N. V. Long, et al., RSC Adv. 4, 6383 (2014), © 2014. (IV) HRTEM images of Pt–Pd
core–shell nanoparticles. Reprinted with permission from [34], N. V. Long, et al., Acta Mater. 59, 2901 (2011). © 2011. All Rights Reserved. (V) M–H
curve of new -Fe2O3microparticles with grain and grain boundary structure showing supermagnetic nature for hyperthermia therapy. (VI) SEM images
of new -Fe2O3microparticles. Reprinted with permission from [78], N. V. Long, et al., Colloids Surf., A 456 (2014). © 2014. All Rights Reserved.
production of a number of permeability mediators. The
phenomenon of enhanced permeability and retention
(EPR) effect for lipid and macromolecular agents was
observed to be universal in solid tumors in the find-
ings of Maeda and co-workers.910 The enhanced vascular
permeability will sustain an adequate supply of nutrients
and oxygen for rapid tumor growth. On the other hand, the
EPR provides a great opportunity for more selective tar-
geting of lipid- or polymer-conjugated anti-cancer drugs,
such as SMANCS and PK-1 to the tumor. The important
10094 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
(A)
(C)
Fe3O4
Fe-Pt
(B)
Fe3O4-SiO2
Nanorods
Fe-Pt Fe-Pt
Fe-Pt
Fe-Pt Fe-Pt
Figure 2. (A) TEM images of magnetite nanoparticles (12 nm).
Reprinted with permisson from [117], (a) J. Park, et al., Nat. Mater.
3, 891 (2004). © 2004. All Rights Reserved. (B) Silica shell-magnetic
core nanoparticles. Reprinted with permission from [117], (b) C. Vogt,
et al., J. Nanopart. Res. 12, 1137 (2010). © 2010. (C) Types of magnetic
Fe–Pt nanoparticles by the chemical methods. Reprinted with permission
from [55], D. Ung, et al., Cryst. Eng. Comm. 11, 1309 (2009). © 2009.
American Scientific Publishers.
characteristics of EPR and its modulation for improv-
ing delivery of macromolecular drugs to the tumor are
described. The EPR effect showed evidences of vascula-
ture in tumors and healthy organs or tissues. Nonethe-
less, blood vessels in tumors are leakier according to
their very fast growth. Interestingly, tumor cells are less
densely packed than cells in healthy tissue. The vascu-
lar permeability effect in tumor tissues, macromolecu-
lar drug delivery, and pathophysiology of tumor vessels
were carefully investigated in EPR.7–10 It is certain that
DDSs are categorized into various approaches of drug tar-
geting, which include passive targeting, active targeting,
and physical targeting.7–10 It is known that the potential
applications of nanotechnology and nanomaterials (in the
range of 100 nm) for the treatment, diagnosis, monitoring,
and control of animal and human biological systems are
referred to as nanomedicine.11–18 The common trials in
animals and humans are aimed at assessing a hoped low
toxicity along with a high efficacy. Briefly, the important
roles of size, composition, morphology, and structure of
nanomaterials and drugs are very meaningful in DDSs to
treat the diseases.4For instance in Japan, nanomedicine is
developed with many projects by the Ministry of Health,
Labour and Welfare (MHLW).11 So far, a nanodrug using
curcumin has been commercialized with high quality prod-
ucts used in treatment of cancerous diseases for skin and
face of humans.17
In our critical review, we have mainly presented the
latest results and highly exciting research aspects of
inorganic/organic particle systems prepared by chemi-
cal and physical methods with potential applications in
biology and medicine (traditional biomedicine) as well
as nanomedicine (nanotechnology). The inorganic/organic
particle systems are well combined with bioconjugates
with potential biomedical applications for drug delivery,
therapeutic hyperthermia, and magnetic resonance imaging
(MRI) in the important assays of the treatment of cancers
with the utilization of functional magnetic nanoparticles
from the latest research of scientists in the world. To this
end, nanomedicine technology must be further studied in
health and safety of humans through the clinic assays for
issues of toxicity and risks to our society, and its appli-
cations for humans should be done with low cost before
complete comerciallization.
2. NANOPARTICLES
To begin these interestingly practical topics of biomed-
ical applications,1–3 we have known that the term can-
cer includes a group of diseases that spring from by
abnormal changes in reproduction, growth and function
of cells in humans. The normal cells become abnormal
after a number of DNA mutations and they uncontrol-
lably proliferate. They can invade near tissues by local
invasion or by long-distance invasion (metastasis) via lym-
phatic system or blood vessels. Metastasis is one main
cause of death. The treatment methods of cancers are
to detect, prevent, and block their processes of devel-
opment at various stages, especially at the early and
beginning periods. In biomedical applications of inor-
ganic and organic particles, DDSs play extremely impor-
tant roles to carry and protect anti-cancer or anti-disease
drugs, biomolecules, and other targeted pharmaceuticals
according to treatment time.1–36They are reliably formed
by the stable bioconjugated particles for treating can-
cers. Both the functional bio-conjugated microsystems
and nanosystems can be potentially used for treatment of
tumors and other serious diseases, including cardiovas-
cular disease, rheumatic disease, gastrointestinal/hepatic
disease, infections such as bacterial, viral, parasitic, and
implant infections, tuberculosis, inflammation, diseases
of skin, face, eye, ear, brain, muscle, lung, kidney, and
liver etc. In DDSs approaches, abovementioned particles
are the main cores for biomedical multifunctional sys-
tems, such as microsystems, nanosystems, and micronano
systems with main features of targeting, drug delivery,
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10095
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
(A)
(B)
Uniform magnetic nanosystem
CoFe2O4-MnFe2O4
Tumor growth
and its volum
e
are controlled.
Figure 3. (A) TEM and high-resolution TEM images of 15 nm CoFe2O4–MnFe2O4nanoparticles. (B) In vivo hyperthermia treatment in the case of
diseased mouse with tumors. Reprinted permission from [151], (a) J. Lee, et al., Nat. Nanotechnol. 6, 418 (2011). © 2011. All Rights Reserved.
and treatment of diseases etc. exhibiting high selectiv-
ity for treating malignances that are found in animals
and humans that originate from themselves and from
living and surrounding environments.12–19 Such surface
functionalization of nanoparticles with bioconjugates can
lead to design various useful platforms for biomedical
applications. Multifunctional nanosystems lead to reduce
the dose of drugs, to avoid side effects, to target useful
anti-cancer drugs towards the diseased locations and to
avoid side effects in healthy cells and tissues. The main
features of particles (nanoparticles and microparticles) are
the sizes, the shapes, and the morphologies. They should
be homogeneous in terms of structure, composition, size,
and shape for important biomedical applications. Typically,
metal nanoparticles with bioconjugates in the nanosized
range of 10 nm are adopted in a platform to carry various
anticancer drugs to the local diseased regions.1–3
In many cases of biomedical applications, it is well
known that Au nanoparticles are demonstrated for molec-
ular imaging, drug delivery, in vivo cancer diagnosis
and therapy. The functional group of amino (–NH2and
thiol (–SH) groups can be used for bioconjugation to
Au nanoparticles (Fig. 2). At the nanosized range, Au
nanoparticles have high atomic density, and high surface
10096 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
area. Therefore, these Au nanoparticles can absorb radi-
ation much more than tissue cells, thus resulting in a
better local radiation-driven treatment of tumors. There-
fore, Au nanoparticles are used in medical applications
with various large advantages and benefits. At present,
common and rare diseases are typically classified as car-
diovascular diseases, malignant/benign tumors, infectious
diseases, proliferative and inflammatory disorders, syn-
drome diseases etc.612–15 To treat some cases of cancers,
Au nanoparticles can be directly injected intravenously.
It is generally accepted that they are accumulated in the
diseased areas, where there are the blood vessels that
can carry them, and functional Au nanoparticles reach
tumor areas efficiently. In this field, Pt nanoparticles were
not much considered in in vivo cancer diagnosis and
therapy. On the other hand, Ag nanoparticles were pri-
marily used for preventing bacteria, against infectious
diseases. Moreover, Fe-based nanoparticles, e.g., Fe3O4
(Fig. 2(A)), -Fe2O3,FeS
2,Fe
3S4,Fe
3O4–SiO2, and oth-
ers have excellent biomedical applications in magnetic
hyperthermia and magnetic resonance imaging (MRI).20–23
Figure 1 shows the various types of important functional
particles, such as noble metal nanoparticles, dendrimers,
e.g., poly(amido)amine (PAMANs), ligands, liposomes,
polymeric micelles for studying normal and tumor cells,6b
mesoporous SiO2nanoparticles for treating breast can-
cer in vitro and in vivo,6c polymeric nanoparticles, and
quantum dots (QDs).6To enhance delivery therapy to the
local diseased areas, QDs are commonly used for can-
cer imaging and treatment with high contrast. To treat
diseases efficiently, artificial virus-based nanoparticles are
also designed in testing and treatment of effects of various
viruses, such as HIV-1.6d In addition, various other parti-
cles are also used as magnetic nanoparticles (Co, Ni, and
Fe-based nanoparticles as well as their alloy and core–shell
nanostructures), carbon nanomaterials, and virus-based
nanoparticles. Their typical applications will be very cru-
cial in a near future for clinical trials in humans. Recently,
innovative drug delivery, diagnosis, treatment, and ther-
apy of diseases with biomedical applications through DDS
micro, nano, and/or micronano systems have been com-
prehensively investigated and developed.19–23
To date, cancer is one of the most common diseases of
human society, which derives from an uncontrolled growth
of a single cell. In particular, cancer is caused by mutations
in DNA that lead to the unlimited growth of cells, which
often causes mortality in patients.
To treat some aggressive cancers, scientists try to
open up a new therapeutic approach that turns traditional
medicines based on natural herbs (curcumin, pomegranate,
green tea etc.) combined with clinical approach methods,
and based on the key foundation of modern nanomedicine
through nanotechnology,2425170b In particular, curcumin
is considered as one of the most powerful natural drugs
against many kinds of various cancers without side effects.
At the same time, curcumin seems to enhance specific
receptors of tumor cells towards some targeting-anticancer
drugs, and reduces the high toxicity of chemotherapy,
and radiation therapy for treating invasive cancers. Most
interestingly, curcumin has the high ability to fight breast
cancer, lung cancer, brain tumor, leukemia, liver, intes-
tine, esophagus, colon cancer, melanoma, and gynecologi-
cal cancers etc. Based on preclinical testing and scientific
research associated with clinical trials, the curcumin com-
bined with nanosystems (typically chitosan nanoparticles,
Au nanoparticles for detecting biological molecules, sil-
ica nanoparticles and various biofunctional engineered
nanosystems26) is very successfully and widely used as
DDSs. However, for biomedical applications, it has to
be emphasized that efficiently synergistic collaborations
between researchers and scientists in many various areas
are indispensable to develop useful biomedical and diag-
nostic solutions by nanotechnology and traditional phar-
maceutics. In principle, biomedical applications through
nanomaterials and nanotechnology need to adopt particle
systems in the size range of 1–100 nm being the most
useful advantage at clinical level.
2.1. Metal Nanoparticles
So far, noble metal nanoparticles, typically such as Au,
Ag, Cu, and Pt nanoparticles (in fact Pt complexes to
be preferred) etc. have been widely investigated in the
biomedical trials in the treatments of the known dis-
eases of animal and human. Metal nanoparticles with
the same sizes and morphologies generally lead uni-
form nanosystem as a platform for biomedical applica-
tions. At present, noble nanoparticles, e.g., Au, Ag, Pt
and their specific compounds show excellent applications
in nano-biomedicine, especially for intrinsic therapeutic
applications of noble metal nanoparticles.2627 Importantly,
Au nanoparticle has become one of the most practical
nanoparticles for nanomedicine. It can be used as thera-
peutic element, and drug delivery in a process of treating
cancer disease. Additionally, Au nanoparticles are chemi-
cally facilely synthesized with uniform characterization of
the size and the shape. In many reports, Au nanoparti-
cles have been investigated in anti-cancer nanotechnology.6
In recent years, the scientists have proved that the injec-
tion of Au nanoparticles for treating cancers selectively
influences tumor cells. Most importantly, the other cells in
the healthy normal tissues are not affected or they have
very small side effects by Au nanoparticles. In the assays,
Au nanoparticles exhibited exciting phenomena in Surface
Plasmon Resonance (SPR), and Surface-Enhanced Raman
(SER) bands for biomedical applications for detection of
biomolecules or biomarkers.6e
For biomedical application, silver (Ag) nanoparticle is
also of great importance. Most typically, the efficiently
practical ability and role of Ag nanoparticles are proved
for preventing, protecting, and killing various danger-
ous and harmful bacteria appearing in animal, human,
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10097
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
and agriculture. In this aspect, the unique capabilities of
Ag nanoparticles can be considered as “nano bullets” to
kill bacteria in clinical medicine and biology.28 Like Au
nanoparticles, at a nanosized range, antivirus activity of
Ag nanoparticles increases much higher than that of ion
Ag in solution. Notably, Ag nanoparticles significantly kill
most of fungal infections, harmful bacteria and viruses.
In particular, bacteria’s immunity cannot be developed in
respective to Ag nanoparticles’s effect. It is known that
Ag nanoparticles are considered to be non-toxic, non-
allergic, non-accumulation inside the body of human, and
they are also known to be friendly, and not harmfully to
the surrounding environments. Similar to Ag nanoparticles,
copper (Cu) nanoparticles with their efficiently powerful
antimicrobial characteristics are discussed,29 but Cu and
CuO possibly show a very high toxicity to health of human
at the high degree of doses and contents. Based on our crit-
ical review, the development of nanomedicine with the use
of nanomaterials and nanoparticles are very fast to ther-
apeutic applications.29–31 Recently, soluble Pt(II) species
have been efficiently used for treating cancers,3233 exploit-
ing their cellular uptake and DNA platination at nontoxic
dosages. In addition to biomedical applications, Pt and
Pt–Pd bimetal nanoparticles have potential applications of
catalyst materials for fuel cells. The main reasons are that
their synthesis processes have been facile and efficient by
chemical and physical methods,243435a and discussed in
detail.35b Thus, bimetal nanoparticles belong to a group
of bimetallic nanosystem with potential applications for
nanomedicine.
2.2. Magnetic Nanoparticles
In general, magnetic nanoparticles with various great
advantages because of unique magnetism show the most
practical and important applications in biomedicine.22ab
In the biomedical applications, magnetic nanoparticles are
mainly used in drug delivery, MRI, and contrast agents
in the efficient treatment of cancers. The most typical
Fe3O4,Fe
2O3,Fe
xOy, and Fe-based oxides are consid-
erably studied in meaning to be “magnetic carriers.2236
Similarly, their ferrites, MFe2O4nanoparticles (M =Cu,
Ni, Co, Mn, Zn, and Mg) are also investigated for the
most interesting biomedical applications by significant
enhancement of high-contrast agents,36a typically such as
Ni1xZnxFe2O4nanoparticles,36b and superparamagnetic
Fe oxide nanoparticles.40 Additionally, Co, Ni, Fe-based
nanoparticles can be considered as magnetic nanovec-
tors for drug delivery.36–42 These above results lead that
Co, Ni, and Fe-based bimetal nanoparticles were stud-
ied for the assays of biomedical applications in ani-
mal and human,36–40 especially MFe2O4nanoparticles,41
Fe–Pt nanoparticles with ferromagnetic properties.42 There
is much progress in applications of magnetic nanopar-
ticles in biomedicine in both theoretical and practical
aspects.4344 The onset of ferromagnetism at the nanoscale
was meaningfully discussed in detail in the cases of Pd
and Pt nanoparticles. To increase the stability and dura-
bility, the thin Au or SiO2coatings can be produced on
the surfaces of FePt and Fe3O4nanoparticles to form
novel core–shell nanosystems.44 They can potentially be
useful in biomedical applications with bioconjugates still
under further the investigations. At present, oxide tran-
sition nanoparticles are of interest to researchers. Metal
transition oxide nanoparticles, e.g., Co-based, Ni-based,
and Fe-based nanoparticles lead to be a group of magnetic
oxide nanosystems.45 In addition to the simple synthesis
and preparation methods, they are promising candidates
for MRI and biomedical applications,45–48 especially the
type of superparamagnetic nanoparticles for new drug
delivery systems.47 However, the biocompatibility and tox-
icity of nanoparticles and pharmaceuticals still need to be
further investigated in the long-term and short-term clini-
cal assays in animals and humans in terms of time, con-
tent and dose of pharmaceuticals.4950 Recently, controlled
MnFe2O4nanoparticles and Gd complex-based nanocom-
posites were used as tunable and enhanced T1/T2-
weighted MRI contrast agents.51a For that purpose in MRI,
superparamagnetic CoFe2O4nanoparticles (6 nm) were
facilely synthesized in oleic acid-water-pentanol system at
180 C.51b According to the recent results, superparamag-
netic Fe3O4nanoparticles show a variety of potential appli-
cations of MRI, cell separation, hyperthermia, and drug
delivery in biomedicine and nanomedicine because they do
not exhibit the permanent magnetism in the case of lack
of external magnetic field.
2.3. Alloy and Core–Shell Nanoparticles
Recently, novel Fe-based, Co-based, and Ni-based bimetal-
lic alloys and core/shell nanoparticles have extensively
discovered to be the best candidates, particularly empha-
sizing on the advantages of high stability and dura-
bility in biomedical applications.51–59 According to the
latest research results, currently -Fe2O3oxide, Fe–Pd,
Co–Pt, Fe–Pt (Fig. 2), and Fe–Pt–Pd, and complex
FePt–CdSe core–shell nanoparticles have been used for
medical applications.5455 In this context, there is no
research on Pt–Pd nanoparticles for treating cancers that
has been reported. On the contrary, the nanoparticles
were shown to be one of the most advantageous tools
in catalysis and fuel cells. Among Fe-based nanoparti-
cles, Fe–Pt nanoparticles can be very potentially used
for biomedical applications, such as MRI contrast agent
because of high bioadaptability, durability and stability but
some low toxicity. According to one research, dumbbell-
like PtPd–Fe3O4nanoparticles can be used for catalysis
and associated biomedical applications,56 Fe3O4–SiO2-Au
core–shell nanosystems,57 and other bimetal nanoparticles,
Au–Ag, Ag–Au core–shell bimetallic nanoparticles for
SPRs and SERs applications for detection of biomolecules
or biomedical molecules,58aband Fe3O4–SiO2(Fig. 2(B)),
10098 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
oxide-based inorganic/organic and nanoporous spherical
particles,59 respectively.
3. SYNTHESIS
In traditional technology and modern nanotechnology,
chemical and physical synthesis methods or many com-
bined methods have been very successfully proved for
the facile preparation of metal, oxide and complex
nanoparticles.5859 At present, both bottom-up and top-
down technological approaches have been well applied
for producing nanoparticles with controllable size, shape,
morphology, and composition. The main differences
among the above preparation methods are just to be
the homogeneous or inhomogeneous characterization of
size, shape, composition, and structure of the prepared
nanoparticles. Typically, the successful preparation meth-
ods of shaping and controlling nanoparticles included co-
precipitation, thermal decomposition, reduction, micelle
synthesis, hydrothermal synthesis, sonoelectrodeposition,
ultrasonication, etc.5860 Sanchez and co-workers have dis-
cussed common synthetic routes of metal Fe-, Co-, and
Ni-P nanoparticles with the characteristics of their phase
diagrams.61 In this context, Cu-, Fe-, Ni-based nanoparti-
cles necessarily allows new biomedical applications. Gold
(Au) nanoparticles (100 nm) were prepared by modified
polyol methods.62 At present, Rh, Au, Pt, Pd, and Pt–Au
nanoparticles and their compounds are successfully syn-
thesized by polyol methods.63–71 The various sizes and
shapes of Pt nanoparticles were controlled in many size
ranges of 10 nm, 20 nm, and 30 nm in some our works
(Fig. 2). To date, Fe-based metal and bimetal nanoparticle
nanosystems are the most important products that are used
for biomedical applications.72–81 They can be produced by
various methods, such as polyol reduction, precipitation,
thermal decomposition, microemulsion, hydrothermal syn-
thesis under high pressure, sonochemical synthesis etc.
Significantly, Fe oxides possess a variety of specific
crystal structural phases, such as -Fe2O3,-Fe2O3,
-Fe2O3,Fe
3O4,etc.,
72–76 resulting in their various
biomedical applications. At present, scientists have just
very successfully used the pure Fe metal and Fe oxide
particles for biomedical applications. On the other hands,
bimetallic Fe nanoparticles are also promising candidates
towards scientific innovations in biology and medicine.77
In particular, the large -Fe2O3oxide particles were suc-
cessfully produced by polyol methods, drying and heat
treatment in air.78–80 In spite of large size, it is sug-
gested that the as-prepared -Fe2O3oxide microparticles
(microsystems) with the large size of 1–10 m (Fig. 1)
exhibited a superparamagnetic nature as the same as that of
Fe sulfide and Fe oxide nanoparticles (nanosystems) with
very small particle size.78 They can be used as potential
candidates for biomedical applications, such as treatment,
imaging, and therapy of tumors.
4. BIOMEDICAL APPLICATION
According to the recent practical applications, current par-
ticle systems for biomedicine can be classified in the dif-
ferent categories in respective to their particle size, such as
macroparticles (50–200 m), microparticles (1–100 m),
and nanoparticles (1–10 nm, 1–100 nm, even in a wide
range of 1–1000 nm). In addition, the definitions and
concepts of nanosized and microsized ranges can be
overlapped. A lot of special intention of scientists was
paid towards biofunctional nanosysems with the size of
nanoparticles around 10 nm and 30 nm,8283 although
their clearance of small nanoparticles out of human organ
and body is slower than that of large particles. In recent
works, only some reports have been given in accordance
with functional microparticles and their advantages with
biomedical applications about biology and medicine. This
is an open direction in the next studies of scientists. At
present, engineered nanoparticles combined with linkers
and bio-conjugates are being very necessarily tested in
biology and medicine or nanomedicine. They are noble
metal nanoparticles, such as Au, Ag, and Pt nanoparticles.
In fact, Au nanoparticles have very wide applications for
biosensor, molecular imaging, drug delivery, in vivo can-
cer diagnosis, and therapy. Comparing with Au nanopar-
ticles, Ag nanoparticles can be provided for SERs tests
in biosensing, and biomedical and chemical applications,
especially with the good antimicrobial effects. In addi-
tion to noble metal nanoparticles, dendrimers are also used
for biomedical applications, drug carriers, imaging agents,
gene delivery. Liposomes are widely studied for biomed-
ical applications in drug delivery, and gene encoding.
Another nanosystem, polymeric micelles, is used for drug
delivery (water-insoluble drugs) and nutraceutical deliv-
ery systems. In the biomedical applications, silica (SiO2
nanoparticles with advantage of more stability and dura-
bility than polymeric nanoparticles are widely used for
drug delivery.284 Among engineered nanosystems, poly-
meric nanoparticles are widely used for controlled release
of drug for cancer treatment, and applications for biosen-
sor, drug delivery, immunoassay, cell separation, protein
delivery, gene expression vector. A nanosystem, e.g., quan-
tum dots (QD) can be used as a platform for drug delivery,
diagnostics, medical imaging, and other biomedical appli-
cations. CdSe QDs can be used in drug delivery, diagnos-
tics and nanomedicine to enhance the efficacy of treating
the diseases of animal and human,285 for example QDs
versus organic dyes as fluorescent labels in photodynamic
therapy.8586 In recent years, the scientists have tried to
use carbon nanomaterials for biomedical applications.87
To prevent and kill dangerous diseases resulted from var-
ious viruses, and specific virus-based nanoparticles are
developed for testing and treatment of various viruses.
With the most advantages of drug delivery, MRI, high-
contrast agent, Co-, Ni-, and Fe-based magnetic nanopar-
ticles have been investigated in the biomedical assays.8788
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10099
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
In nanomedicine, the useful methods of surface function-
alisation of nanoparticles were proposed for biomedical
applications in many significant works.8990 In various sig-
nificant works, scientists proved that Au nanoparticles can
be potentially used for biosensor, in molecular imaging,
especially for drug delivery therapy, and the assays in vivo
cancer diagnosis and therapy, and other applications.91–100
Alternatively, Ag nanoparticles have potential biomedical
applications of preventing and killing infectious diseases,
such as harmful bacteria and viruses.101102 It is shown that
Ag and its compounds typically show a clear strong tox-
icity to many various bacteria, viruses, algae and fungi.
However, Ag and its compounds do not significantly show
toxicity to humans and if they are, that toxicity is very low.
It is certain that Ag nanoparticles proved typical biological
properties of anti-bacteria, and anti-inflammatory in their
growth and development of bacteria.101102 In addition, Ag
nanoparticles show very good drug delivery, and SERS
measurements for detection dyes in diagnosis and imag-
ing. An in vivo human time-exposure study of orally dosed
commercial Ag nanoparticles was presented. The authors
exactly demonstrated that 14-day monitored human oral
dosing of a commercial oral Ag nanoparticle product does
not produce observable clinical toxicity markers.103 There-
fore, fundamental parameters of toxicity thresholds and
levels of biomedical nanoparticles are used in human that
need to be further studied according to time, dose and
content, and the trials need to be studied in additional
organ systems of human. So far, Ag nanoparticles have
proved very high ability to prevent, block, destroy, and
kill bacterial infections in the assays,104 such as influenza
(A/H5N1), diarrhea (Escherichia coli), and cholera (Vibrio
cholera) but Ag nanoparticles have shown their possible
toxicology at high contents in biomedical applications.
Recently, functionally designed nanoparticles have been
discussed in medicine, biology, and nanomedicine (Fig. 1),
as well as fuel cells,6105–107 and typically for biosens-
ing applications with Pt nanowires.108109 In the out-
standing biomedical applications, Pt nanoparticles show
great potential for treating various cancers. In this case,
the scholars revealed that a fast ion radiation with Pt
nanoparticles improves future cancer therapy protocols.32a
Beyond Pt complexes and Pt nanoparticles, Au com-
plexes can probably be most useful as good anticancer
agents.32 Interestingly, a strategy was proposed on a com-
bination of Pt nanoparticles with irradiation by fast ions
used in “hadron therapy” towards lethal damage in DNA,
and other biomedical applications via Pt nanoparticles.110
Beside the above engineered nanoparticles, polymeric
nanoparticles in DDSs also show very good biomedical
applications in controlled release of drug, cancer treatment,
biosensors, drug delivery, immunoassay, cell separation,
protein delivery, and gene expression vector.111–116 Poly-
meric particles with high bio-adaptability can be obtained
from natural polymers or chemically-synthesized polymers
while magnetic nanoparticles with facile chemical synthe-
sis and preparation processes have very special biomedical
applications in vitro and in vivo analysis to animal and
human,117–127 such as Fe3O4nanoparticles and SiO2–Fe3O4
core–shell nanoparticles.117 In the former applications, Fe-
based nanoparticles are typically used for cell separation
and purification. In the latter applications, they can be
potentially used for targeted drug delivery, high-contrast
magnetic resonance imaging (MRI), and hyperthermia to
cancer treatment. Apart from Fe and Fe2O3oxide par-
ticles with micro- and nano-structures, other magnetic
nanoparticles associated with surfactants and polymer and
biofunctionalisation with biomolecules also offer good
biomedicine applications,121 specially emphasizing on Fe–
Pt, Fe3O4,CoFe
2O4,Fe
3O4–Au, Co–SiO2,Fe
3O4–SiO2,
Fe2O3–CdSe, and other Fe-based particles with core–
shell structures. The core–shell structures greatly show
advantages of biomedical applications in nanomedicine.
Ultra-large-scale syntheses of nanoparticles were presented
(Fig. 2(A)).
In comparison of the magnetic, radiolabeling, and
hyperthermic properties as well as biodistribution proper-
ties of hybrid nanoparticles bearing CoFe2O4and Fe3O4
metal or metal oxide cores,128 scholars proved that metal
cores can have a significant influence on the hyperthermic
properties of overall nanoformulation, and radiolabeling.
However, the properties of in vivo biodistribution remain
unaffected. Both the shell structure and the size seem to
have the significant influence on the in vivo fate of hybrid
magnetic nanoparticles. Among designed nanoparticles,
Fe-based superparamagnetic nanoparticles and their ferrite
compounds are abundant, cheap, and easy to be synthe-
sized for making various multifunctional particle systems
with bioconjugates. Recently, Franchini and his colleagues
have presented the assays of magnetic nanoparticles for
biomedical applications.128 The development of stable
aqueous suspensions of PEGylated superparamagnetic iron
oxide nanoparticles (SPIONs) has been intensively stud-
ied for those biomedical applications.129abSPIONs in MRI
technology enable important development in clinical imag-
ing. Beside nanoparticles discussed, recently we have suc-
cessfully synthesized TiO2nanorods.130 In this context,
TiO2is being a promising candidate to nanomedicine. Sci-
entists show that TiO2nanomaterials have good photo-
catalytic properties, good biocompatibility but they can
exhibit low toxicity in photodynamic therapy for cancer
treatment, drug delivery systems, and cell imaging.131
5. DRUG DELIVERY
At present, nanomedicine includes further studies of med-
ical diagnostics, nanotherapeutics, stem cells, and tissue
engineering.4In various biomedical applications, mag-
netic nanoparticles are efficiently surrounded by common
biodegradable polymers as carriers that can be lipo-
somes for drug delivery, erythrocytes for drug delivery
10100 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
and cell separation, phospholipids for enzyme immobiliza-
tion, albumin for drug delivery and cell separation, starch
for drug delivery, MRI, and irradiation, poly(lactic acid)
for irradiation.132 With the above biomedical applications,
dextran carriers can be very efficiently used with mag-
netic nanoparticles for cell separation, enzyme immobi-
lization, MRI, drug delivery, and therapeutic hyperthermia.
Besides, chitosan, carboxydextran, polyalkylcyanoacry-
late, and polyethyleneimine carriers can be used with
magnetic nanoparticles for drug delivery. Different from
biodegradable polymers, and other common polymers, typ-
ically such as ethyl-cellulose, specific synthetic polymers,
polystyrene, and polymethylmetacrylate can be used with
magnetic nanoparticles. The category of ethyl-cellulose
is used for penetrating into artery. Specific synthetic
polymers are used for cell separation and for treat-
ing and kill various dangerous viruses. For the biofunc-
tionalization of the surfaces of engineered nanoparticles,
various polymers can be used with magnetic nanoparti-
cles for biomedical applications. In general, it includes
polyethylene glycol, dextran, and polyvinylpyrrolidone
(PVP) for enhancing circulatory system. Fatty acids are
used for stabilizing suspension system, and providing car-
boxyl groups. A typical polymer, e.g., polyvinyl alcohol
(PVA) is used for homogeneous dispersion of nanosys-
tems. Moreover, polyacrylic acids are used for increasing
high bio-compatibility of functional magnetic nanoparti-
cles. In the clinic assays, polypeptides, phosphorylcholine,
poly(lactide-co-glycolide), poly(N-isopropylacryl amide),
chitosan, and gelatin can be used for biomedical appli-
cations with magnetic nanoparticles. They are used with
Au and magnetic nanoparticles, and various nanoparticles
for drug delivery,132–145 especially as CoFe2O4nanoparti-
cles for the treatment of cancers.140141 Recently, against
glioblastoma,142 targeted delivery methods of Ag nanopar-
ticles and alisertib were carried out in vitro and in vivo
synergistic effect. Of course, nanoparticles have the most
advantages over microparticles in drug delivery. The small-
est capillaries in the body are 5 to 6 m in diameter.142b
The size of particles being distributed into the bloodstream
must be significantly smaller than 5 m, and should not
form aggregates, and ensure that the particles do not form
an embolism.
6. HYPERTHERMIA
In very exciting magnetic hyperthermia or therapeutic
hyperthermia, most of scientists utilize Fe-based parti-
cles that are directly injected into the local regions of
malignancies and heated by a magnetic field in their
research. This method is useful to treat various tumors.
When a magnetic field is applied, the particles vibrate and
generate heat under external magnetic field, which in turn
destroys the tumor cells. At a high temperature, tumor
cells can be destroyed by magnetic hyperthermia at a wide
range of heating temperature 41 C–70 C.6The com-
pounds containing Fe metal or Fe oxide particles (nanopar-
ticles and microparticles) are very commonly used in
magnetic hyperthermia. Specifically, scientists suggest that
external magnetic field is safe and not much harmful to
the body and organs of human. Therefore, the magnetic
nanoparticles are crucially used in biomedical applica-
tions of diagnosis and treatment of various diseases. The
large tumors are usually treated by hyperthermia. In recent
exciting advances of tumor therapies, efficacy of hyper-
thermia requires that temperatures within tumors remain
above 43 C for 30 to 60 min, while safety considerations
limit the operating temperatures in normal tissues to below
42 C. In special cases, temperature range of 40–46 C can
be used for treating seriously large tumors. It is clear that
superparamgentic nanoparticles, such as Fe3O4and Fe2O3
are typically used for therapeutic hyperthermia.146–151 The
necessary clinical trials are used for mouse, rat, and rabbit
or other animals. In fact, Au nanoparticles are also used
for the similar purposes of hyperthermia through the com-
parison of Surface plasmon resonance (SPR).133 Recently,
Cheon and co-workers have performed exciting anti-tumor
study in mice test.151a In comparison with CoFe2O4and
MnFe2O4nanoparticles, they found the most typical mag-
netism of CoFe2O4–MnFe2O4nanoparticles at 300 K
with the persuasively hard evidences of superparamagnetic
nature and zero coercivity (Fig. 3). The attractive results
have been shown to demonstrate the very good potential
of magnetic nanoparticles for nanomedicine in the treat-
ment of invasive cancers of liver, brain, breast etc. (Fig. 4).
Thus, the Co1xZnxFe2O4nanoparticles can be prepared
for biomedical applications because of their specific mag-
netic properties.151 In addition, magnetic core–shell noble
metal/Fe-based oxide nanoparticles, such as Au–Fe2O3,
Au–Fe3O4, Au–CoFe2O4, etc. were used as MRI contrast
agents in cell culture with DNA-templated nanoparticle
chains.151b At the same time, they also discovered the
superior therapeutic efficacy of these nanoparticles to a
common anticancer drug. They have applied CoFe2O4
MnFe2O4hyperthermia with the size of magnetic Co, Mn
and Fe based nanoparticles in the homogeneous range of
about 20 nm. They have investigated dose dependency
on tumor volume (measured 18 days) after the treat-
ment and tumor growth was well controlled. The tumors
become much smaller comparable to before treatment by
hyperthermia. For magnetic core–shell nanoparticle hyper-
thermia and doxorubicin treatments, doses (75 gand
300 g) were needed to nearly and completely eliminate a
tumor with a volume of 100 mm3. For Feridex hyperther-
mia, even very high 1,200 g nanoparticle dose did not
adequately suppress and treat tumor growth. Thus, the high
and good efficacy of core–shell ferrite nanoparticle hyper-
thermia was clearly proved in biomedical assays in mice.
Accordingly, the large tumors are greatly reduced in both
volume and size. It is suggested that the local hyperthermia
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10101
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
Figure 4. Potential biomedical applications of magnetic nanoparticles.
will be most usefully combined with chemotherapy. How-
ever, in our opinion, the long-term treatment of some cases
of tumor diseases will be done with nanomedicine. There-
fore, practical nanodrugs and therapies need to be inex-
pensive when they can be widely applied for human use.
7. MAGNETIC RESONANCE IMAGING
Magnetic nanoparticles offer very practical applications in
magnetic resonance imaging (MRI). In MRI technology,
nuclear magnetic moment of protons is used as a sen-
sitive probe of the chemical neighborhood of protons in
different tissues and organs of the human body.6119149a
In principle, nuclear moments are aligned by means of
an external magnetic bias field 0.2–3 T and precession
of the spins is excited by transverse RF pulses at the
proton resonance frequency of about 43 MHz T1.149a
After applying the pulse sequence, the induced magne-
tization decays and the longitudinal (T1and transverse
(T2relaxation times of the processing nuclear magnetic
moments show tissue-specific differences that are used to
generate image contrast.149a Recently, Garwood and col-
leagues have proved quantification of Fe-oxide nanopar-
ticles at high concentration for thermal therapies.149b In
the latest outstanding research, the scientists show that
T
2mapping is difficult because of the very fast signal
decays in magnetism of magnetic nanoparticles. They dis-
covered that sweep imaging with Fourier transformation
(SWIFT) sequence is combined with the Look-Locker
method to map T1(Longitudinal/spin-lattice relaxation) of
Fe-oxide nanoparticles at high concentrations for thermal
therapies to overcome the current limitations of MRI. In
research efforts of scientists, Fe-oxide nanoparticles were
used with concentrations less than 53.6 mM. This is an
excellent finding, comparable to conventional methods of
T2(Transverse/spin–spin relaxation) or T
2mapping.149b
A new technique of the method of SWIFT MRI was also
applied for detection of breast cancer metastasis to the
lung with significant enhancement of MRI imaging.149c In
International workshop (2014) of MRI section, Professor
Garwood and scholars in his research group proved that
SWIFT method with the introduction of magnetic nanopar-
ticles into aqueous environments can be very clearly
visualized with positive contrast, and with a high Fe con-
centration up to at least 3 mg Fe/mL.149d In general,
imaging is well performed by controlling external field
gradients so that the resonance condition is fulfilled only
in a restricted local region, and then scanning the reso-
nant volume over the body part to be imaged. The tissue-
specific differences of the relaxation times T1and/or T2
or T
2can be used for construction of the so-called T1-
or T2-weighted images showing optimal contrast of tissue
features.149a However, most of the MRI treatments must
create a local impact mainly only on the tumor (diseased
region), and does not affect the different sections of differ-
ent cells and tissues of the healthy organs in the inside of
human. In ferromagnetic material, when the size of particle
is reduced into the nanometer-sized range, and at a cer-
tain critical value, their ferromagnetic properties are con-
verted into a typical superparamagnetic nature. However,
even Fe2O3oxide particles have the very large particle size
(1–10 m) with grain and grain boundary structures as the
10102 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
same size as the cells in human body.78 They are known
as Fe2O3oxide particulates. It is indicated that they also
showed an important superparamagnetic feature. There-
fore, large Fe2O3oxide particles can be very promising
candidates in nanomedicine for the treatment of very large
tumors, such large brain tumors. The characteristics of
superparamagnetic nanoparticles depend strongly on mag-
netic field but this magnetism disappears when external
magnetic field is not supplied. This is the key foundation
in potential biomedical application for clinical MRI tech-
niques because the superparamagnetic nanoparticles with
nanodrugs will be controlled under magnetic field. They
can accumulate mostly only in the area of cancer tis-
sues for curing them more efficiently. There are not much
works and reports relating to Fe-based microsystems for
biomedical applications.155–158 Although Fe oxides show
high bio-adaptability, and the best bio-adaptability with
bioconjugates to the inner organs of animal and human,
their potential toxicity remained that side effects can occur
in the organs.153154 The proposed methods of molecu-
lar dynamics simulations were used for studying interac-
tion between Fe3O4and biocompatible polymer.155 This
clearly proved that they are layers attached with index
planes of Fe3O4. Certainly, MRI contrast enhancement was
achieved by the designed spinel ferrite nanoparticles,156157
such as MnFe2O4,andCoFe
2O4. At present, scientists
are of great interest in the large brain tumors through
functional magnetic microsystems, and nanosytems, espe-
cially critical for nanotherapeutic approaches for brain can-
cer management.158 Potentially, SPIONs are meaningful to
therapy and treatment of various malignancies. In this very
urgent area, we really need to create novel kinds of innova-
tive magnetic nanocarriers for drug delivery, imaging, and
therapy against large brain tumors.5On these cases, SPI-
ONs or Fe-based nanoparticles with the polymeric coating
of poly(lactic-coglycolicacid)-block-poly(ethylene glycol)
(PLGA-b-PEG) can be efficiently used as a multifunc-
tional platform to carry and deliver imaging and thera-
peutic agents to the local diseased areas inside the body
of animal and human in the clinic assays. In this way,
targeted drug delivery with polymeric nanoparticles could
significantly improve the ability of diagnostic and/or ther-
apeutic agents to reach locally their target inside the brain.
To achieve the aims, the diagnostic agents can be used as
iron magnetic (Fe) based nanoparticles with size and shape
control to determine MRI information. Fe-based nanopar-
ticles will be covered by organic molecules to entrap
the Fe-based nanoparticles inside polymeric nanoparticles.
Therefore, -Fe2O3,Fe
3O4or -Fe2O3Au–Fe3O4,FePt,
Fe–Pt–Pd, Fe–Co, especially as MnFe2O4,andZnFe
2O4
based nanoparticles will be used for the ultimate goals of
MRI in theranostic applications because of the enhanced
high-contrast imaging agents.159–162 In addition, magnetic
MnFe2O4nanoparticles with 100 nm are prepared by
chemical method.163 Here, the MnFe2O4nanoparticle-
based immunoassays are carried out for rapid detection
of influenza infections by using an integrated microflu-
idic system. When compared with the 4.5 m magnetic
beads, the optical signals of the MnFe2O4nanoparticles
were twice as sensitive. So far, health safety-toxicity and
risk assessments of nanomaterials on animal and human
health as well as the surrounding environments have been
clearly identified.164165 Because of possible toxicity of
nanomaterials, several important biomedical applications
have been limited in the trials in human. In this con-
text, scientists are trying to find the solutions to address
these problems. Notably, the cytotoxicity of nanoparti-
cle encapsulated AsPt compositions on the human were
confirmed at around 37 C in the clinic assays (in vivo,
in vitro,orex vivo cells, tissues, and other organs inside
animal and human).166 In such circumstances, the inten-
sive investigations of nanomaterials-related risks and ben-
efits are being carried out in various kinds of nanoparticles
for use, safety recommendations, specifications and stan-
dards, regulation and policy in products etc.,167–173 espe-
cially critical for toxicity and safety in environment, life,
and health of animal and human. The toxicity and its dif-
ferent effects of magnetic nanoparticles on animal and
human depending on time must be confirmed in the short
or long periods or permanent existence. Tomitaka and
co-researchers presented an in vitro cytotoxicity test of
Fe3O4and NiFe2O4, which was performed using human
cervical carcinoma cells (HeLa).168 Fe3O4nanoparticles
have the size of 20–30 nm, and NiFe2O4nanoparticles
have the size of 20–30 nm. The in vitro cytotoxicity test
showed that exposure of the NiFe2O4nanoparticles to
HeLa cells causes a significant dose-dependent reduction
in the cell number as compared to Fe3O4nanoparticles.
Thanh and colleagues discussed the low and high toxicity
of nanosystems in detail with various doses and content
of nanoparticles and nanodrugs used for most of tumor
cases.5b4354558990124170a For detection and quantifica-
tion of anticancer drugs tagged to Fe3O4nanoparticles,
Huong and coauthors have very successfully presented the
use of Fe3O4nanoparticles (10 nm) with alginate and
curcumin.170b The ultra-high detection sensitivity of the
biosensor shows a value of 30% that was used for checking
magnetic biomarkers in biological systems. In our main
viewpoints, the safely suitable levels of a specific dose
and content of targeting drug and nanoparticles must be
controlled at the lowest toxicity. In fact, only the limit of
very low toxicity can be reliably accepted for the treatment
methods and deliver therapy of oncological patients.
8. CONCLUSIONS
In this review, we have primarily focused on biofunctional
engineered particle systems in relation to potential and
practical biomedical applications of diagnosis and therapy
of life-threatening and common diseases in animals and
humans, especially for tumor diseases. The bioconjuga-
tion methods and techniques are also shortly introduced to
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10103
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
design multifunctional inorganic/organic particle systems.
Here, the particle systems can be micro-, nano- and hybrid
micronano-systems with bioconjugates for nanomedicine.
The concrete characterizations of size and shape, dose and
content of functional nanoparticles should be classified in
relation to efficacy of treating many diseases. Although
they are very promising candidates for medicine and biol-
ogy, they show a certain degree of toxicity for animals
and humans. The future challenges of next research in tra-
ditional biology and medicine, and nanomedicine will be
focused on clarifying the toxicological aspects of nanopar-
ticles and nanomaterials in in vitro and in vivo trials before
they can be widely introduced for their clinical use.
Acknowledgments: We greatly thank and appreci-
ate the financial supports and projects sponsored by
Chinese Academy of Sciences (Visiting Fellowship for
Researchers from Developing Countries, Grant No.
2013FFGB0007) and China Postdoctoral Science Foun-
dation (No. 2014M551462) from Shanghai Institute of
Ceramics, Chinese Academy of Sciences, and other Uni-
versities for our research on Novel Magnetic Nanoparticles
for Catalysis, Biology and Medicine (Nanomedicine). This
study was supported by a fund from the National Natural
Science Foundation of China (No. 51471182).
References and Notes
1. R. A. J. Freitas, Nanomedicine 1, 2 (2005).
2. (a) R. A. Freitas, Jr., Nanomedicine, Volume IIA: Biocompatibility,
Landes Bioscience, Georgetown, Texas, U.S.A. (2003);(b)G.T.
Hermanson, Bioconjugate Techniques, Elsevier Inc. (2008).
3. T. C. Yih and C. Wei, Nanomedicine 1, 191 (2005).
4. H. Bullinger, Technology Guide: Principles—Applications—Trends
(Editor), Springer-Verlag, Berlin, Heidelberg (2009).
5. (a) G. A. Hughes, Nanomedicine 1, 22 (2005); (b) Y. Cheng, R. A.
Morshed, B. Auffinger, A. L. Tobias, and M. S. Lesniak, Adv. Drug
Deliv. Rev. 66, 42 (2014).
6. (a) N. V. Long, C. M. Thi, and M. Nogami, Curr. Phys. Chem. 4, 1
(2014); (b) G. Sahay, J. O. Kim, A. V. Kabanov, and T. K. Bronich,
Biomater. 31, 923 (2010); (c) H. Meng, W. X. Mai, H. Zhang,
M. Xue, T. Xia, S. Lin, X. Wang, Y. Zhao, Z. Ji, J. I. Zink, and
A. E. Nel, ACS Nano 7, 994 (2013); (d) X. Yu, A. Feizpour, N. P.
Ramirez, L. Wu, H. Akiyama, F. Xu, S. Gummuluru, and B. M.
Reinhard, Nat. Commun. 5, 4136 (2014); (e) P. M. Kosaka, V. Pini,
J. J. Ruz, R. A. da Silva, M. U. González, D. Ramos, M. Calleja,
and J. Tamayo, Nat. Nanotechnol. 9, 1047 (2014).
7. E. C. Unger, T. A. Fritz, T. Matsunaga, V. Ramaswami,
D. Yellowhair, and G. Wu, Therapeutic drug delivery systems. U.S.
Patent 5580575 (1996).
8. G. A. Hughes, Nanomedicine 1, 22 (2005).
9. H. Maeda, J. Wu, T. Sawa, Y. Matsumura, and K. Hori, J. Control.
Rel. 65, 271 (2000).
10. H. Maeda, Advan. Enzyme Regul. 41, 189 (2001).
11. http://www.mhlw.go.jp/english/.
12. R. Singh and H. S. Nalwa, J. Biomed. Nanotechnol. 7, 489 (2007).
13. M. Ferrari, Nat. Rev. Cancer 5, 161 (2005).
14. G. Taubes, Science 321, 356 (2008).
15. T. H. Hai, L. H. Phuc, L. K. Vinh, B. D. Long, T. T. Kieu, N. N.
Bich,T.N.Lan,N.Q.Hien,L.H.A.Khoa,andN.N.V.Tam,
Int. J. Nanotechnol. 8, 383 (2011).
16. C. Binns, Introduction to Nanoscience and Nanotechnoloy, John
Wiley & Sons, Inc. (2003).
17. (a)S.Prasad,A.K.Tyagi,andB.B.Aggarwal,Cancer. Res. Treat.
46, 2 (2014);(b)L.Pari,D.Tewas,andJ.Eckel,Arch Physiol.
Biochem. 114 (2008).
18. Z. Gao, L. Zhang, J. Hu, and Y. Sun, Nanomed. Nanotechnol. Biol.
Med. 9, 174 (2013).
19. I. Safarik and M. Safarikova, Chemical Papers 63, 497 (2009).
20. (a) T. Kim and T. Hyeon, Nanotechnology 25, 012001 (2014);
(b)A.S.BarnardandS.P.Russo,J. Phys. Chem. C 113, 5376
(2009).
21. G. Y. Tonga, K. Saha, and V. M. Rotello, Adv. Mater. 26, 359
(2014).
22. (a) D. Singh, J. M. McMillan, X. Liu, H. M. Vishwasrao,
A. V. Kabanov, M. Sokolsky-Papkov, and H. E. Gendelman,
Nanomedicine 9, 469 (2014); (b) X. P. Nguyen, D. L. Tran, P. T.
Ha,H.N.Pham,T.T.Mai,H.L.Pham,V.H.Le,H.M.Do,
T. B. H. Phan, T. H. G. Pham, D. T. Nguyen, T. M. N. Hoang,
K. Lam, and T. Q. Nguyen, Adv. Nat. Sci.: Nanosci. Nanotechnol.
3, 033001 (2012); (c) A. Singh and S. K. Sahoo, Drug Discovery
Today 19, 474 (2014).
23. M. Dwivedi, S. Sharma, P. Shukla, P. R. Mishra, S. K. Paliwal, and
A. K. S. Rawat, J. Biomater. Tissue Engin. 4, 198 (2014).
24. N. K. Kazem, A. Abolfazl, P. M. Mohammad, and S. J. Woo, Asian
Pac. J. Cancer. Prev. 14, 6595 (2013).
25. B. Korzeniowska, R. Nooney, D. Wencel, and C. McDonagh,
Nanotechnol. 24, 442002 (2013).
26. R. R. Arvizo, S. Bhattacharyya, R. A. Kudgus, K. Giri,
R. Bhattacharya, and P. Mukherjee, Chem. Soc. Rev. 41, 2943
(2012).
27. K. Fan, C. Cao, Y. Pan, D. Lu, D. Yang, J. Feng, L. Song, M. Liang,
and X. Yan, Nat. Nanotechnol. 7, 459 (2012).
28. K. Wong and X. Liu, Med. Chem. Commun. 1, 125 (2010).
29. R. Betancourt-Galindo, P. Y. Reyes-Rodriguez, B. A. Puente-
Urbina, C. A. Avila-Orta, O. S. Rodríguez-Fernández, G. Cadenas-
Pliego, R. H. Lira-Saldivar, and L. A. García-Cerda, J. Nanomater.
2014, 980545 (2014).
30. L. Zhang, F. X. Gu, J. M. Chan, A. Z. Wang, R. S. Langer, and
O. C. Farokhzad, Clin. Pharmacol. Ther. 83, 761 (2007).
31. M. K. Teli, S. Mutalik, and G. K. Rajanikant, Curr. Pharm. Des.
16, 1882 (2010).
32. (a)E.Porcel,S.Liehn,H.Remita,N.Usami,K.Kobayashi,
Y. Furusawa, C. L. Sech, and S. Lacombe, Nanotechnol. 21, 085103
(2010); (b) C. Nardon, G. Boscutti, and D. Fregona, Anticancer
Res. 34, 487 (2014); (c) F. P. Víctor, D. P. Fernando, R. M.
F. Manuel, and G. R. Óscar, Conjugates comprising nanopar-
ticles coated with platinum containing compouds. WO Patent
2010069941A1 (2010); (d) C. Nardon, G. Boscutti, and D. Fregona,
Anticancer Res. 34, 487 (2014).
33. H. Gehrke, J. Pelka, C. G. Hartinger, H. Blank, F. Bleimund,
R. Schneider, D. Gerthsen, S. Bräse, M. Crone, M. Türk, and
D. Marko, Arch. Toxicol. 85, 799 (2011).
34. N. V. Long, T. Asaka, T. Matsubara, and M. Nogami, Acta Mater.
59, 2901 (2011).
35. (a) N. V. Long, N. D. Chien, H. Hirata, T. Matsubara, M. Ohtaki,
and M. Nogami, J. Crys. Growth 320, 78 (2011); (b) A. D.
Banadaki and A. Kajbafvala, J. Nanomater. 2014, 985948 (2014).
36. (a) R. M. Cornell and U. Schwertmann, The Iron Oxides: Structure,
Properties, Reactions, Occourences and Uses, John Wiley & Sons,
Inc., Verlag GmbH, Co. KGaA, Weinheim (2003); (b) S. Singh,
Manpreet Singh, N. K. Ralhan, R. K. Kotnala, and K. C. Verma,
Adv. Sci. Eng. Med. 6, 688 (2014).
37. J. Klostergaard and C. E. Seeney, Nanomedicine 8, S37 (2012).
38. S. Sun and H. Zeng, J. Am. Chem. Soc. 124, 8204 (2002).
39. N. J. Darton, B. Hallmark, X. Han, S. Palit, N. K. H. Slater, and
M. R. Mackley, Nanomedicine 4, 19 (2008).
10104 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
40. E. M. Rajesh, K. Shamili, R. Rajendran, M. Elango, and S. R. M.
Shankar, Adv. Sci. Eng. Med. 6, 268 (2014).
41. S. Sun, H. Zeng, D. B. Robinson, S. Raoux, P. M. Rice, S. X.
Wang, and G. Li, J. Am. Chem. Soc. 126, 273 (2004).
42. S. Sun, C. B. Murray, D. Weller, L. Folks, and A. Moser, Science
287, 1989 (2000).
43. Q. A. Pankhurst, N. K. T. Thanh, S. K. Jones, and J. Dobson,
J. Phys. D: Appl. Phys. 42, 224001 (2009).
44. P. Crespo, P. de la Presa, P. Marín, M. Multigner, J. M. Alonso,
G. Rivero, F. Yndurain, J. M. González-Calbet, and A. Hernando,
J. Phys.: Condens. Matter 25, 484006 (2013).
45. A. Doaga, A. M. Cojocariu, C. P. Constantin, R. Hempelmann, and
O. F. Caltun, AIP Conference Proceedings 1564, 123 (2013)
46. A. K. Gupta and M. Gupta, Biomater. 26, 3995 (2005).
47. T. Neuberger, B. Schöpf, H. Hofmann, M. Hofmann, and B. von
Rechenberg, J. Magn. Magn. Mater. 293, 483 (2005).
48. M. Mahmoudi, H. Hofmann, B. Rothen-Rutishauser, and A. Petri-
Fink, Chem. Rev. 112, 2323 (2012).
49. H. Markides, M. Rotherham, and A. J. El Haj, J. Nanomaterials
2012, 614094 (2012).
50. M. Calero, L. Gutiérrez, G. Salas, Y. Luengo, A. Lázaro, P. Acedo,
M. P. Morales, R. Miranda, and A. Villanueva, Nanomedicine
10, 733 (2014).
51. (a) Z. Wang, J. Liu, T. Li, J. Liu, and B. Wang, J. Mater. Chem.
B2, 4748 (2014); (b) A. Repko, D. Niž ˇ
nanský, and J. Poltierová-
Vejpravová, J. Nanopart. Res. 13, 5021 (2011).
52. (a) K. Chatterjee, S. Sarkar, K. J. Rao, and S. Paria, Adv. Col-
loid Interface Sci. 209, 8 (2014); (b) L. Hung, J. Chang, Y. Tsai,
C. Huang, C. Chang, C. Yeh, and G. Lee, Nanomedicine 10, 819
(2014).
53. R. G. Chaudhuri and S. Paria, Chem. Rev. 112, 2373 (2012).
54. T. T. Trinh, D. Mott, N. T. K. Thanh, and S. Maenosono, RSC Adv.
1, 100 (2011).
55. D. Ung, L. D. Tung, G. Caruntu, D. Delapostas, Y. Alexandrous,
I. Prior, and N. T. K. Thanh, Cryst. Eng. Comm. 11, 1309 (2009).
56. X. Sun, S. Guo, Y. Liu, and S. Sun, Nano Lett. 12, 4859 (2012).
57. D. Ho, X. Sun, and S. Sun, Acc. Chem. Res. 44, 875 (2011).
58. (a) Y. Yang, J. Shi, G. Kawamura, and M. Nogami, Scripta Mater.
58, 862 (2008); (b) Y. Huang, Y. Yang, Z. Chen, X. Li, and
M. Nogami, J. Mater. Sci. 43, 5390 (2008).
59. K. Shiba, M. Tagaya, R. D. Tilley, and N. Hanagata, Sci. Technol.
Adv. Mater. 14, 023002 (2013).
60. P. Tartaj, M. D. P. Morales, S. Veintemillas-Verdaguer, T. González-
Carreño, and C. J. Serna, J. Phys. D: Appl. Phys. 36, R182 (2003).
61. S. Carenco, D. Portehault, C. Boissière, N. Mézailles, and
C. Sanchez, Adv. Mater. 26, 371 (2014).
62. N. V. Long, M. Ohtaki, M. Yuasa, S. Yoshida, T. Kuragaki, C. M.
Thi, and M. Nogami, J. Nanomater. 2013, 793125 (2013).
63. N. V. Long, C. M. Thi, M. Nogami, and M. Ohtaki, New J. Chem.
36, 1320 (2012).
64. N. V. Long, M. Ohtaki, T. D. Hien, R. Jalem, and M. Nogami,
J. Nanopart. Res. 13, 5177 (2011).
65. N. V. Long, T. D. Hien, T. Asaka, M. Ohtaki, and M. Nogami,
J. Alloys Compd. 509, 7702 (2011).
66. N. V. Long, N. D. Chien, H. Hirata, M. Ohtaki, T. Hayakawa,
and M. Nogami, Adv. Nat. Sci: Nanosci. Nanotechnol. 1, 035012
(2010).
67. (a) N. V. Long, C. M. Thi, Y. Yang, M. Nogami, and M. Ohtaki,
J. Nanosci. Nanotechnol. 13, 4799 (2013); (b) N. V. Long,
M. Nogami, C. M. Thi, and M. Ohtaki, J. Adv. Micros. Res. 7, 98
(2012).
68. N. V. Long, C. M. Thi, M. Nogami, and M. Ohtaki, Recent Patents
Mater. Sci. 5, 175 (2012).
69. N. V. Long, N. D. Chien, M. Uchida, T. Matsubara, J. Randy, and
N. Masayuki, Mater. Chem. Phys. 124, 1193 (2010).
70. N. V. Long, T. Hayakawa, T. Matsubara, N. D. Chien, M. Ohtaki,
and M. Nogami, J. Exp. Nanosci. 7, 426 (2012).
71. N. V. Long, N. D. Chien, T. Hayakawa, T. Matsubara, M. Ohtaki,
and M. Nogami, J. Exp. Nanosci. 7, 133 (2012).
72. A. S. Teja and P. Y. Koh, Cryst. Growth Character. Mater. 55, 22
(2009).
73. M. Colombo, S. Carregal-Romero, M. F. Casula, L. Gutiérrez, M. P.
Morales, I. P. Böhm, J. T. Heverhagen, D. Prosperi, and W. J. Parak,
Chem. Soc. Rev. 41, 4306 (2012).
74. A. Lu, E. L. Salabas, and F. Schüth, Angew. Chem. Int. Ed. 46, 1222
(2007).
75. D. L. Huber, Small 1, 482 (2005).
76. S. Laurent, D. Forge, M. Port, A. Roch, C. Robic, L. V. Elst, and
R. N. B. Muller, Chem. Rev. 108, 2064 (2008).
77. W. J. Liu, T. T. Qian, and H. Jiang, Chem. Eng. J. 236, 448 (2014).
78. N. V. Long, Y. Yang, C. M. Thi, Y. Cao, and M. Nogami, Colloids
Surf., A 456, 184 (2014).
79. N. V. Long, Y. Yang, M. Yuasa, C. M. Thi, Y. Cao, T. Nann, and
M. Nogami, RSC Adv. 4, 8250 (2014).
80. N. V. Long, Y. Yang, M. Yuasa, C. M. Thi, Y. Cao, T. Nann, and
M. Nogami, RSC Adv. 4, 6383 (2014).
81. N. Poudyal and J. P. Liu, J. Phys. D: Appl. Phys. 46, 043001 (2013).
82. B. Felice, M. P. Prabhakaran, A. P. Rodríguez, and S. Ramakrishna,
Mater. Sci. Eng., C C41, 178 (2014).
83. G. A. Hughes, Nanomedicine 1, 22 (2005).
84. W. Zhou, P. Gao, L. Shao, D. Caruntu, M. Yu, J. Chen, and C. J.
O’Connor, Nanomedicine 1, 233 (2005).
85. T. Nann, Nano. Biomed. Eng. 3, 137 (2011).
86. U. Resch-Genger, M. Grabolle, S. Cavaliere-Jaricot, R. Nitschke,
and T. Nann, Nat. Methods 5, 763 (2008).
87. C. S. S. R. Kumar, Biofunctionalization of Nanomaterials, Nano-
technologies for the Life Sciences, Wiley-VCH Velag GmbH &
Co., KgaA, Weinheim (2005),Vol.1.
88. C. Yang, A. Rait, K. F. Pirollo, J. A. Dagata, N. Farkas, and E. H.
Chang, Nanomedicine 4, 318 (2008).
89. N. T. K. Thanh and L. A. W. Green, Nano. Today 5, 213 (2010).
90. C. Blanco-Andujar, L. D. Tung, and N. T. K. Thanh, Annu. Rep.
Prog. Chem., Sect. A 106, 553 (2010).
91. V. Vergoni, G. Tosi, R. Tacchi, M. A. Vandelli, A. Bertolini, and
L. Costantino, Nanomedicine 5, 369 (2009).
92. P.K.Jain,I.H.El-Sayed,andM.A.El-Sayed,Nanotoday 2, 18
(2007).
93. X. Xing, Y. Chang, and I. Kang, Sci. Technol. Adv. Mater.
11, 014101 (2010).
94. P.C.Chen,S.C.Mwakwari,andA.K.Oyelere,Nanotechnol. Sci.
App. 1, 45 (2008).
95. E. Boisselier and D. Astruc, Chem. Soc. Rev. 38, 1759 (2009).
96. A. Kopwitthaya, K. Yong, R. Hu, I. Roy, H. Ding, L. A. Vathy,
E. J. Bergey, and P. N. Prasad, Nanotechnol. 21, 315101 (2010).
97. Y. Yang, M. Tanemura, Z. Huang, D. Jiang, Z. Li, Y. Huang,
G. Kawamura, K. Yamaguchi, and M. Nogami, Nanotechnol.
21, 325701 (2010).
98. J. Stone, S. Jackson, and D. Wright, WIREs Nanomed.
Nanobiotechnol. 3, 100 (2011).
99. P. K. Jain, K. S. Lee, I. H. El-Sayed, and M. A. El-Sayed, J. Phys.
Chem. B 110, 7238 (2006).
100. G. Kawamura, Y. Yang, and M. Nogami, Appl. Phys. Lett.
90, 261906 (2007).
101. M. M. Kemp, A. Kumar, S. Mousa, E. Dyskin, M. Yalcin,
P. Ajayan, R. J. Linhardt, and S. A. Mousa, Nanotechnol.
20, 455104 (2009).
102. J. S. Kim, E. Kuk, K. N. Yu, J. Kim, S. J. Park, H. J. Lee, S. H.
Kim, Y. K. Park, Y. H. Park, C. Hwang, Y. Kim, Y. Lee, D. H.
Jeong, and M. Cho, Nanomedicine 3, 95 (2007).
103. M. A. Munger, P. Radwanski, G. C. Hadlock, G. Stoddard,
A. Shaaban, J. Falconer, D. W. Grainger, and C. E. Deering-Rice,
Nanomedicine 10, 1 (2014).
104. Q. H. Tran, V. Q. Nguyen, and A. T. Le, Adv. Nat. Sci: Nanosci.
Nanotechnol. 4, 033001 (2013).
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10105
Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles Long et al.
105. Y. Cao, J. Zhang, Y. Yang, Z. Huang, N. V. Long, and M. Nogami,
J. Nanosci. Nanotechnol. 14, 1194 (2014).
106. N. Sanvicens and M. P. Marco, Trends Biotechnol. 26, 425 (2008).
107. G. Doria, J. Conde, B. Veigas, L. Giestas, C. Almeida,
M. Assunção, J. Rosa, and P. V. Baptista, Sensors 12, 1657 (2012).
108. P. T. H. Thuy, T. P. Duy, D. M. Chien, T. G. Bui, V. L. Nguyen,
and H. D. Tong, Int. J. Nanotechnol. 10, 166 (2013).
109. V. B. Pham , T. H. Le, X. T. T. Pham, V. V. To, N. T. D. Dan g, T. T.
T. Le, T. G. Bui, V. L. Nguyen, M. C. Dang, and D. H. Tong, Int.
J. Nanotechnol. 10, 178 (2013).
110. H. Mohammadi, A. Abedi, A. Akbarzadeh, M. J. Mokhtari, H. E.
Shahmabadi, M. R. Mehrabi, S. Javadian, and M. Chiani, Int. Nano
Lett. 3, 28 (2013).
111. R. Dinarvand, N. Sepehri, S. Manoochehri, H. Rouhani, and
F. Atyabi, Int. J. Nanomedicine 6, 877 (2011).
112. R. Singh and J. W. Lillard, Jr., Exp. Mol. Pathol. 86, 215 (2009).
113. C. P. Reis, R. J. Neufeld, A. J. Ribeiro, and F. Veiga, Nanomedicine
2, 8 (2006).
114. S. M. Moghimi, A. C. Hunter, and J. C. Murray, Pharm. Rev.
53, 283 (2001).
115. J. Panyam and V. Labhasetwar, Adv. Drug Delivery Rev. 55, 329
(2003).
116. K. Tahara, S. Furukawa, H. Yamamoto, and Y. Kawashima, Int. J.
Pharm. 392, 311 (2010).
117. (a)J.Park,K.An,Y.Hwang,J.Park,H.Noh,J.Kim,J.Park,
N. Hwang, and T. Hyeon, Nat. Mater. 3, 891 (2004); (b) C. Vogt,
M. S. Toprak, M. Muhammed, S. Laurent, J. Bridot, and R. N.
Müller, J. Nanopart. Res. 12, 1137 (2010).
118. L. H. Reddy, J. L. Arias, J. Nicolas, and P. Couvreur, Chem. Rev.
112, 5818 (2012).
119. W. Andrä, U. Häfeli, R. Hergt, and R. Misri, Application of Mag-
netic Particles in Medicine and Biology, Handbook of Magnetism
and Advanced Magnetic Materials. Edited by H. Kronmüller and
S. Parkin, Novel Materials, Wiley-Interscience (2007),Vol.4.
120. O. Ketebu, P. Cumpson, and E. Yu, Adv. Sci. Eng. Med. 6, 531
(2014).
121. J. Gao, H. Gu, and B. Xu, Acc. Chem. Res. 42, 1097 (2009).
122. N. V. Long, C. M. Thi, Y. Yong, Y. Cao, H. Wu, and M. Nogami,
Recent Patents Nanotechnol. 8, 52 (2014).
123. B. I. Kharisov, H. V. Rasika Dias, O. V. Kharissova, V. M. Jiménez-
Pérez, B. O. Pérez, and B. M. Flores, RSC Adv. 2, 9325 (2012).
124. N. T. K. Thanh, Magnetic Nanoparticles from Fabrication to Clin-
ical Applications Theory to Therapy Chemistry to Clinic Bench to
Bedside, CRC Press, Taylor & Francis Group, LLC (2012).
125. M.R.Dewi,W.M.Skinner,andT.Nann,Australian J. Chem.
67, 663 (2014).
126. H. Lida, K. Takayanagi, T. Nakanishi, and T. Osaka, J. Colloid
Interf. Sci. 314, 274 (2007).
127. B. Peeples,V. Goornavar, C. Peeples, D. Spence, V. Parker, C. Bell,
D. Biswal, G. T. Ramesh, and A. K. Pradhan, J. Nanopart. Res.
16, 2290 (2014).
128. D. Psimadas, G. Baldi, C. Ravagli, M. C. Franchini, E. Locatelli,
C. Innocenti, C. Sangregorio, and G. Loudos, Nanotechnol.
25, 025101 (2014).
129. (a) K. Hervé, L. Douziech-Eyrolles, E. Munnier, S. Cohen-
Jonathan, M. Soucé, H. Marchais, P. Limelette, F. Warmont, M. L.
Saboungi, P. Dubois, and I. Chourpa, Nanotechnol. 19, 465608
(2008); (b) A. Beduneau, Z. Ma, C. B. Grotepas, A. Kabanov, B. E.
Rabinow, N. Gong, R. L. Mosley, H. Dou, M. D. Boska, and H. E.
Gendelman, PLoS One 4, e4343 (2009).
130. N. V. Long, P. V. Viet, L. V. Hieu, C. M. Thi, Y. Yong, and
M. Nogami, J. Adv. Sci. Engin. Med. 6, 1 (2014).
131. Z. F. Yin, L. Wu, H. G. Yang, and Y. H. Su, Phys. Chem. Chem.
Phys. 15, 4844 (2013).
132. X. Zhou, A. L. Porter, D. K. R. Robinson, M. S. Shim, and Y. Guo,
Nanomedicine 10, 889 (2014).
133. L. Dykmana and N. Khlebtsov, Chem. Soc. Rev. 41, 2256 (2012).
134. J. Dobson, Drug Dev. Res. 67, 55 (2006).
135. M. Arruebo, R. Fernández-Pacheco, M. R. Ibarra, and
J. Santamaría, Nano Today 2, 22 (2007).
136. C. Sun, J. S. H. Lee, and M. Zhang, Adv. Drug Delivery Rev.
60, 1252 (2008).
137. R. A. Petros and J. M. DeSimone, Nat. Rev. Drug Discov. 9, 615
(2010).
138. D.L.Tran,V.H.Le,H.L.Pham,T.M.N.Hoang,T.Q.Nguyen,
T. T. Luong, P. T. Ha, and X. P. Nguyen, Adv. Nat. Sci.: Nanosci.
Nanotechnol. 1, 045013 (2010).
139. J. Chomoucka, J. Drbohlavova, D. Huska, V. Adam, R. Kizek, and
J. Hubaleka, Phar macol. Res. 62, 144 (2010).
140. G. Baldi, D. Bonacchi, M. C. Franchini, D. Gentili, G. Lorenzi,
A. Ricci, and C. Ravagli, Langmuir 23, 4026 (2007).
141. E. Locatelli, P. Matteini, F. Sasdelli, A. Pucci, M. Chiariello,
V. Molinari, R. Pini, and M. C. Franchini, Chem. Commun.
50, 7783 (2014).
142. (a) E. Locatelli, M. Naddaka, C. Uboldi, G. Loudos,
E. Fragogeorgi, V. Molinari, A. Pucci1, T. Tsotakos, D. Psimadas,
J. Ponti, and M. C. Franchini, Nanomedicine 9, 839 (2014);
(b) M. L. Hans and A. M. Lowman, Nanoparticles for Drug Deliv-
ery, Nanomaterials Handbook, Taylor and Francis Group, LLC
(2006).
143. E. Locatelli, W. Bost, M. Fournelle, J. Llop, F. Arena, V. Lorusso,
and M. C. Franchini, J. Nanopart. Res. 16, 2304 (2014).
144. Z. Poon, J. A. Lee, S. Huang, R. J. Prevost, and P. T. Hammond,
Nanomedicine 7, 201 (2011).
145. A. Sinha, A. Shaporev, N. Nosoudi, Y. Lei, A. Vertegel, S. Lessner,
and N. Vyavahare, Nanomedicine 10, 1003 (2014).
146. K. K. Jain, The Handbook of Nanomedicine. Humana Press,
Springer Science+Business Media, LLC (2008).
147. P. I. Soares, I. M. Ferreira, R. A. Igreja, C. M. Novo, and J. P.
Borges, Recent Pat. Anticancer Drug Discov. 7, 64 (2012).
148. Z. Zhou, L. Wang, X. Chi, J. Bao, L. Yang, W. Zhao, Z. Chen,
X. Wang, X. Chen, and J. Gao, ACS Nano 7, 3287 (2013).
149. (a) Q. A. Pankhurst, J. Connolly, S. K. Jones, and J. Dobson,
J. Phys. D: Appl. Phys. 36, R167 (2003); (b) J. Zhang,
R. Chamberlain, M. Etheridge, D. Idiyatullin, C. Corum,
J. Bischof, and M. Garwood, Magn. Reson. Med. 71, 1982 (2014);
(c) N. Kobayashi, D. Idiyatullin, C. Corum, J. Weber, M. Garwood,
and D. Sachdev, Magn. Reson. Med. 73, 1812 (2015);(d)M.Gar-
wood, J. Zhang, M. L. Etheridge, H. L. Ring, K. R. Hurley,
L. Utecht, A. Petryk, S. Jeon, D. Idiyatullin, C. Hogan, C. L.
Haynes, J. C. Bischof, and P. J. Hoopes, Advanced Magnetic Reso-
nance Imaging Approaches to Thermal Therapy and Nanomedicine
Biodistribution, Society for Thermal Medicine Annual Meeting
Minneapolis, Minnesota, United State (US) (2014).
150. E. Fantechi, C. Innocenti, M. Zanardelli, M. Fittipaldi, E. Falvo,
M. Carbo, V. Shullani, L. D. C. Mannelli, C. Ghelardini, A. M.
Ferretti, A. Ponti, C. Sangregorio, and P. Ceci, ACS Nano 8, 4705
(2014).
151. (a) J. Lee, J. Jang, J. Choi, S. H. Moon, S. Noh, J. Kim, J. Kim,
I. Kim, K. I. Park, and J. Cheon, Nat. Nanotechnol. 6, 418 (2011);
(b) H. Jaganathan, D. L. Hugar, and A. Ivanisevic, ACS Appl. Mater.
Interfaces 3, 1282 (2011).
152. D. P. Cormode, B. L. Sanchez-Gaytan, A. J. Mieszawska, Z. A.
Fayad, and W. J. M. Mulder, NMR Biomed. 26, 766 (2013).
153. G. Liu, J. Gao, H. Ai, and X. Chen, Small 9, 1533 (2013).
154. H. B. Na, I. C. Song, and T. Hyeon, Adv. Mater. 21, 2133
(2009).
155. L. Qiang, T. Yang, Z. Li, H. Wang, X. Chen, and X. Cui, Colloids
Surf., A 456, 62 (2014).
156. J. Lodhia, G. Mandarano, N. J. Ferris, P. Eu, and S. F. Cowell,
Biomed. Imaging Interv. J. 6, e12 (2010).
157. G. Mandarano, J. Lodhia, P. Eu, N. J. Ferris, R. Davidson, and S. F.
Cowell, Imaging Interv. J. 6, e13 (2010).
10106 J. Nanosci. Nanotechnol. 15, 10091–10107,2015
Long et al. Biomedical Applications of Advanced Multifunctional Magnetic Nanoparticles
158. L. S. del Burgo, R. M. Hernández, G. Orive, and J. L. Pedraz,
Nanomedicine 10, 905 (2014).
159. M. Ryvolova, J. Chomoucka, J. Drbohlavova, P. Kopel, P. Babula,
D. Hynek, V. Adam, T. Eckschlager, J. Hubalek, M. Stiborova,
J. Kaiser, and R. Kizek, Sensors 12, 14792 (2012).
160. Wahajuddin and S. Arora, Int. J. Nanomedicine 7, 3445
(2012).
161. T. J. Chen, T. H. Cheng, Y. C. Hung, K. T. Lin, G. C. Liu, and
Y. M. Wang, J. Biomed. Mater. Res. A 87, 165 (2008).
162. G. Invernici, S. Cristini, G. Alessandri, S. E. Navone, L. Canzi,
D. Tavian, C. Redaelli, F. Acerbi, and E. A. Parat, Recent Pat.
Anticancer Drug Discov. 6, 58 (2011).
163. L. Hung, J. Chang, Y. Tsaic, C. Huang, C. Chang, C. Yeh, and
G. Lee, Nanomedicine 10, 819 (2014).
164. H. Spielmann, T. Seidle, V. Kral, G. Schoeters, M. Schäfer-Korting,
A. Rowan, and E. McIvor, Alternative testing strategies: Progress
report 2010, Replacing, reducing and refining use of animals in
research. Progress report 2010-EU, Health Biotechnology, AXLR8
Consortium, European Commission (2010), ISBN 978-3-9814126-
1-1.
165. S. Singh and H. S. Nalwa, J. Nanosci. Nanotechnol. 7, 3048 (2007).
166. H. Chen, S. Pazicni, N. L. Krett, R. W. Ahn, J. E. Penner-Hahn,
S.T.Rosen,andT.V.OHalloran,Angew. Chem. Int. Ed. 48, 9295
(2009).
167. T. R. Daniels, T. Delgado, J. A. Rodriguez, G. Helguera, and M. L.
Penichet, Clin. Immunol. 121, 144 (2006).
168. A. Tomitaka, T. Koshi, S. Hatsugai, T. Yamada, and Y. Takemura,
J. Magn. Magn. Mater. 323, 1398 (2011).
169. S. Rizzitelli, P. Giustetto, C. Boffa, D. D. Castelli, J. C. Cutrin,
S. Aime, and E. Terreno, Nanomedicine 10, 901 (2014).
170. (a) L. Yildirimer, N. T. K. Thanh, M. Loizidoua, and A. M.
Seifalian, Nano Today 6, 585 (2011); (b) J. Devkota, J. Wingo, T. T.
T. Mai, X. P. Nguyen, N. T. Huong, P. Mukherjee, H. Srikanth, and
M. H. Phan, J. Appl. Phys. 115, 17B503 (2014).
171. J. T. Jenkins, D. L. Halaney, K. V. Sokolov, L. L. Ma, H. J. Shipley,
S. Mahajan, C. L. Louden, R. Asmis, T. E. Milner, K. P. Johnston,
and M. D. Feldman, Nanomed. Nanotechnol. Biol. Med. 9, 356
(2013).
172. Z. Gao, L. Zhang, J. Hu, and Y. Sun, Nanomed. Nanotechnol. Biol.
Med. 9, 174 (2003).
173. D. Singh, J. M. McMillan, A. V. Kabanov, M. Sokolsky-Papkov,
and H. E. Gendelman, Nanomedicine 9, 501 (2014).
Received: 9 July 2014. Revised/Accepted: 12 March 2015.
J. Nanosci. Nanotechnol. 15, 10091–10107, 2015 10107
... Despite the promising applications of Fe-NMPs, the impact of these nanoparticles (NPs) on the environment [29] and human health [30] is still a concern, as they may accumulate in soil and water resources and potentially cause toxicity to living organisms [31,32]. So far, the impact of Fe-based NPs on plants is differentiated, depending on the plant species, dose, exposure route, and NP size, whereas the studies with porphyrins in plants are nearly non-existent. ...
... Among the biomedical applications magnetite nanoparticles play a crucial role in the diagnosis and treatment of cancer, biocatalysis, analysis of pharmaceuticals, synthetic cell engineering, biosensors, and immobilization of biomolecules such as proteins [21,22]. Imperfectly stabilized Fe 3 O 4 nanoparticles are characterized by their high chemical activity and sensitivity to oxidation, often leading to a reduction or complete loss of magnetic properties. ...
Article
The presented article designs hydrogel structures with magnetite nanoparticles in the composition, involving various synthetic and natural polymers. The synthesis of magnetite nanoparticles in the hydrogel composition was accomplished through the co-precipitation method, and the immobilization of protein using trypsin as a model drug was investigated. The incorporated of micro-quantities of magnetite Fe3O4 nanoparticles into the gel matrix enhances the affinity for the enzyme, resulting in a significant improvement in specific activity and stability compared to gels without magnetite Fe3O4 nanoparticles. In other words, increasing the degree of immobilization of magnetite Fe3O4 nanoparticles enhances both the immobilization degree and the biological activity of the enzyme. This phenomenon manifests as a continuous interaction between the gel composition and the magnetite nanoparticles of the enzyme
... Such probes can reveal cellular or molecular lesions and increase treatment accuracy [2,3]. To be successful, these probes must (i) have significant detection capability, (ii) be able to assemble in the desired targeted area, and (iii) be biocompatible [4,5]. Diagnostic probes that use nanoparticles (NPs) as contrast agents can target tumors to enhance the detection of cancers [6], which afflict 10 million people annually according to the World Health Organization. ...
Article
Full-text available
Purpose: We synthesized folic acid-conjugated Fe3O4/Au-pralidoxime chloride Nanoparticles (Fe2O3/Au@PAM NPs) for use as dual-modal contrast agents for Magnetic Resonance Imaging (MRI) and Computed Tomography (CT) in the diagnosis of breast cancer. Materials and Methods: Fe2O3/Au@PAM NPs labeled or not to folic acid were synthesized and analyzed by dynamic light scattering, transmission electron microscopy, and vibrating sample magnetometry. The ability of these NPs to create image contrast was also investigated in silico and in vitro (in MCF-7 breast cancer cells and A549 lung cancer cells) with CT and MRI. Results: Dynamic light scattering and transmission electron microscopy revealed that the Fe2O3/Au@PAM NPs were nearly spherical. The average diameter of Fe2O3/Au NPs increased from 11.6 nm to 98 nm after folic acid conjugation. The saturation magnetization values of Fe2O3/Au@PAM NPs with and without folic acid conjugation were 25.56 and 32.6 emu/g, respectively. Conjugation of folic acid to NPs greatly improved their uptake by cancer cells. The additional coating of NPs with FA reduced the T2 relaxation time and signal intensity for MRI. Folic acid-labeled MCF-7 cells had a radiodensity measurement of 208 Hunsfield Units (HU) compared to 95 HU for A549 cells. For breast cancer cells, NPs labeled with folic acid significantly improved the X-ray absorption coefficient as a sign of active cellular uptake compared to NPs without labeling. Conclusion: Folic acid-labeled Fe2O3/Au@PAM NPs can serve as dual CT/MRI contrast agents and improve the sensitivities of both modalities for the detection of cancer cells.
... Such probes can reveal cellular or molecular lesions and increase treatment accuracy [2,3]. To be successful, these probes must (i) have significant detection capability, (ii) be able to assemble in the desired targeted area, and (iii) be biocompatible [4,5]. Diagnostic probes that use nanoparticles (NPs) as contrast agents can target tumors to enhance the detection of cancers [6], which afflict 10 million people annually according to the World Health Organization. ...
Preprint
Full-text available
Purpose: We synthesized folic acid-conjugated Fe 3 O 4 /Au-pralidoxime chloride Nanoparticles (Fe 2 O 3 /Au@PAM NPs) for use as dual-modal contrast agents for Magnetic Resonance Imaging (MRI) and Computed Tomography (CT) in the diagnosis of breast cancer. Materials and Methods: Fe 2 O 3 /Au@PAM NPs labeled or not to folic acid were synthesized and analyzed by dynamic light scattering, transmission electron microscopy, and vibrating sample magnetometry. The ability of these NPs to create image contrast was also investigated in silico and in vitro (in MCF-7 breast cancer cells and A549 lung cancer cells) with CT and MRI. Results: Dynamic light scattering and transmission electron microscopy revealed that the Fe 2 O 3 /Au@PAM NPs were nearly spherical. The average diameter of Fe 2 O 3 /Au NPs increased from 11.6 nm to 98 nm after folic acid conjugation. The saturation magnetization values of Fe 2 O 3 /Au@PAM NPs with and without folic acid conjugation were 25.56 and 32.6 emu/g, respectively. Conjugation of folic acid to NPs greatly improved their uptake by cancer cells. The additional coating of NPs with FA reduced the T2 relaxation time and signal intensity for MRI. Folic acid-labeled MCF-7 cells had a radiodensity measurement of 208 Hunsfield Units (HU) compared to 95 HU for A549 cells. For breast cancer cells, NPs labeled with folic acid significantly improved the X-ray absorption coefficient as a sign of active cellular uptake compared to NPs without labeling. Conclusion: Folic acid-labeled Fe 2 O 3 /Au@PAM NPs can serve as dual CT/MRI contrast agents and improve the sensitivities of both modalities for the detection of cancer cells.
... Cancer is one of the most common diseases nowadays and derives from the uncontrollable growth of a single cell [1]. There are many types of cancer with few typical or common characteristics, and its treatment is challenging. ...
Article
Full-text available
Cancer is one of the most common diseases nowadays and derives from the uncontrollable growth of a single cell. Magnetic nanoparticles (NpMag) offer various possibilities for use in the biomedical area, including drug delivery mediated by magnetic fields. In the current study, we evaluated the in vitro effects of iron-oxide magnetic nanoparticles conjugated with the antitumor drug doxorubicin (Dox) on human breast cancer cells. Our results revealed that magnetic nanoparticles with Dox (NpMag+Dox) induce cellular redox imbalance in MCF-7 cells. We also demonstrate that iron-oxide nanoparticles functionalized with Dox induce oxidative stress evidenced by DNA damage, lipid peroxidation, cell membrane disruption, and loss of mitochondria potential. As a result, NpMag+Dox drives MCF-7 cells to stop the cell cycle and decrease cell migration. The association of NpMg+Dox induced a better delivery of Dox to MCF cells, mainly in the presence of a magnetic field, increasing the death of MCF cells which might reduce the toxicity for healthy cells providing a better efficacy for the treatment. Thus, iron-oxide nanoparticles and doxorubicin conjugated may be candidate for anticancer therapy.
... Các vật liệu tổ hợp chứa hai pha từ tính có thể ẩn chứa những hiệu ứng và tính chất nổi trội như trao đổi hiệu dịch, liên kết trao đổi, trạng thái siêu thuận từ, lực kháng từ tăng cường, từ trở khổng lồ,… [7] - [11]. Liên kết trao đổi hoàn hảo giữa 2 pha từ tính có thể nâng cao khả năng ứng dụng của nam châm nano tổ hợp [9], y-sinh [3]; trao đổi hiệu dịch lớn mang đến tiềm năng ứng dụng trong bộ nhớ truy cập ngẫu nhiên, điện tử spin [11], phối hợp trở kháng tốt tạo tiền đề tốt cho ứng dụng trong hấp thụ sóng điện từ trong vùng tần số cao [8]. ...
Article
Ảnh hưởng của nhiệt độ ủ lên cấu trúc, hình thái và tính chất từ của các mẫu bột tổ hợp nano Fe/Fe3O4 đã được khảo sát. Các mẫu bột nano được chế tạo bằng cách kết hợp giữa nghiền bi năng lượng cao và ủ nhiệt tại các nhiệt độ 623, 723 và 823 K. Cấu trúc, hình thái các mẫu được đặc trưng bởi nhiễu xạ tia X, hiển vi điện tử quét. Các phép đo này chỉ ra sự tiến triển của các pha tinh thể của các mẫu khi nhiệt độ tăng. Đồng thời hình thái của các mẫu thay đổi bất thường tại các nhiệt độ ủ khác nhau: dạng hạt (623 K), dạng lá (723 K) và dạng thanh (823 K). Các phép đo từ nhiệt độ phòng cho thấy từ độ bão hòa giảm khi nhiệt độ ủ tăng, trong khi đó lực kháng từ tăng. Bên cạnh đó, sự có mặt của chuyển pha Verwey gần 120 K chứng tỏ phẩm chất tinh thể tốt của pha Fe3O4. Nghiên cứu của chúng tôi chứng minh khả năng điều khiển hình thái và từ tính trong hệ Fe/ôxit Fe thông qua sự ôxy hóa có điều khiển tại các nhiệt độ khác nhau.
... In addition to Mn-based nanomaterials, Fe-based nanoplatforms could also achieve the integration of treatment and imaging by releasing Fe 2+ (100). Existing clinical CAs are mainly gadolinium (Gd) chelates, which have short relaxation times and nephrotoxicity (101,102). Superparamagnetic iron oxide nanoparticles (SPIONPs) have been commercialized as a type of contrast agent for MRI, but their clinical application is limited owing to their poor T2-weighted imaging. To improve the imaging capability of SPIONPs, one strategy is to develop quasi-amorphous and hierarchical Fe 2 O 3 supraparticles. ...
Article
Full-text available
Hypoxia is an important component of tumor microenvironment and plays a pivotal role in cancer progression. With the distinctive physiochemical properties and biological effects, various nanoparticles targeting hypoxia had raised great interest in cancer imaging, drug delivery, and gene therapy during the last decade. In the current review, we provided a comprehensive view on the latest progress of novel stimuli-responsive nanomaterials targeting hypoxia-tumor microenvironment (TME), and their applications in cancer diagnosis and therapy. Future prospect and challenges of nanomaterials are also discussed.
Article
Full-text available
Magnetic nanoparticles (MNPs) are appealing materials as assistant to resolve environmental pollution issues and as recyclable catalysts for the oxidative degradation of resistant contaminants. Moreover, they can significantly influence the advancement of medical applications for imaging, diagnostics, medication administration, and biosensing. On the other hand, due to unique features, excellent biocompatibility, high curie temperatures and low cytotoxicity of the Iron-based nanoparticles, they have received increasing attention in recent years. Using an external magnetic field, in which the ferrite magnetic nanoparticles (FMNPs) in the reaction mixtures can be easily removed, make them more efficient approach than the conventional method for separating the catalyst particles by centrifugation or filtration. Ferrite magnetic nanoparticles (FMNPs) provide various advantages in food processing, environmental issues, pharmaceutical industry, sample preparation, wastewater management, water purification, illness therapy, identification of disease, tissue engineering, and biosensor creation for healthcare monitoring. Modification of FMNPs with the proper functional groups and surface modification techniques play a significant role in boosting their capability. Due to flexibility of FMNPs in functionalization and synthesis, it is possible to make customized FMNPs that can be utilized in variety of applications. This review focuses on synthesis, modifications, and applications of Iron-based nanoparticles.
Article
Full-text available
Citation: Kicheeva, A.G.; Sushko, E.S.; Bondarenko, L.S.; Kydralieva, K.A.; Pankratov, D.A.; Tropskaya, N.S.; Dzeranov, A.A.; Dzhardimalieva, G.I.; Zarrelli, M.; Kudryasheva, N.S. Functionalized Magnetite Nanoparticles: Characterization, Bioeffects, and Role of Reactive Oxygen Species in Unicellular and Enzymatic Systems. Abstract: The current study evaluates the role of reactive oxygen species (ROS) in bioeffects of mag-netite nanoparticles (MNPs), such as bare (Fe 3 O 4), humic acids (Fe 3 O 4-HA), and 3-aminopropyltri-ethoxysilane (Fe 3 O 4-APTES) modified MNPs. Mössbauer spectroscopy was used to identify the local surrounding for Fe atom/ions and the depth of modification for MNPs. It was found that the Fe 3 O 4-HA MNPs contain the smallest, whereas the Fe 3 O 4-APTES MNPs contain the largest amount of Fe 2+ ions. Bioluminescent cellular and enzymatic assays were applied to monitor the toxicity and anti-(pro-)oxidant activity of MNPs. The contents of ROS were determined by a chemiluminescence luminol assay evaluating the correlations with toxicity/anti-(pro-)oxidant coefficients. Toxic effects of modified MNPs were found at higher concentrations (>10 −2 g/L); they were related to ROS storage in bacterial suspensions. MNPs stimulated ROS production by the bacteria in a wide concentration range (10 −15-1 g/L). Under the conditions of model oxidative stress and higher concentrations of MNPs (>10 −4 g/L), the bacterial bioassay revealed prooxidant activity of all three MNP types, with corresponding decay of ROS content. Bioluminescence enzymatic assay did not show any sensitivity to MNPs, with negligible change in ROS content. The results clearly indicate that cell-membrane processes are responsible for the bioeffects and bacterial ROS generation, confirming the ferroptosis phenomenon based on iron-initiated cell-membrane lipid peroxidation.
Book
Full-text available
Today’s widespread activities in nanoscience and technology are actually rooted in the ideas of some leading scientists of the last century. Among them, the foremost name was Richard P. Feynman. He delivered a legendary talk entitled “there is plenty of room at the bottom” in the Annual general body meeting of the American Physical society on December 29, 1959 [1] at California Institute of Technology. In that talk he discussed about the ideas of manipulating and controlling things at the atomic scale. In this famous lecture the great scientist uttered also these famous words “the principles of physics, as far as I can see, do not speak against the possibility of maneuvering things atom by atom”. He also envisioned building circuits having sizes few nanometers that can be used as elements in more powerful computers. In that time these ideas were regarded ‘too speculative’. But nearly twenty years later from this prophetic talk, many inventions and discoveries began to appear justifying Feynman’s time ahead ideas. Soon the technology advisors and predictors realized the importance of these discoveries and declared that a new revolution is imminent namely nanotechnology, just as in the past we experienced industrial revolution and information revolution. Drexler expanded Feynman’s ideas and definition in a stimulating and lateral thinking way in his book, ‘Engines of Creation, the Coming Age of Nanotechnology’ [2]. We can quote Drexler to have some ideas about nanotechnology: “Nanotechnology is the principle of manipulation atom by atom, through control of the structure of matter at the molecular level. It entails the ability to build molecular systems with atom-by-atom precision, yielding a variety of nanomachines.” In recognition with this reality National Science and Technology Council (NSTC) of USA created an integrating working group on nanoscience, engineering and technology in 1998. Then in the year 2001 they announced the National Nanotechnology Initiative (NNI) programme with a large amount of fund in the budgetary provisions [3]. The major objective of this initiative was to create a common platform for the academia, industries and also private sector for working on this new technology. Following this example, most of the advanced countries in the world and many developing countries including China and India invested heavily on research and development in this new field of science and technology.
Article
Full-text available
The requirements for early diagnostics as well as effective treatment of insidious diseases such as cancer constantly increase the pressure on development of efficient and reliable methods for targeted drug/gene delivery as well as imaging of the treatment success/failure. One of the most recent approaches covering both the drug delivery as well as the imaging aspects is benefitting from the unique properties of nanomaterials. Therefore a new field called nanomedicine is attracting continuously growing attention. Nanoparticles, including fluorescent semiconductor nanocrystals (quantum dots) and magnetic nanoparticles, have proven their excellent properties for in vivo imaging techniques in a number of modalities such as magnetic resonance and fluorescence imaging, respectively. In this article, we review the main properties and applications of nanoparticles in various in vitro imaging techniques, including microscopy and/or laser breakdown spectroscopy and in vivo methods such as magnetic resonance imaging and/or fluorescence-based imaging. Moreover the advantages of the drug delivery performed by nanocarriers such as iron oxides, gold, biodegradable polymers, dendrimers, lipid based carriers such as liposomes or micelles are also highlighted.
Article
Full-text available
In our research, Pt nanoparticles by polyol method using AgNO3 were studied by UV-vis-IR spectroscopy, X-ray diffraction analysis, transmission electron microscopy (TEM) and high-resolution (HR) TEM methods. Their specific surfaces were investigated in the controlled size and morphology in the range of 10 nm without AgNO3, and in the range of 20 nm with AgNO3. Here, AgNO3 is crucial to appearance of polyhedral Pt nanoparticles but their convex or concave roughness characterizations are confirmed in the range of 20 nm without AgNO3. The high crystallization degree of the as-prepared Pt nanoparticles was discovered in the HRTEM evidences. In addition, the interesting complexity of surface roughness showed important evidences of bonding and chemical changes. Re-nucleations and re-crystallizations at their connections by surface attachment were discovered. The results show importance of controlling size and morphology, and big challenge for HRTEM method is predicted, which is how to measure many nanoparticles with high resolution with good quality of fine lattice-fringe systems around 10 nm.
Article
Full-text available
In the last decade the use of nanomaterials has been having a great impact in biosensing. In particular, the unique properties of noble metal nanoparticles have allowed for the development of new biosensing platforms with enhanced capabilities in the specific detection of bioanalytes. Noble metal nanoparticles show unique physicochemical properties (such as ease of functionalization via simple chemistry and high surface-to-volume ratios) that allied with their unique spectral and optical properties have prompted the development of a plethora of biosensing platforms. Additionally, they also provide an additional or enhanced layer of application for commonly used techniques, such as fluorescence, infrared and Raman spectroscopy. Herein we review the use of noble metal nanoparticles for biosensing strategies--from synthesis and functionalization to integration in molecular diagnostics platforms, with special focus on those that have made their way into the diagnostics laboratory.
Book
This textbook is aimed primarily at the senior undergraduate and first year graduate students from the various engineering and sciences departments including physics, chemistry, materials engineering, chemical engineering, electrical engineering, mechanical engineering, bioengineering, and biology. Researchers in the areas of nanomaterials and nanoscience will also find the book useful for building the background necessary to understand the current literature and as a reference book. The text assumes only a basic level of competency in physics, chemistry and mathematics. Some of the background material and introductory matter are included in the first few chapters and as appendices. Although this material may be familiar to some of the students, it is the author's experience after teaching such a course for many years that this cannot be taken for granted and moreover, serves as a ready reference to understand the text. As the area of nanoscience, nanotechnology and nanomaterials is a fast developing one, an approach which equips the students to comprehend the developing field rather than providing a large volume of information is essential. With this in view, while providing a broad perspective, the book emphasizes basics of nanoscience and nanoscale materials and goes into sufficient depth for the reader to be able to handle numerical problems. The treatment is kept at a level which is easily comprehensible to an undergraduate student. Solved examples are provided in each chapter to aid understanding and a set of problems is given at the end of each chapter. © 2013 by World Scientific Publishing Co. Pte. Ltd. All rights reserved.
Chapter
Toxicology is the branch of medicine that deals with the study of the adverse effects of chemicals and biological agents on the human body. It is the study of symptoms, mechanisms, treatments, and detection of poisoning. The broad scope of toxicology covers not only the adverse effects of therapeutics but also environmental agents and poisons. Nanotoxicology covers safety issues relevant to nanomaterials.
Chapter
Clinical application of molecular technologies to elucidate, diagnose, and monitor human diseases is referred to as molecular diagnosis. It is a broader term than DNA (deoxyribonucleic acid) diagnostics and refers to the use of technologies that use DNA, RNA (ribonucleic acid), genes, or proteins as bases for diagnostic tests. The scope of the subject is much wider and includes in vivo imaging and diagnosis at the single-molecule level. A more detailed description of molecular diagnostics is presented elsewhere (Jain 2012a).
Article
The influence of the oleic acid surface coating on Fe3O4 and NiFe2O4 nanoparticles on their magnetic and calorimetric characterization was investigated. Fe3O4 nanoparticles (particle sizes of 15–20 and 20–30nm) and NiFe2O4 nanoparticles (particle sizes of 20–30nm) were dispersed in oleic acid. The surface coating resulted in a decrease in the dipole–dipole interaction between the particles, which in turn affected the coercivity and heat dissipation of the nanoparticles. The coercivity of the oleic-acid-coated nanoparticles was found to be lower than that of the uncoated nanoparticles. The temperature rise in the oleic-acid-coated nanoparticles was greater than that of the uncoated nanoparticles; this temperature rise was associated with the relaxation losses. The viscosity dependence on the self-heating temperature of Fe3O4 nanoparticles (15–20 and 20–30nm) under an ac magnetic field was measured. The temperature rise for both the Fe3O4 nanoparticles (15–20 and 20–30nm) exhibited a strong dependence on viscosity at each magnetic field frequency, and the contribution of Brownian relaxation loss to the temperature rise was revealed. Moreover, an in vitro cytotoxicity test of Fe3O4 and NiFe2O4 was performed using human cervical carcinoma cells (HeLa), and the cytotoxicity of NiFe2O4 nanoparticles was compared to that of Fe3O4 nanoparticles.
Chapter
Magnetic particles (MPs) have proved to be valuable tools for manipulation of cells or biomolecules, for transportation of chemical substances or transfer of energy to defined target sites in biological systems, and for clinical diagnostics and therapeutics—both in vitro as well as in vivo. MPs receive their magnetic properties and responsiveness to magnetic fields most often from the proven biocompatible iron oxides magnetite (Fe3O4) and maghemite (γ-Fe2O3). The small size of MPs allows them to pass through capillary vessels during blood circulation. In nanoparticulate form or under the influence of strong magnetic fields, some MPs can even extravasate through capillary walls into surrounding tissue and reach many of the cells in the human body. Magnetically assisted delivery of chemo- or radiotherapeutics to as well as the generation of heat (hyperthermia) at defined target sites in the body can thus be achieved for treatment purposes, such as tumor therapy. Furthermore, MPs can serve as site- and function-specific contrast agents and thus enhance the diagnostic potential of magnetic resonance imaging (MRI). On the biotechnological side, magnetic labeling of cells and biomolecules with MPs followed by magnetic separation has been utilized for the isolation and analysis of nucleic acids and specific cells, for protein purification, for the detection of pathogenic bacteria and viruses, and for gene transfection.
Article
Controlled release of drugs from nanostructured functional materials, especially nanoparticles (NPs), is attracting increasing attention because of the opportunities in cancer therapy and the treatment of other ailments. The potential of magnetic NPs stems from the intrinsic properties of their magnetic cores combined with their drug loading capability and the biochemical properties that can be bestowed on them by means of a suitable coating. Here we review the problems and recent advances in the development of magnetic NPs for drug delivery, focusing particularly on the materials involved.