Vous êtes sur la page 1sur 16

Downloaded from http://rsfs.royalsocietypublishing.

org/ on July 7, 2018

Working principle and application of


magnetic separation for biomedical
rsfs.royalsocietypublishing.org
diagnostic at high- and low-field
gradients
Sim Siong Leong1, Swee Pin Yeap1,2 and JitKang Lim1,3
Review 1
School of Chemical Engineering, Universiti Sains Malaysia, Nibong Tebal, Penang 14300, Malaysia
2
Faculty of Engineering, Technology and Built Environment, UCSI University, 56000 Cheras Kuala Lumpur,
Cite this article: Leong SS, Yeap SP, Lim JK.
Malaysia
2016 Working principle and application of 3
Department of Physics, Carnegie Mellon University, Pittsburgh, PA 15213, USA
magnetic separation for biomedical diagnostic SSL, 0000-0001-8247-0169
at high- and low-field gradients. Interface
Focus 6: 20160048. Magnetic separation is a versatile technique used in sample preparation for
http://dx.doi.org/10.1098/rsfs.2016.0048 diagnostic purpose. For such application, an external magnetic field is applied
to drive the separation of target entity (e.g. bacteria, viruses, parasites and
cancer cells) from a complex raw sample in order to ease the subsequent
One contribution of 12 to a theme issue task(s) for disease diagnosis. This separation process not only can be achieved
‘Multifunctional nanostructures for diagnosis via the utilization of high magnetic field gradient, but also, in most cases, low
and therapy of diseases’. magnetic field gradient with magnitude less than 100 T m21 is equally feas-
ible. It is the aim of this review paper to summarize the usage of both high
gradient magnetic separation and low gradient magnetic separation (LGMS)
Subject Areas:
techniques in this area of research. It is noteworthy that effectiveness of the
biomedical engineering, chemical engineering, magnetic separation process not only determines the outcome of a diagnosis
nanotechnology but also directly influences its accuracy as well as sensing time involved. There-
fore, understanding the factors that simultaneously influence the efficiency of
Keywords: both magnetic separation process and target detection is necessary. Moreover,
for LGMS, there are several important considerations that should be taken into
low gradient magnetic separation, high
account in order to ensure its successful implementation. Hence, this review
gradient magnetic separation, paper aims to provide an overview to relate all this crucial information by
magnetophoresis, magnetic particles, linking the magnetic separation theory to biomedical diagnostic applications.
biomedical diagnostic, disease detection

Authors for correspondence:


Swee Pin Yeap 1. Introduction
e-mail: yeapsw@ucsiuniversity.edu.my Fast, selective and accurate detection of diseases is critical in clinical diagnosis
JitKang Lim as it allows physicians to provide more precise and preliminary medical assess-
e-mail: chjitkangl@usm.my ments to patients. However, biological samples are usually exceptionally
complex due to the presence of multiple components. In order to perform dis-
ease diagnosis, it is essential to isolate the specific target entity (which is the
infectious agent to be detected) from the complex raw samples. This sample
preparation step is needed prior to analysis in order to speed up the screening
process [1] as well as to enhance the detection limit [2].
Separation of targeted entity for diagnostic purpose is possible through the
application of magnetophoretic force. The idea implemented here is to magnet-
ically isolate the target entity, either those with or without intrinsic magnetic
responsive characteristic (target entity without magnetic property must be
magnetically labelled with magnetic particles) [3], from a complex mixture by
the application of an external magnetic field (figure 1). Owing to the presence
of magnetic field gradient, magnetic materials will be magnetically aligned and
driven to the region with the highest magnetic field strength by magneto-
phoretic force [4]. This phenomenon is known as magnetophoresis which
Electronic supplementary material is available involves the motion of magnetic particles relative to their non-magnetic
online at http://dx.doi.org/10.6084/m9.fig- surrounding medium under a non-homogeneous magnetic field [5]. The mag-
share.c.3493029. netically trapped and concentrated samples can then be used for the subsequent

& 2016 The Author(s) Published by the Royal Society. All rights reserved.
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

(a) (b) (c) dedicated to its development in order to realize the true poten- 2
tial of LGMS technique for biomedical diagnostic as well as

rsfs.royalsocietypublishing.org
in other applications. Moreover, the underlying mechanism
of magnetophoresis also determines the LGMS rate and the
magnet effectiveness of this separation method [4]. Concurrently, the
time consumed to separate target entity from a complex
sample is one of the key factors that decides the duration of
the diagnostic process. Therefore, a thorough understanding
of the theoretical background of the LGMS process is impera-
tive in the design of magnetic separator used in a biomedical
diagnostic application. Other factors, such as magnetic particle
size, magnetic particle concentration and magnetization, also

Interface Focus 6: 20160048


target non-target non-target play inevitable roles in determining the effectiveness of
Figure 1. (a) Sample with a mixture of various components containing both LGMS for target detection. In this regard, it is expected that
target and non-target entities, (b) isolation of target entities using magnetic there will be several trade-offs originating from the complex
field, (c) concentrated target after removal of the non-targets. Note that the tar- interplay of all these factors and they are yet to be revealed
gets must possess intrinsic magnetic responsiveness, or have been pre-labelled and explored in depth.
with magnetic responsive particles. Hence, in this paper, it is our goal (i) to review the utiliz-
ation of MS technique in sample preparation for diagnosis of
diseases, (ii) to highlight the feasibility of LGMS (less than
routine analysis of target identification. This technique, com- 100 T m21) in biomedical diagnostic application, (iii) to discuss
monly known as ‘magnetic separation’ (MS), has gained important factors that simultaneously affect the effectiveness of
popularity in the preparation of samples for diagnostic pur- LGMS and target detection, and (iv) to discuss some criteria
pose. There are many advantages offered by the utilization of that are worth considering for future implementation of
this technique. First and foremost, physical contact between LGMS technique in biomedical diagnostic application.
the magnetic source and the biological samples can be omitted
throughout the entire separation process which in turn makes
MS to be biologically non-invasive and does not exert a detri- 2. Magnetic separation in isolation of target
mental effect on the biological components [6]. In addition,
MS technique is also known to be high-throughput, low-cost entity for diagnostic purpose
[7] and less energy intensive when a permanent magnet is As mentioned in the Introduction, MS technique has been
employed as magnetic source [8]. used in sample preparation for disease diagnosis. One of the
Owing to the benefits offered by MS technique, a great deal active researches in this area is the magnetic isolation of
of effort has been allocated in the design of different types of malaria-infected red blood cells (i-RBCs) for clinical diagnosis
magnetic separator, which allow the MS process to occur of malaria. Malaria, a life-threatening disease, is triggered by
either in a batch or continuous manner [9]. In addition, MS Plasmodium parasites [16]. The malaria parasites convert hae-
technique is feasible in wide-ranging length scales as it is moglobin in healthy cells to haemozoin [17] which contains
not only workable in a laboratory test tube but also displays iron(III) (Fe3þ) that exhibits a relatively stronger magnetic sus-
excellent performance in miniaturized devices [10,11]. Nam ceptibility compared with iron(II) (Fe2þ) in haemoglobin [12].
and co-workers employed microfluidic device with two outlets Owing to the presence of iron(III), i-RBCs display stronger
to separate late stage malaria-infected red blood cells (RBCs) paramagnetic effect than healthy red blood cells (h-RBCs)
which possessed relatively higher magnetic susceptibility in [12]. Thus, by taking advantage offered by the difference of
comparison with healthy RBCs [12]. Pamme & Wilhelm [13] their intrinsic magnetic susceptible property, fractionation of
demonstrated continuous MS of magnetically labelled cancer i-RBCs and h-RBCs can be achieved through the application
cells in a microfluidic separator with multiple outlets leading of external magnetic field (figure 2a) [12]. This technique
to the fractionation of the cancer cell–magnetic parti- serves as an alternative to the conventional isolation method
cle complexes according to their magnetic dipole moment. that relies on the density of i-RBCs [19]. By applying a similar
In addition, Chen and co-workers filled up their magnetic concept, the separation of deoxyhemoglobin RBCs (para-
separation chamber with irregular-shaped iron particles with magnetic) from white blood cells (diamagnetic) also can be
diameter ranging from 25 to 75 mm, in order to generate conducted by MS technique [20]. In this regard, the blood
highly localized and strong magnetic field gradient upon cell sorting can be achieved solely depending on the difference
exposure to an external magnetic field to allow the effective in intrinsic magnetic properties. For instance, the absence of
separation of HIV virions that had been labelled with magnetic bound oxygen to Fe atom causes deoxygenated RBCs to pos-
particles [14]. sess relatively higher magnetic susceptibility in comparison
In spite of all the sophisticated applications mentioned with that of oxygenated RBCs [3]. In fact, the feasibility of
earlier, MS itself is a subject of research with many open- MS technique in the isolation of RBCs from whole blood
ended questions yet to be answered. Particularly, MS technique samples has been discussed way back in the 1970s by Melville
can be further classified into high gradient magnetic separa- et al. [21] in their work published in Nature.
tion (HGMS) and low gradient magnetic separation (LGMS) However, it should be highlighted that most biological
according to the magnitude of magnetic field gradient compounds lack inherent magnetic properties in most situ-
employed in the separation process. In comparison with ations. Thus, it is necessary to artificially enhance the
HGMS, the design rules for LGMS are ill defined [15], and magnetic property of the target entity so that it can be separ-
hence, more theoretical and experimental works must be ated from the other non-magnetic components under an
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

(a) sample flow 3


(h-RBC and i-RBC) sheath flow h-RBC sample

rsfs.royalsocietypublishing.org
(b)
i-RBC sample
(i) (ii)
i iii
sheath flow
ii iv
permanent
h-RBC
magnet
i-RBC
(i) (ii) (iii) (iv)
Ni Ni Ni Ni 10 mm
injection of sample hydrodynamic separation process collection of
(h-RBC + i-RBC) focusing by magnetic force separated sample

Interface Focus 6: 20160048


(c)
magnetic field
(i) S (ii) no magnet 500 mm (iii) with magnet 500 mm
N
M(b)

M(a) buffer

sample
non-M y mixture
non-M, M(a), M(b)
x

Figure 2. (a) Schematic diagram showing the principle of i-RBC separation from a blood sample using a microfluidic device containing ferromagnetic wire. An
external magnetic field was applied normal to the ferromagnetic nickel (Ni) wire in order to create high magnetic field gradient. Paramagnetic i-RBCs were drawn
towards the Ni wire and isolated out from the h-RBCs, reprinted with permission from [12], copyright & 2013 American Chemical Society. (b) SK-BR3 cells incu-
bated (i) with IO that had been conjugated with Ab (dark blue), and (ii) with carboxyl-functionalized IO that had not been conjugated with Ab (less blue) (note:
Prussian blue staining was implemented to confirm the presence of Fe ions on the cell surface), reprinted with permission from [18], copyright 2011 Elsevier. (c)(i)
Schematic diagram illustrating a free-flow MS technique at which laminar flow was applied in x-direction, whereas magnetic field was applied in y-direction. Note
that the non-magnetic material moved along the laminar flow, while those with magnetic responsiveness were deflected from the direction of laminar flow. This
separation technique was used to separate HeLa cells in which HeLa cells that had been incubated with magnetic particles (ii) flowing straight through the channel
when there was no applied magnetic field and (iii) deflected in their flow direction upon application of magnetic field, reprinted with permission from [13],
copyright & 2006 Royal Society of Chemistry.

externally applied magnetic field. This intention can be plays an essential role in the specific binding of magnetic par-
accomplished by labelling the target entity with magnetic ticles onto the targets. Likewise, Chen and co-workers have
particles before subjecting to MS. Magnetite (Fe3O4), maghe- also shown the high selectivity of their antibody-conjugated
mite (g-Fe2O3) and commercially available Dynabeads are magnetic beads towards Salmonella enterica over other species
some of the commonly used magnetic particles for this of bacteria (Escherichia coli, Shigella spp., Staphylococcus aureus
purpose. This approach has been used in the detection of and spirillumcholera) due to the specific antibody–antigen rec-
various biological components such as bacteria, viruses ognition feature [22]. In other words, magnetic particles that
and parasites [10,14,22– 25]. In addition, early diagnosis of serve as magnetic carriers in the MS process can be made
cancer can also be achieved by magnetic separation of circu- more versatile to label different targets based on the different
lating tumour cells (CTCs) that had been specifically labelled strategies/compounds used to functionalize their surface.
with magnetic particles [18]. Besides attaching onto the target cell surface, magnetic
Owing to the reason stated above, surface modification of particles might be further internalized into the cell body.
the magnetic particles with specific molecules that only bind For instance, Pamme & Wilhelm [13] found the internaliz-
to the target entity, but not on other non-target components, ation of g-Fe2O3 nanoparticles with diameter less than
is the main prerequisite. For instance, Xu and co-workers [18] 10 nm into the target cells (i.e. mouse macrophages and
surface modified their iron oxide nanoparticles with a water- human ovarian cancer (HeLa) cells) in large quantity can
soluble polymer which was then followed by bio-conjugation later be used to facilitate the cell sorting via MS technique.
with anti-HER2 antibody. The as-prepared iron oxide- Here, the MS process was performed in continuous mode
antibody conjugates (IO-Ab) were then incubated in a fresh within a customized microfluidic chip where the magnetic
human whole blood that was spiked with SK-BR3 (a human particle-labelled cells were deflected from their original path-
breast cancer cell line that is HER2-positive). The cells labelled way and driven towards the region with the strongest
with IO-Ab were then magnetically isolated by using a Super- magnetic field gradient (figure 2c).
MagTM separator before analysis on the captured cells was More interestingly, diagnosis can also be performed by
conducted. It was reported that IO-Ab were preferentially measuring the amount of magnetic particles that are still
labelled to SK-BR3 cells in comparison with normal cells remained in the sample solution after the MS process. This
found in the blood sample. As illustrated in figure 2b, it can approach has been demonstrated in a recent work published
be seen that the amount of IO-Ab attached onto the cell is over- by Chen and co-workers in the detection of Salmonella enterica,
whelming (figure 2b(i)) in comparison to that of IO without Ab which is a food pathogen that affect human gastrointestinal
functionalization (figure 2b(ii)). This result has directly con- tract. In this work, antibody-conjugated magnetic particles of
firmed that the antibody (attached on magnetic particles) size 250 nm (MP250) and 30 nm (MP30) were first dispersed
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

free 4
(a) magnetic (b)
inlet

rsfs.royalsocietypublishing.org
particle

magnetic
particle

magnetic
particle
captured aggregate

N
S

magnetic
particle
circulating

Interface Focus 6: 20160048


flow
magnetically
susceptible
wires
outlet magnet
magnetic
layer of captured
particle
magnetic particles
magnet
Figure 3. Illustration of HGMS and LGMS processes. (a) The HGMS column is packed with randomly entangled magnetizable wires, which act as magnetic field
‘dehomogenizer’ to produce strong and localized magnetic field gradient near to it. Magnetic particles which are flowing through the column will experience an
exceptionally strong magnetophoretic force in the vicinity of the magnetizable wires and be captured on them. (b) Illustration of LGMS process. Permanent magnet
is used to create magnetic field gradient across the magnetic particle solution in which magnetic particles are going to be separated. The magnetic particles migrate
towards the region with highest magnetic field gradient (which is usually the region adjacent to the magnet) and are separated from the solution. Particle aggrega-
tion (due to the magnetic dipole – dipole interaction) and continuous circulating flow (as a consequence of hydrodynamic effect) occur throughout the entire process
and contribute to the distortion of the magnetophoretic pathway of magnetic particles.

into the biological sample in order to promote the formation magnetically responsive particles from their suspension [28].
of MP30-target-MP250 complex. Upon the application of an To date, HGMS has been proved to be feasible in mineral
external magnetic field, the magnetophoretic separation rates processing [29–31], waste removal [32–34], biotechnology
of MP250 and MP30-target-MP250 complex are considerably [35], as well as drug delivery [36,37]. In general, the operation
higher compared with that of MP30 owing to their greater of HGMS involves a column which is filled with randomly
magnetic dipole moment. Accordingly, the amount of the entangled magnetically susceptible wires and surrounded
target entity in the initial biological sample can be inferred by an electromagnet (figure 3a) [38]. By turning on the
from the transverse relaxation time of the water molecules electromagnet around/besides the column, the magnetically
(T2 value) surrounding the MP30 which remained within susceptible wires tend to dehomogenize the magnetic field
the biological sample after the MS process [22]. The posi- such that high magnetic field gradient (more than 100 T m21)
tive outcomes obtained from this novel approach definitely is produced in the vicinity of the magnetizable wires. As the
encourage more exciting exploration in this research area in suspension of magnetic particles is channelled through
the future. the column, the magnetic particles are captured on the magnet-
The MS process can also be conducted in a negative selec- ically susceptible wires and hence being isolated from the initial
tion manner. In this regard, the undesired entities are labelled solution. In contrast, LGMS involves only a simple set-up in
with magnetic particles and thus being separated during the which the separation of magnetic materials from the solution
MS process. On the contrary, the target entity, which is is performed typically by a hand held permanent magnet with-
unlabelled and magnetically irresponsive, is still suspended out any kind of packing. Under a non-homogeneous magnetic
in the biological sample after the MS process [26]. This field produced by the permanent magnet, magnetic particles
approach is appealing as the target entity remains unaltered are driven to migrate towards the magnetic source by magneto-
throughout the MS process. However, it might suffer low phoretic force and therefore being isolated out of the solution
target yield due to non-specific loss of the target entity or (figure 3b). Because the magnitude of magnetic field gradient
inadequate removal of undesired biological components [27]. involved in this set-up is generally less than 100 T m21, this
One of the factors that determines the successful implemen- type of MS technique is denoted as LGMS.
tation of all aforementioned MS applications is the magnetic
separator design. Hence, in the next section, design and
principle of MS technique are thoroughly discussed. 3.1. Working principle of high gradient magnetic
separation
Owing to the remarkable difference between magnetic suscep-
tibility of magnetizable wires and that of suspending medium,
3. Strategies of magnetic separator design the magnetic field within close range of the wires is signifi-
Basically, MS technique can be classified into two distinct cantly distorted which sequentially induces an intense
classes, namely HGMS and LGMS, according to the magnitude magnetic field gradient in that region [28,39]. Magnetic field
of magnetic field gradient employed in the separation process. gradient as high as 1260 T m21 can be created near the wires
In fact, HGMS has widely been implemented in the isolation of within the HGMS column when the electromagnet is switched
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

on [29]. As magnetophoretic attraction force experienced of magnetic particles, namely viscous drag and thermal 5
by a magnetic particle is directly proportional to the magnetic motion [43]. Viscous drag is the resistance encountered by

rsfs.royalsocietypublishing.org
field gradient (see equation (3.1)), the magnetic particle experi- an object that performs a relative motion with respect to
ence a huge magnetophoretic force as it passes through the the surrounding fluid. The magnitude of viscous drag can
region adjacent to the magnetizable wire [40]: be evaluated by the Stokes equation [46]:
Fm ¼ 43pr3 mo MrH: ð3:1Þ Fd ¼ 6phrv, ð3:2Þ

Here, Fm is magnetophoretic force exerted on single magnetic where Fd is viscous drag experienced by magnetic particle, h
particle, r is radius of magnetic particle, mo is the permeability is viscosity of the suspending fluid and n is magnetophoretic
of free space, M is volumetric magnetization of magnetic particle velocity of magnetic particle relative to fluid. Besides, thermal
and rH is magnetic field gradient. Such a gigantic magneto- motion is random fluctuation of magnetic particles that dis-
phoretic force is able to overcome viscous drag and Brownian rupts the deterministic pathway of magnetic particles along

Interface Focus 6: 20160048


fluctuation which oppose the magnetophoretic motion of mag- the magnetic field gradient. The intensity of thermal fluctu-
netic particles. Finally, the magnetic particles will be captured ation can be reflected by the magnitude of diffusion
on the wires and isolated from the solution. coefficient D of the given particle in the suspension as
Even though HGMS has been well established and widely demonstrated in the Stokes–Einstein equation [47]:
implemented in various applications, there are several draw-
kT
backs associated with the utilization of HGMS. First and D¼ , ð3:3Þ
6phr
foremost, the installation and operation cost of the HGMS is
extremely high as huge amount of energy is required for the elec- where k is Boltzmann constant and T is absolute temperature.
tromagnet to generate a magnetic field that is sufficiently intense According to equations (3.1) and (3.2), magnetophoretic
to induce a successful collection of magnetic particles [32]. Apart force Fm varies linearly with respect to r 3 while the opposing
from that, the highly complex and inhomogeneous magnetic viscous drag Fd is directly proportional to r. Hence, as the
field within the HGMS column prevents the further develop- magnetic particle (and hence r) is small, viscous drag
ment of analytical model to accurately describe the HGMS dominates the magnetophoretic force and thus the magnetic
process which in turn renders theoretical study of the given pro- particles migrate slowly towards the magnet pole. Conse-
cess exceptionally strenuous [28]. Last but not least, magnetic quently, more time is required to execute a successful
particles which have been attached on the wires are difficult to separation. Apart from that, the thermal fluctuation of magnetic
be removed due to the complicated set-up of the column. The particle is more intense as its size is small (see equation (3.3)).
retention of magnetic particles on the wires will greatly reduce This motion randomizes the magnetophoretic pathway of mag-
the separation efficiency of the HGMS column [41]. netic particle and this has caused the difficulty in controlling the
motion of magnetic particles. The relevant dimensionless num-
bers include Reynolds number (Re) and Péclet number (Pe)
which characterize the ratio of inertia force (magnetophoretic
3.2. Working principle of low gradient magnetic force in this case) to viscous drag force and diffusive force,
separation respectively [15]. According to this dimensionless analysis,
In order to overcome the disadvantages of HGMS described magnetophoretic force is more dominant than viscous drag
above, another MS scheme was introduced to induce the and diffusion in defining the motion of magnetic nanoparticle
magnetophoretic collection of magnetic particles from sus- providing that Re and Pe are larger than unity. Consequently,
pension. This separation technique is able to perform the these criteria must be fulfilled prior to the more comprehensive
magnetic particle collection without the intricate set-up of design of the given LGMS process.
magnetizable wires within the HGMS column. On the con- Despite the arguments mentioned above, LGMS is still a
trary, the only part required is a permanent magnet which feasible separation technique in the removal of magnetic
produces non-uniform magnetic field all over the magnetic particles from their suspension. Yavuz et al. [42] reported that
particle solution subjected to MS [42]. Owing to the non- magnetic nanoparticles as tiny as 12 nm can be collected
homogeneity of the magnetic field throughout the magnetic within a reasonable timescale by applying low magnetic field
particle solution, magnetophoretic motion is initiated in gradient. This phenomenon is mainly due to the self-aggregation
which the magnetic particles migrate towards the region of magnetic particles under an external magnetic field [48]. For
where the magnetic field gradient is the highest (figure 3b) instance, when a magnetic particle is exposed to an external
[43]. In such a way, magnetic particles are removed from magnetic field, it will be magnetized and acquire net magnetic
the suspending medium and the separation is accomplished. dipole moment. In order to lower the magnetic interaction
Because magnetic field strength declines rapidly with the dis- potential energy, magnetic particles with net magnetic dipole
placement from the magnet, magnetic field gradient across moment tend to align themselves in the direction of the magnetic
the entire volume of magnetic particle solution subjected to field lines and being driven towards the regions with higher
separation is usually lower than 100 T m21 even though a magnetic field [49]. In the course of the magnetophoretic
strong neodymium ferrite boron magnet (abbreviated as migration of magnetic particles, some particles will come close
NdFeB magnet and is a permanent magnet with extremely to one another. Because each magnetic particle with net mag-
strong remanent magnetization) is employed [44,45]. netic dipole moment is behaving as a ‘small’ magnet, magnetic
Yet, one of the most pronounced disadvantages of LGMS particles which are sufficiently near will experience magnetic
is originated from the weak magnetophoretic force (as a con- attraction among each other and finally ‘stick’ together [50].
sequence of low magnetic field gradient) encountered by the Accordingly, magnetic particles tend to collapse with one
magnetic particles to be separated [15]. There are two factors another forming larger aggregate [51]. Owing to their higher
which oppose the deterministic magnetophoretic migration magnetic volume, magnetic particle aggregate acquires much
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

larger magnetophoretic force which is able to overcome the given experimental condition were reported as 96% and 97%, 6
aforementioned opposing forces (i.e. viscous drag force and ther- respectively. It should be noted that the magnetic field gradient

rsfs.royalsocietypublishing.org
mal fluctuation) and move collectively towards the magnetic can be high at the location near to the surface of the magnetic
source with much magnetophoretic velocity [42]. Hence, by source and decay with the distance away from the magnetic
the aid of aggregation, magnetic particles can be separated source. As an example, Lin and co-workers designed a mag-
by low magnetic field gradient generated by a hand-held netic bioseparator by using two NdFeB magnet columns [1].
permanent magnet within a reasonable timescale. A small space was left between the two magnet columns and
being employed as the MS zone. Even though the magnetic
field gradient in the separation zone can be as high as approxi-
3.3. Application of high and low gradient magnetic mately 300 T m21 at the surface of the magnet columns, it
separations in biomedical diagnostic decays to 0 T m21 at the middle of the separation zone. This
Application of both HGMS and LGMS in biomedical diagnos- phenomenon has caused the average magnetic field gradient

Interface Focus 6: 20160048


tic has been proved to be feasible. Several published works in within the separation zone to be approximately 90 T m21
this area of research are summarized in table 1. The concept ori- (table 1). By using this set-up, it only took around 2 min to sep-
ginated from HGMS has been used to separate E. coli bacteria arate the E. coli that had been labelled with 180 nm diameter
from whole blood sample [10] as well as to concentrate and magnetic particles. Hence, LGMS appears as a beneficial and
purify HIV viral product in human plasma [14]. In order to effective tool in separation of particular cells, bacteria or viruses
generate localized and strong magnetic field gradient which for diagnostic purpose.
enables rapid and efficient separation of magnetically labelled Nevertheless, in order to design and optimize the diagno-
E. coli bacteria, magnetizable substance is employed to deho- sis process by incorporating LGMS technique, it is extremely
mogenize the magnetic field created by the magnetic source. vital to understand some of the control parameters that influ-
In the work reported by Xia and co-workers, magnetic field ence the performance of the process in the first place. This
gradient as high as 290 T m21 can be generated by NiFe micro- will be discussed in the next section.
comb that is acting as magnetic field dehomogenizer (table 1)
[10]. This recorded magnetic field gradient value is much
higher than the one that can be achieved within a microfluidic
channel (approx. 20 T m21) in the absence of NiFe microcomb. 4. Effectiveness of low gradient magnetic
In the purification of human immunodeficiency virus type 1 separation in biomedical diagnostic
virions, irregular iron oxide spheres were placed within the
microfluidic separator to act as magnetic field gradient con- application
centrator. Under this configuration, the solution containing
viral product can be concentrated within 20 min and yields a
4.1. Factors that influence low gradient magnetic
solution that is 40–80 times more concentrated in comparison separation rate
with the initial solution [14]. On the other hand, Pivetal and co- Size of magnetic particles plays a very important role in con-
workers designed a micro-magnet array which can develop trolling the LGMS kinetics. As depicted in equation (3.1), the
magnetic field gradient as high as 2.5  105 T m21 to position magnitude of Fm experienced by a magnetic particle is directly
and immobilize magnetic particle-labelled E. coli (row 11 in proportional to r 3. Here, force balance between Fm and Fd
table 1) [57]. reveals that the magnetophoretic velocity, which determines
Meanwhile, there are plenty of biomedical diagnostic the separation time, varies linearly with respect to r 2. Thus,
applications which have employed the concept of LGMS in in an LGMS process, particles with larger size are separated
their operation unwittingly and resulted in positive outcomes. first before the small ones when they are subjected to the
For instance, in the continuous sorting of monocytes and same magnetic field [58].
macrophages by magnetic nanoparticles, permanent magnet There exists plenty of literature in relating the MS effi-
(NdFeB) was used to create the magnetic field throughout ciency to the magnetic particle size. Lately, Lin and co-
the microfluidic chip which generates magnetic field gradient workers showed that E. coli O157:H7 which had been labelled
within the range of 30–80 T m21 (row 7 in table 1) [55]. with 180 nm magnetic particles can be successfully isolated in
Despite the low magnetic field gradient (less than less than 2 min within a magnetic bioseparator of mean mag-
100 T m21), excellent purification (more than 88%) and high- netic field gradient approximately 90 T m21 [1]. Meanwhile,
throughput (10–100 cells/s) could be accomplished in this when the same experiment was conducted by using magnetic
magnetophoresis set-up. Apart from that, in the separation particles of size 30 nm, the optimal MS duration was notably
of cancer cells from fresh whole blood as reported by Xu increased to 60 min, which is about 30 times slower in
and co-workers, SuperMagTM separator (Ocean NanoTech) comparison with the MS process that employed 180 nm
was used to trigger the magnetophoretic motion of the magne- magnetic particles. Thus, magnetic particles of larger size
tically labelled tumour cells (row 5 in table 1) [18]. Enrichment are preferential in fast detection/screening applications. By
factor of cancer cell to normal cell as high as 1 : 10 000 000 reversing this concept, Chen and co-workers used a mixture
has been achieved under the low magnetic field gradient of magnetic particles of two different sizes (i.e. 30 nm (MP30)
(100 T m21) created by permanent magnets within the mag- and 250 nm (MP250)) in bacteria and virus detection [22].
netic separator. Furthermore, in the work done by Song and Their preliminary study showed that MP250 was successfully
co-workers, magnetic scaffold was used to create magnetic precipitated in 1 min upon subjecting to a magnetic field of
field gradient below 100 T m21 for the isolation and detection 0.01 T; while MP30 remains suspended even after prolonging
of particular target cell from analytical samples (see table 1 the MS time to 1 h under the same magnetic field (figure 4a).
for detail) [54]. For instance, the magnetophoretic capture effi- By employing the advantage offered by such a huge distinc-
ciencies of leukaemia cells and prostate cancer cells under the tion in MS time, the concentration of the target entity within a
Table 1. Details of magnetic particles (MPs) or cells with intrinsic magnetic property used in biomedical diagnosis based on magnetic separation concept.

types of
a
magnetic particles , magnetic
or cells with intrinsic separator
magnetic size of MPs (batch/ magnetic field magnetic field application in
responsiveness (diameter) magnetic properties functionalizing layer concentration of MPs continuous) strength gradient biomedical diagnostic ref.
b 26 21
deoxygenated RBCs 7.7 mm Dx ¼ 0.265  10 — not counted batch mean ¼ 1.40 T mean ¼ 131 T m determine [52]
methemoglobin RBCs Dx b ¼ 0.301  1026 0.5  106 cells ml21 magnetophoretic
mobility of the cells
g-Fe2O3 8.4 nm 3.1  105A m21c carboxyl groups 1015 particles ml21 continuous 0.4 T 50 T m21 sorting of mouse [13]
macrophages and
human ovarian
cancer cells (HeLa
cells)
superparamagnetic green 1.6 mm n.a. streptavidin 3.1  109 beads ml21 continuous 0.048 Td 290 T m21d separation of living [10]
fluorescent bead E. coli bacteria
(43% iron oxide)
parasite-infected n.a. n.a. — n.a. continuous 1.426 T 804 T m21 concentrate erythrocytes [53]
erythrocytes parasitized by
human malaria
species
Fe3O4 27 + 5 nm n.a. oleic acid þ amphiphilic 2 mg Fe ml21 batch n.a. 100 T m – 1 separation of tumour [18]
polymer þ antibodies against human cells from fresh
epithelial growth factor receptor 2 whole blood
(anti-HER2 or anti-HER2/neu)
fluorescent–magnetic 211 nm 8.18 emu g21c monoclonal antibody (mAb) 10  1010 nanobioprobes ml21 batch 0.4 T 0– 100 T m21e detection and extraction [54]
bifunctional of leukaemia cells
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

nanoparticles and prostate cancer


cells
g-Fe2O3 8.7 nm 6.6  10214 A m2f (for carboxylate functional groupsh 1 mM (extracellular iron continuous 0.15– 0.26 T (from 30– 80 T m21 sorting of monocytes [55]
1 pg of irong) concentration) middle of the and macrophages
separation chamber based on amount of
to surface of the MPs internalize into
magnet) the cells
deoxygenated RBCs 6– 8 mm n.a. — 5  107 cells ml21 semi 0– 1.68 T 1750 T m21 RBC debulking [56]
methemoglobin RBCs n.a.

(Continued.)
7

Interface Focus 6: 20160048 rsfs.royalsocietypublishing.org


Table 1. (Continued.)

types of
a
magnetic particles , magnetic
or cells with intrinsic separator
magnetic size of MPs (batch/ magnetic field magnetic field application in
responsiveness (diameter) magnetic properties functionalizing layer concentration of MPs continuous) strength gradient biomedical diagnostic ref.

Fe3O4 30 nm n.a. streptavidin þ antibodies against 40 mg ml21 for 30 nm MPs batch 1.15– 1.65 T 0– 300 T m21 isolate E. coli O157:H7 [1]
E. coli (mean ¼ 1.35 T) (mean ¼ 90 T m21) cells
180 nm carboxyl functional 100 mg ml21 for 180 nm MPs
groups þ streptavidin þ antibodies
against E. coli
magnetic beads 30 nm, n.a. capture antibody (Ab1), detection 0.2 g l21i batch 0.01 T n.a. detection of S. enterica [22]
250 nm antibody (Ab2)
microbeads 50 nm n.a. streptavidinj n.a. batch  0.12 to0.29 T  2.89  104 – selective arraying of E. [57]
2.5  10 T m21 coli bacteria by
micro-magnet
a
Magnetic particles that were used to label on target entity which is originally non-magnetically responsive.
b
Dx ¼ net magnetic susceptibility of the RBC in aqueous suspension¼ xRBC – xH2 O .
c
Saturation magnetization.
d
The values of magnetic field strength and magnetic field gradient provided were the maximum (which occurs at the location closest to the magnet within the whole domain of flowing channel) under the influence of NiFe microcomb.
e
Simulation based on the geometry configuration provided by using COMSOL Multiphysics (see electronic supplementary material for the simulation result).
f
Magnetic moment.
g
1 pg ¼ 8  105 nanoparticles.
h
The carboxylate functional groups here are to stabilize the magnetic particles in aqueous medium.
i
The concentration of magnetic beads is estimated in the solution where magnetic separation occurs.
j
During immunomagnetic labelling, the streptavidin-coated microbeads were mixed with bacterial cells that already pre-treated with biotinylated anti E. coli antibodies.
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

Interface Focus 6: 20160048 rsfs.royalsocietypublishing.org


Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

(a) (b) 9
(i) (iii)

rsfs.royalsocietypublishing.org
50 mm

(ii) (iv)

MP250 MP30
(1 min) (60 min)

Interface Focus 6: 20160048


(c) (i) (ii)
(d)
normalized absorbance (arb. units)

1.5 1.0 magnet

50 mT mm–1
0.9
0.025 g l–1
0.8
normalized opacity

0.05 0 g l–1
10 g l–1 exit
1.0 0.7 0.075 g l–1
1 g 1–1 0.25 g l–1
6
0.6 0.50 g l–1
0.1 g 1–1 0.70 g l–1
0.5 5
0.01 g 1–1 1 g l–1
4
0.4 2.5 g l–1
0.5 0.3
5 g l–1
3
10 g l–1
0.2 2
0.1 1
1 mm
0 0 12 mm
0 100 200 0 200 400 600 800 diameter cells
time (s) time (s)

Figure 4. (a) MP250 was separated in 1 min by using magnetic field of 0.01 T. Meanwhile, MP30 remains well dispersed even after 60 min subjected to the same
magnetic field, reprinted with permission from [22], copyright 2015 American Chemical Society. (b) Optical micrographs of solution with 1 g ml21 of 0.41 mm
superparamagnetic microsphere (Estaporw M1 – 030/40) are demonstrated in (i) and (ii). Figure (i) was observed under the absence of magnetic field, while figure
(ii) is the image taken after 120 s of magnetic field exposure. The alignment of superparamagnetic microsphere along the magnetic field was observed in (ii) and
this is an indication of magnetic particle aggregation under low gradient magnetic field, reprinted with permission from [48], copyright & 2008 American Chemical
Society. Optical micrograph of 425 nm magnetic nanoparticles in deionized water (iii) before the application of magnetic field and (iv) a few seconds after the
exposure to magnetic field generated by NdFeB magnet, reprinted with permission from [45], copyright & 2008 American Institute of Physics Publishing LLC. Chain
formation ( particle aggregation) was observed in (iv) and this phenomenon has proven the occurrence of cooperative magnetophoresis. (c) The magnetophoresis
profile for (i) Estaporw M1 – 030/40 particles, reprinted with permission from [48], copyright & 2008 American Chemical Society, and (ii) 30 nm bare iron oxide
magnetic nanoparticles, reprinted with permission from [58], copyright & 2014 American Chemical Society. Both (i) and (ii) show that the magnetophoresis process
is accelerated as magnetic particle solution with higher concentration is employed. The variation of magnetophoresis rate with the concentration of the magnetic
particle solution indicates the significance of particle aggregation throughout the magnetophoresis. (d ) Illustration of magnetic separation chamber used for the
fractionation of mouse macrophages or human ovarian cancer cells (HeLa cells) according to the cell magnetic moment, reprinted with permission from [13],
copyright & 2006 Royal Society of Chemistry.

biological sample can be measured by dispersing the magnetic [50,51]. Particle aggregation is initiated by the magnetic inter-
particle mixture into the given solution. Under this scenario, the action between magnetic dipole moment possessed by each
interaction between antibody (from magnetic particles) and magnetic particle. As suggested by De Las Cuevas and co-
antigen (from targets) would cause the formation of large workers, the significance of magnetic interaction between
MP250-target-MP30 complex. The target entities (bacteria or two magnetic particles can be estimated by magnetic Bjerrum
virus) which were embedded in MP30-target-MP250 complex length lB which is formulated as [48]
can then be rapidly removed upon exposure to a weak magnetic
 1=3
field due to the gigantic size of MP250-target-MP30 complex in 8pm0 M2
lB ¼ r2 , ð4:1Þ
comparison with MP30. The unreacted MP30 which were still 9kT
suspended in the solution after the MS will change the T2
value of the surrounding water molecules. The change of T2 where m0 is the permeability of free space. Particularly,
values was then correlated to the concentration of the target magnetic interaction is dominant over the thermal random
(bacteria or virus) in the initial biological sample. Results from motion and particle aggregation is initiated as the separation
the studies mentioned above have highlighted the importance distance between both magnetic particles is smaller than
of selecting magnetic particles of suitable size for successful the magnetic Bjerrum length. Because the average distance
employment of MS technique in biomedical diagnosing. between magnetic particles suspended in the solution
In addition to particle size, the separation kinetic of MS is decreases with the particle concentration of the solution,
also strongly dependent on the concentration of the magnetic magnetic interaction between particles (which induces parti-
particles. This phenomenon is attributed to the cooperative cle aggregation) is exceptionally substantial in the LGMS of
nature of the magnetic particle system in which magnetic par- highly concentrated magnetic particle solutions. In addition,
ticles tend to aggregate and move collectively towards the according to thermodynamic self-assembly theory as proposed
magnetic source upon exposure to an external magnetic field by Andreu et al. [59], the onset of particle aggregation in
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

magnetic particle solution subjected to magnetophoresis can or volume [62]) of magnetic particles is also an important cri- 10
be predicted by aggregation parameter N* which is defined as terion which affects the magnetophoretic separation rate of

rsfs.royalsocietypublishing.org
qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi magnetic particles in any related biomedical diagnostic appli-
N  ¼ fo eG1 , ð4:2Þ cation. The linear correlation between magnetophoretic force
experienced by magnetic particles and their magnetization
where fo is the volume fraction of magnetic particles in the has been demonstrated in equation (3.1). Magnetic particles
given solution and G is the magnetic coupling parameter with stronger magnetization (and hence more intense magnetic
which is given by dipole moment) experience larger magnetophoretic force
under the same external magnetic field and undergo more
m0 m2s
G¼ : ð4:3Þ rapid MS. One related example which employs this principle
2pd3 kT
is demonstrated in the work reported by Pamme & Wilhelm
Here, ms is the total magnetic dipole moment possessed by [13], in which target cells were fractionated according to their

Interface Focus 6: 20160048


one magnetic particle at saturation and d is the diameter of mag- magnetic loading. The target cells were firstly incubated with
netic particle. As N* is larger than unity, particle aggregation is nano-sized magnetic particles in order to allow the internaliz-
initiated by the magnetic interaction between neighbouring ation of magnetic particles into the cells. Cells that were loaded
magnetic particles [60]. Besides, the magnitude of N* is also with more magnetic particles possessed higher magnetic
representing the average number of particles within a particle moment; while those loaded with fewer magnetic particles
aggregate in the cooperative regime. Both magnetic Bjerrum exhibited lower magnetic moment. When the mixture of cells
length lB [48] and aggregation parameter N* [59] analysis (with different magnetic moments) was driven to flow through
suggests that the particle aggregation is more significant at a microfluidic chamber where the magnetic field gradient was
higher particle concentrations. The direct consequence of particle applied in the direction perpendicularly to the flow velocity,
aggregation is the formation of larger-sized magnetic particle the cells start to deflect from their original flow direction
clusters with much stronger magnetic response in comparison under the influence of magnetophoretic force. Cells with
to individual magnetic particles. In the work reported by De higher magnetic moment were deflected to a greater extent
Las Cuevas et al. [48], they observed instantaneous align- towards the location where the external magnet was applied
ment of Estapor M1–030/40 particles (the diameter of which is and vice versa (figure 4d). By this way, mixture of cells is fractio-
0.41 mm) upon the application of an external magnetic field nated according to their magnetic loading due to their different
(figure 4b(i)(ii)). Meanwhile, Schaller et al. [45] also reported the MS responsiveness. Similarly, this principle has also been
similar result in their attempt to investigate the motion of mag- successfully applied in the sorting of monocytes and macro-
netic particles under an external magnetic field (figure 4b(iii)(iv)). phages based on the amount of magnetic particles that have
Formation of particle aggregates, which possess larger been internalized into the cells [55].
size and more intense magnetic response, leads to accelerated
MS such that magnetic particles can be segregated from the
solution within a shorter timescale. Under this condition,
4.2. Factors that simultaneously affect the performance
magnetic particles interact along their magnetophoretic of target detection
migration pathway; thus, this phenomenon is designated as Apart from affecting the separation rate, particle size is also
cooperative magnetophoresis. In line with the same coopera- important in determining the efficiency of target detection.
tive concept, MS is faster as the concentration of magnetic Generally, micrometre-sized (1 –10 mm) and nanometre-
particles increased (figure 4c) [4,48,58]. Therefore, this prin- sized (50 –100 nm) magnetic particles are popular for the
ciple can be used in biomedical diagnostic applications isolation of target entities from complex biological samples
which employ MS technique to isolate a target entity. Even for diagnostic purpose [14]. Even though magnetic particles
though the separation of the target entity by external mag- of larger size are preferable in terms of their shorter LGMS
netic field can be accelerated by labelling the target entity duration (as discussed in §4.1), they pose several shortcom-
with larger magnetic particles, the utilization of larger mag- ings as compared with the nano-sized counterparts. First,
netic particles offers some drawbacks. First and foremost, the surface area to volume ratio of micrometre-sized particles
due to the fact that the ratio of surface area to volume is redu- is lower compared with nano-sized particles which in turn
cing with the increase in magnetic particle size, the available causes the reduction in the binding capacity [18] and capture
surface sites for labelling become fewer. Furthermore, the efficiency [63] for magnetic particles with larger size. Second,
relative size between magnetic particle and target entity also gravitational sedimentation is the common issue encountered
has a significant effect on the sensitivity in certain biomedical by large and dense particles [14,27], hence, continuous agita-
detection which exerts extra constraint on the selection of the tion is needed to keep them dispersed. On the contrary, small
magnetic particle size (will be discussed in more detail in the particles with nano-sized dimension are usually dispersed
next section) [61]. Henceforth, cooperative nature of magnetic more uniformly within the suspension [1] and exhibit faster
particles throughout the LGMS process provides an alternative reaction kinetics [1,63]. Additionally, due to their less steric
route to optimize the separation rate of magnetically labelled exclusion property, higher amount of nano-sized particles
target entity in biomedical diagnostic application. By increas- can be bound onto the surface of a target cell in comparison to
ing the concentration of magnetic particles supplied to the that of micrometre-sized particles (as illustrated in figure 5a)
sample filled with the target entity, the magnetophoretic separ- [18]. A clear comparison between target cells that were
ation rate of magnetically labelled target entity can be greatly attached with magnetic particles of different sizes is given in
enhanced upon cooperative movement of the aggregated figure 5b. Figure 5b(i) shows immobilization of nano-sized
magnetically responsive species. magnetic particles on the surface of E. coli [64]; while
In addition to the size and concentration effects, magnetiza- figure 5b(ii) shows E. coli O157:H7 cells which were bound to
tion (which is the total magnetic dipole moment per unit mass antibody-coated magnetic beads (Dynabeads). In addition,
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

(a) (b) 11
(i)

rsfs.royalsocietypublishing.org
membrane 1 mm (ii)
of target
cell

100 nm magnetic
particles

Interface Focus 6: 20160048


30 nm 200 nm

(c) (d) (e) 10 mg ml–1


1 mg ml–1
600 0.5 mg ml–1
105 0.1 mg ml–1

probe excess
100 equivalence

DT2 (ms)
400
95
T2 (ms)

target excess
90 200
85
high T2 80 0
low T2
75 1 10 102 103 104 105 106 107
0.001 0.01 0.1 1 concentration of S. enterica (cfu ml–1)
no. target analytes/ no. probes

Figure 5. (a) A pictorial drawing (not drawn to scale) showing membrane of a target cell labelled with magnetic particles of various sizes (more nano-sized
magnetic particles can be attached on the cell surface than the micrometre-sized counterparts due to less steric exclusion) [18]. (b)(i) Transmission electron
microscopy image showing E. coli immobilized with 3-mercaptophenylboronic acid/1-decanethiol-modified magnetic nanoparticles, reprinted with permission
from [64], copyright & 2013 Multidisciplinary Digital Publishing Institute; (ii) electron micrograph (scale bar, 5.14 mm) of E. coli O157:H7 cells captured by Dyna-
beadsw M-280 Streptavidin (Dynal, Lake Success, NY, USA) coated with biotinylated antibodies, reprinted with permission from [65], copyright & 2002 John Wiley
and Sons. (c) Solution with well-dispersed magnetic particles provides high T2 value. Once the target cell was mixed with the magnetic particle solution, interaction
between antibody (from the functionalization layer of magnetic particles) and antigen (from the target cells) would induce the aggregation between magnetic
particles and cells which results in large magnetic particles– cells complex. Owing to the presence of the larger-sized complex, T2 value of the solution becomes
lower, reprinted with permission from [61], copyright & 2009 American Chemical Society. (d) Variation of T2 value with the number ratio of target cells to
magnetic particle probes, reprinted with permission from [61], copyright & 2009 American Chemical Society. (e) The effect of magnetic particle (MP30) concen-
tration (0.1, 0.5, 1 and 10 mg ml21) on the sensitivity of the detection process. It can be observed that the sensitivity (change of T2 value) of the detection method
is decaying with the concentration of MP30, reprinted with permission from [22], copyright & 2015 American Chemical Society.

Pamme & Wilhelm [13] predicted that the number of magne- individual magnetic particles) tend to cause magnetic field dis-
tic particles bound to the surface of a target cell can be tortion around them and diphase the spins of water protons
significantly reduced from 2  106 to 102 and 100 when the which are diffusing through the region more significantly
diameter of the magnetic particles is enlarged from 9 nm to (figure 5c) [61]. Thereby, the concentration of the target entity
400 nm and 2.8 mm, respectively. This prediction implies that in the initial solution can be inferred from the percentage
smaller magnetic particles are more appropriate for use in change of T2 value measured by a relaxometer [66]. However,
the analysis of high-density cell surface markers; while the degree of magnetic particle aggregation (and hence change
for detection of rare cell surface markers, larger magnetic par- of T2 value) is highly dependent on the number ratio of the
ticles are more appropriate. Moreover, using smaller particles target entity to magnetic particles. For instance, when magnetic
approaching the size of surface markers, the resolution of particles with a diameter of 70 nm were employed as the probe
diagnostic process could be improved significantly. to analyse the concentration of 8 nm entity (BSA-(Tag)6) which
Apart from acting as one of the non-trivial parameters has a smaller size than the magnetic particle probe, T2 value
which govern the dynamical behaviour of cooperative LGMS tends to decrease with increasing concentration of target
(separation rate), concentration of magnetic particle solution entity until a minimum (denoted as equivalence point) is
also affects the performance of LGMS-aided biomedical diag- reached (as shown in figure 5d). However, beyond the equival-
nostic application in term of other aspects. After dispersing ence point, T2 value increases with target entity concentration
antibody-conjugated magnetic particles into the biological as the target entities which are present in excess have saturated
sample containing the target entity, the binding between anti- the binding sites of magnetic particles and prevent the further
body (from magnetic particles) and antigen (from the target aggregation of magnetic particles [61]. This phenomenon is
entity) fosters the aggregation of magnetic particles in the sol- known as prozone effect. The intervention of prozone effect
ution which in turn changes the spin–spin relaxation time (T2) has distorted the anti-correlation between concentration of
of the surrounding water protons. In general case, T2 value the target entity and T2 value when the number ratio of
declines with the degree of magnetic particle aggregation as target entity to magnetic particles exceeds the equivalence
particle aggregates (with larger size in comparison to point [66]. Thereby, in order to obtain reliable measurements
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

on concentration of the target entity, it is necessary to disperse of iron oxide (Fe2O3) nanoparticles [72]. Hence, even though 12
sufficient amount of magnetic particles into the given solution the utilization of highly concentrated magnetic particle sol-

rsfs.royalsocietypublishing.org
so that the number ratio of target entity to magnetic particles is ution is capable of increasing the MS rate in biomedical
well below the equivalence point. diagnostic application, there exists certain constraint for us to
Additionally, concentration of magnetic particles also select magnetic particle solution with a suitable concentration
affects the sensitivity of target detection by MS. This phenom- in the design of biomedical diagnostic process. Apart from opti-
enon is observed in a bacteria and virus detection method mizing the MS rate by increasing the concentration of magnetic
proposed which involved strong magnetic responsive particles, it is also necessary to ensure the concentration falls
MP250-target-MP30 complex and poor magnetic responsive within the range that is characterized by excellent sensitivity
unreacted MP30 [22]. Here, unreacted MP30 will still remain and biological harmlessness.
suspended after being subjected to an external magnetic To summarize all the above discussion, figure 6 is presented
field. Because concentration of MP30 dispersed in the solution to highlight the complex interplay between the particle size,

Interface Focus 6: 20160048


has a strong effect on the T2 value, the percentage change of particle concentration and magnetization, which determine
T2 value of the solution after the MS process can be used to the design of LGMS for biomedical diagnostic application.
infer the concentration of the target entity in the initial sol-
ution. However, the sensitivity of this bacteria or virus
detection method is strongly dependent on the concentration
of MP30 supplied in the initial solution (figure 5e). As MP30 5. Concluding remarks and future considerations
have been oversupplied, the fraction of MP30 involved in Over the past few decades, tremendous efforts have been
the formation of MP250-target-MP30 complex is too low to dedicated to the development of MS technique for sample
provide noticeable change in T2 value of the solution after purification prior to the diagnosis of diseases. In this regard,
undergoing MS. On the contrary, when the concentration of understanding the underlying principles that govern the
MP30 in the initial solution is too low, the magnetic signal dynamical behaviour of MS is critical in the optimization of
obtained from T2 value measurement is not sufficiently separation efficiency. Concurrently, along with the progressive
intense to provide reliable result which accurately reflects growth in research works related to magnetophoresis of mag-
the concentration of the target in the initial solution. Further- netic particles under low magnetic field gradient, more
more, the linear relationship between the change of T2 value findings directly related to its working principle have been dis-
and the target concentration can only be observed in a par- covered and disclosed. In the near future, those findings
ticular range of target concentration [22]. Therefore, in definitely should be taken into consideration for any design
order to have an accurate measurement of the target concen- of LGMS technique for biomedical diagnostic application.
tration by using MS method, concentration of the magnetic First and foremost, magnetic particles employed to magne-
particles and the target must be carefully tuned to certain tically label the specified target entity must possess good
range which provide reliable feedback from the analysis. colloidal stability. This prerequisite is needed to ensure the par-
Furthermore, for MS-based in vivo diagnosis, the size and ticles present in well-dispersed condition so that more effective
concentration of magnetic particles employed must be con- mixing and tagging with target entity can be promoted. Apart
sidered carefully as their nanotoxicity is unclear. In this from imparting good colloidal stability to the functionalized
regard, Nel et al. [67] demonstrated that nano-sized magnetic magnetic particles in the initial suspending medium, it is para-
particles are biologically toxic due to their large exposed sur- mount to ensure their colloidal stability is not disrupted by
face area that facilitates harmful interaction with biological different biological matrices (e.g. DPBS, RPMI-1640, human
systems. For instance, Kunzmann and co-workers showed plasma) throughout the magnetic labelling process [18]. Gener-
the decay of cell viability of human monocyte derived ally, stability of magnetic particles can be strengthened by
dendritic cells upon exposure to CSNPs (silica coated iron imparting electrostatic repulsion and steric hindrance to the
oxide nanoparticles) with size of 30 and 50 nm. On the con- given particles. However, it was reported that enhanced col-
trary, this phenomenon is not observed throughout their loidal stability compromised the MS rate of magnetic
exposure to larger CSNPs (70 and 120 nm) [68]. In parallel, particles under low magnetic field gradient [73]. Hence, the
the same authors also observed that micro-sized titanium conflicting effect between colloidal stability and MS efficiency
oxide (TiO2) caused relatively higher DNA damage com- must be carefully assessed in the design of LGMS technique for
pared with their nano-sized counterpart due to their higher biomedical diagnostic application.
surface reactivity [69]. Hence, the toxicity dependency on As discussed in §3.2, several limitations exist in the isolation
magnetic particle size cannot be solely coming from the of tiny size magnetic particles by employing LGMS technique.
trade-off between total exposure surface area and surface In order to perform a successful LGMS, magnetophoretic
reactivity of the given magnetic particles. In addition, con- force exerted on magnetic particles must be sufficiently intense
centration is also one of the major factors that influences to overcome viscous drag, thermal motion and gravitational
bio-compatibility of a magnetic particle solution. This is pulling. To accomplish these requirements, there are several
because magnetic particle solution which is highly concen- well-established rules of thumb which can provide a fast screen-
trated (beyond certain threshold concentration) was found ing of the suitability of any selected magnetic particle system in a
to be toxic to living organism [70]. The toxicity is mainly particular LGMS process. Here, dimensionless number analysis
due to the induction of oxidative stress and generation of (i.e. Reynolds number, Péclet number and Froude number)
reactive oxygen species which will subsequently injure or serves as a beneficial tool in determining whether the LGMS
kill the cells. For a cell –particle mixture, cell viability of a particular system can be successfully conducted [15].
decreases with increasing concentration of magnetic particles Another particle size-related criterion that also must
[71]. For instance, it was reported that the viability of PC12 be carefully addressed is the impact of particle-size
cells diminishes as they are exposed to a higher concentration distribution (PSD). As demonstrated in our previous work,
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

magnetization 13

rsfs.royalsocietypublishing.org
magnet

50 mT mm–1
exit
6
5
4
3
2
1
1 mm
12 mm
diameter cells

selectivity

Interface Focus 6: 20160048


10 mg ml–1
1 mg ml–1
600 0.5 mg ml–1
0.1 mg ml–1

S
DT2 (ms)

400

200

N
0
1 10 102 103 104 105 106 107
concentration of S. enterica (cfu ml–1) LGMS in biomedical MP250 MP30
diagnosis (1 min) (60 min)

sensitivity magnetic
separation rate
1.5
normalized opacity

10 g l–1 magnetic
1.0 1 g 1–1 particles
0.1 g 1–1
0.01 g 1–1 1 mm
0.5
100 nm 30 nm
0
0 100
time (s)
200
membrane of target cell
particle cooperative magnetophoresis magnetic loading particle
concentration size
Figure 6. Schematic diagram demonstrating the relationship between the three major factors ( particle size, particle concentration and magnetization) that influence
the performance of LGMS-aided biomedical diagnosis. Image components reprinted with permission from [13], Copyright & 2006 Royal Society of Chemistry, [22]
Copyright & 2015 American Chemical Society, [45] Copyright & 2008 American Institute of Physics and [48] Copyright & 2008 American Chemical Society.

magnetophoresis profile of an electrosterically stabilized internalize into) a unit target entity [13,18,55]. Hence, the magne-
magnetic nanoparticle suspension under LGMS is greatly tophoresis of this magnetic particle-bound target is a result of
influenced by PSD of the given particle system [58]. Prep- synergistic movement of a large number of magnetic particles.
aration of highly monodisperse magnetic particle system This process operated under the cooperative magnetophoresis
with narrow PSD is essential in order to have a high-quality mode in which magnetic particles move collectively towards
control of the LGMS process. Yet, the synthesis of mono- magnetic source. Additionally, the magnetic particle-bound tar-
disperse magnetic particles is technically challenging as gets might clump to each other during magnetophoretic
aggregation of the monodisperse particles, which results in migration which in turn further disturbs the dynamical behav-
polydisperse magnetic particles, is possible [74]. In addition, iour of the LGMS process. For this case, examining the
the aggregation of magnetic particles is further promoted by magnetophoretic migration of these magnetic particle-bound
the magnetic attraction due to the induced magnetic dipole targets at a microscopic level is highly recommended.
moment possessed by each magnetic particle in the presence Finally, our recent work has revealed that hydrodynamic
of an external magnetic field. For ferro- and ferri-magnetic effect is also playing a vital role in governing the dynamical
particles, magnetic dipole moment exists even without the behaviour of LGMS process. Owing to the momentum trans-
external magnetic field. Also, particle size is exceptionally fer between moving magnetic particles and the surrounding
vital in determining the bio-compatibility of the magne- fluid, continuous sweeping flow is created within the entire
tic particle system (as discussed in §4.2), and tight control of magnetic particle solution that is subjected to LGMS [44].
self-aggregation throughout the diagnosis process is indis- The convective flow generated due to the particle motion is
pensable. Thus, it is not only essential to synthesize highly directed towards the area where the magnetic field gradient
monodispersed magnetic particles, the PSD of the magnetic is lower. Simultaneously, this circulating flow drives mag-
particles throughout the diagnosis process must be closely netic particles located within the same area towards the
monitored and controlled as well. magnet within a relatively shorter timescale in comparison
Timescale involved for disease diagnosis is influenced by the with the motion which is purely driven by magnetophoretic
rate at which MS occurs. Therefore, it is crucial to optimize the force. In such a way, hydrodynamics engenders the fast mag-
separation rate of magnetic particles in any biomedical diagnos- netophoretic collection of magnetic particles from their
tic application. In particular, self-aggregation of magnetic suspension and makes LGMS more applicable for biomedical
particles leading to cooperative magnetophoresis is able to applications in which fast separation kinetics is needed. By
accelerate the LGMS rate. In fact, there were high amount taking advantage of the hydrodynamic interaction, it is
of magnetic particles in the cluster form, which bind to (or believed that the efficiency of diagnosis process could be
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

further enhanced. However, for LGMS that is conducted in framework in this aspect, such that the incorporation of 14
continuous mode, the magnetophoretic pathway of particles LGMS into a biomedical diagnostic application can be

rsfs.royalsocietypublishing.org
might be considerably disrupted by the convective flow facilitated in the upcoming future.
initiated from hydrodynamic effect. In this regard, the inter-
ference of hydrodynamically driven convection has caused
difficulty in the theoretical prediction of deterministic magne- Authors’ contributions. The manuscript was written through contributions
tophoretic pathway of magnetic particles subjected to LGMS. of all authors. All authors have given approval to the final version of
To date, we are only able to calculate the macroscopic the manuscript.
quantities of the LGMS system by solving the Navier – Competing interests. We declare we have no competing interests.
Stokes equation (which is a nonlinear and fourth-order Funding. This project is financially supported by (i) International Foun-
dation for Sciences (IFS) and co-financed by the Organisation for the
partial differential equation in three-dimensional space [75])
Prohibition of Chemical Weapons (OPCW) (grant no. 6050324/I100)
numerically with the aid of computational simulation tools.

Interface Focus 6: 20160048


and (ii) Fundamental Research Grant Scheme (FRGS) grant from
By contrast, theoretical description of the microscopic picture Ministry of Higher Education (MOHE) (grant no. 6071269).
of LGMS incorporating the hydrodynamic effect is still Acknowledgement. Sim Siong Leong acknowledges the support from the
unexplored. Thus, it is imperative to develop a theoretical MOHE Malaysia through the MyPhD Scholarship.

References
1. Lin J, Li M, Li Y, Chen Q. 2015 A high gradient and applications (ed. NT Thanh), pp. 195 –214. Boca 32, 9758–9765. (doi:10.1016/j.biomaterials.2011.
strength bioseparator with nano-sized Raton, FL: CRC press. 08.076)
immunomagnetic particles for specific separation 10. Xia N, Hunt TP, Mayers BT, Alsberg E, Whitesides 19. Bhakdi SC, Ottinger A, Somsri S, Sratongno P,
and efficient concentration of E. coli O157:H7. GM, Westervelt RM, Ingber DE. 2006 Combined Pannadaporn P, Chimma P, Malasit P,
J. Magn. Magn. Mater. 378, 206–213. (doi:10. microfluidic-micromagnetic separation of living cells Pattanapanyasat K, Neumann HP. 2010 Optimized
1016/j.jmmm.2014.11.039) in continuous flow. Biomed. Microdevices 8, high gradient magnetic separation for isolation of
2. He J, Huang M, Wang D, Zhang Z, Li G. 2014 299 –308. (doi:10.1007/s10544-006-0033-0) Plasmodium-infected red blood cells. Malar. J. 9,
Magnetic separation techniques in sample 11. Zborowski M, Williams PS, Moore LR, Chalmers JJ, 38. (doi:10.1186/1475-2875-9-38)
preparation for biological analysis: a review. Zimmerman PA. 2007 New challenges and 20. Qu B-Y, Wu Z-Y, Fang F, Bai Z-M, Yang D-Z, Xu S-K.
J. Pharmaceut. Biomed. Anal. 101, 84– 101. (doi:10. opportunities. In Laboratory techniques in 2008 A glass microfluidic chip for continuous blood
1016/j.jpba.2014.04.017) biochemistry and molecular biology (eds M cell sorting by a magnetic gradient without
3. Gijs MAM, Lacharme F, Lehmann U. 2010 Zborowski, JJ Chalmers), pp. 331–412. Amsterdam, labeling. Anal. Bioanal. Chem. 392, 1317–1324.
Microfluidic applications of magnetic The Netherlands: Elsevier. (doi:10.1007/s00216-008-2382-4)
particles for biological analysis and catalysis. 12. Nam J, Huang H, Lim H, Lim C, Shin S. 2013 21. Melville D, Paul F, Roath S. 1975 Direct magnetic
Chem. Rev. 110, 1518–1563. (doi:10.1021/ Magnetic separation of malaria-infected red blood separation of red cells from whole blood. Nature
cr9001929) cells in various developmental stages. Anal. Chem. 255, 706. (doi:10.1038/255706a0)
4. Lim J, Yeap SP, Leow CH, Toh PY, Low SC. 2014 85, 7316–7323. (doi:10.1021/ac4012057) 22. Chen Y, Xianyu Y, Wang Y, Zhang X, Cha R, Sun J,
Magnetophoresis of iron oxide nanoparticles at low 13. Pamme N, Wilhelm C. 2006 Continuous sorting of Jiang X. 2015 One-step detection of pathogens and
field gradient: the role of shape anisotropy. magnetic cells via on-chip free-flow viruses: combining magnetic relaxation switching
J. Colloid Interface Sci. 421, 170–177. (doi:10.1016/ magnetophoresis. Lab Chip 6, 974–980. (doi:10. and magnetic separation. ACS Nano 9, 3184–3191.
j.jcis.2014.01.044) 1039/b604542a) (doi:10.1021/acsnano.5b00240)
5. Andreu JS, Camacho J, Faraudo J, Benelmekki M, 14. Chen GD, Alberts CJ, Rodriguez W, Toner M. 2010 23. Olsvik O, Popovic T, Skjerve E, Cudjoe KS,
Rebollo C, Martı́nez LM. 2011 Simple analytical Concentration and purification of human Hornes E, Ugelstad J, Uhlen M. 1994 Magnetic
model for the magnetophoretic separation of immunodeficiency virus type 1 virions by separation techniques in diagnostic microbiology.
superparamagnetic dispersions in a uniform microfluidic separation of superparamagnetic Clin. Microbiol. Rev. 7, 43 –54. (doi:10.1128/CMR.
magnetic gradient. Phys. Rev. E 84, 021402. (doi:10. nanoparticles. Anal. Chem. 82, 723 –728. (doi:10. 7.1.43)
1103/PhysRevE.84.021402) 1021/ac9024522) 24. Lien K-Y, Lin J-L, Liu C-Y, Lei H-Y, Lee G-B. 2007
6. Pamme N. 2007 Continuous flow separations in 15. Lim J, Yeap SP, Low SC. 2014 Challenges associated Purification and enrichment of virus samples
microfluidic devices. Lab Chip 7, 1644– 1659. to magnetic separation of nanomaterials at low utilizing magnetic beads on a microfluidic system.
(doi:10.1039/b712784g) field gradient. Sep. Purif. Technol. 123, 171 –174. Lab Chip 7, 868–875. (doi:10.1039/b700516d)
7. Probst CE, Zrazhevskiy P, Gao X. 2011 Rapid (doi:10.1016/j.seppur.2013.12.038) 25. Gundersen SG, Haagensen I, Jonassen TO,
multitarget immunomagnetic separation through 16. Tangpukdee N, Duangdee C, Wilairatana P, Figenschau KJ, de Jonge N, Deelder AM. 1992
programmable DNA linker displacement. J. Am. Krudsood S. 2009 Malaria diagnosis: a brief review. Quantitative detection of schistosomal circulating
Chem. Soc. 133, 17 126–17 129. (doi:10.1021/ Korean J. Parasitol. 47, 93– 102. (doi:10.3347/kjp. anodic antigen by a magnetic bead antigen capture
ja2072324) 2009.47.2.93) enzyme-linked immunosorbent assay (MBAC-EIA)
8. Yavuz CT, Prakash A, Mayo JT, Colvin VL. 2009 17. Hackett S, Hamzah J, Davis TME, St Pierre TG. 2009 before and after mass chemotherapy. Trans. R. Soc.
Magnetic separations: from steel plants to Magnetic susceptibility of iron in malaria-infected Trop. Med. Hyg. 86, 175–178. (doi:10.1016/0035-
biotechnology. Chem. Eng. Sci. 64, 2510– 2521. red blood cells. Biochim. Biophys. Acta 1792, 9203(92)90559-U)
(doi:10.1016/j.ces.2008.11.018) 93 –99. (doi:10.1016/j.bbadis.2008.11.001) 26. Zborowski M. 2007 Commercial magnetic cell
9. Kaminski MD, Chen H, Liu X, Rempfer D, Rosengart 18. Xu H, Aguilar ZP, Yang L, Kuang M, Duan H, Xiong separation instruments and reagents. In Laboratory
AJ. 2012 Removal of blood-borne toxin in the body Y, Wei H, Wang A. 2011 Antibody conjugated techniques in biochemistry and molecular biology
using magnetic nanospheres. In Magnetic magnetic iron oxide nanoparticles for cancer cell (eds M Zborowski, JJ Chalmers), pp. 265– 292.
nanoparticles: from fabrication to clinical separation in fresh whole blood. Biomaterials Amsterdam, The Netherlands: Elsevier.
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

27. Šafařı́k I, Šafařı́ková M. 1999 Use of magnetic use of high performance magnetic nano- 57. Pivetal J, Royet D, Ciuta G, Frenea-Robin M, 15
techniques for the isolation of cells. J. Chromatogr. adsorbents in environmental applications. Haddour N, Dempsey NM, Dumas-Bouchiat F,

rsfs.royalsocietypublishing.org
B Biomed. Sci. Appl. 722, 33 –53. (doi:10.1016/ Chem. Eng. J. 256, 187–204. (doi:10.1016/j.cej. Simonet P. 2015 Micro-magnet arrays for specific
S0378-4347(98)00338-7) 2014.06.119) single bacterial cell positioning. J. Magn. Magn.
28. Moeser GD, Roach KA, Green WH, Alan Hatton T, 42. Yavuz CT et al. 2006 Low-field magnetic separation Mater. 380, 72 –77. (doi:10.1016/j.jmmm.2014.
Laibinis PE. 2004 High-gradient magnetic separation of monodisperse Fe3O4 nanocrystals. Science 314, 09.068)
of coated magnetic nanoparticles. AIChE J. 50, 964 –967. (doi: 10.1126/science.1131475) 58. Yeap SP, Leong SS, Ahmad AL, Ooi BS, Lim J. 2014
2835–2848. (doi:10.1002/aic.10270) 43. Lim J, Lanni C, Evarts ER, Lanni F, Tilton RD, On size fractionation of iron oxide nanoclusters by
29. Oberteuffer J. 1973 High gradient magnetic Majetich SA. 2011 Magnetophoresis of low magnetic field gradient. J. Phys. Chem C 118,
separation. IEEE Trans. Magn. 9, 303 –306. (doi:10. nanoparticles. ACS Nano 5, 217 –226. (doi:10.1021/ 24 042 –24 054. (doi:10.1021/jp504808v)
1109/tmag.1973.1067673) nn102383s) 59. Andreu JS, Camacho J, Faraudo J. 2011 Aggregation
30. Oder R. 1976 High gradient magnetic separation 44. Leong SS, Ahmad Z, Lim J. 2015 Magnetophoresis of superparamagnetic colloids in magnetic fields:

Interface Focus 6: 20160048


theory and applications. IEEE Trans. Magn. 12, of superparamagnetic nanoparticles at low field the quest for the equilibrium state. Soft Matter 7,
428–435. (doi:10.1109/tmag.1976.1059076) gradient: hydrodynamic effect. Soft Matter 11, 2336– 2339. (doi:10.1039/c0sm01424a)
31. Kelland D. 1973 High gradient magnetic 6968 –6980. (doi:10.1039/c5sm01422k) 60. Faraudo J, Andreu JS, Camacho J. 2013
separation applied to mineral beneficiation. 45. Schaller V et al. 2008 Motion of nanometer sized Understanding diluted dispersions of
IEEE Trans. Magn. 9, 307–310. (doi:10.1109/tmag. magnetic particles in a magnetic field gradient. superparamagnetic particles under strong magnetic
1973.1067683) J. Appl. Phys. 104, 093918. (doi:10.1063/1.3009686) fields: a review of concepts, theory and simulations.
32. Toh PY, Yeap SP, Kong LP, Ng BW, Chan DJC, Ahmad 46. McCabe WL, Smith JC, Harriott P. 2005 Unit Soft Matter 9, 6654 –6664. (doi:10.1039/
AL, Lim JK. 2012 Magnetophoretic removal of operations of chemical engineering. New York, NY: c3sm00132f )
microalgae from fishpond water: feasibility of high McGraw-Hill. 61. Koh I, Hong R, Weissleder R, Josephson L. 2009
gradient and low gradient magnetic separation. 47. Berg HC. 1993 Random walks in biology. Princeton, Nanoparticle2target interactions parallel
Chem. Eng. J. 211 –212, 22 –30. (doi:10.1016/j.cej. NJ: Princeton University Press. antibody2protein interactions. Anal. Chem. 81,
2012.09.051) 48. De Las Cuevas G, Faraudo J, Camacho J. 2008 Low- 3618– 3622. (doi:10.1021/ac802717c)
33. Franzreb M, Holl WH. 2000 Phosphate removal by gradient magnetophoresis through field-induced 62. Rosensweig RE. 1997 Ferrohydrodynamics.
high-gradient magnetic filtration using permanent reversible aggregation. J. Phys. Chem. C 112, New York, NY: Dover Publications.
magnets. IEEE Trans. Appl. Supercond. 10, 945 –950. (doi:10.1021/jp0755286) 63. Tamer U, Boyacı İH, Temur E, Zengin A, Dincer İ,
923–926. (doi:10.1109/77.828382) 49. Yellen BB, Hovorka O, Friedman G. 2005 Arranging Elerman Y. 2011 Fabrication of magnetic gold
34. Mariani G, Fabbri M, Negrini F, Ribani PL. 2010 matter by magnetic nanoparticle assemblers. Proc. nanorod particles for immunomagnetic separation
High-gradient magnetic separation of pollutant Natl Acad. Sci. USA 102, 8860 –8864. (doi:10.1073/ and SERS application. J. Nanopart. Res. 13,
from wastewaters using permanent magnets. Sep. pnas.0500409102) 3167– 3176. (doi:10.1007/s11051-010-0213-y)
Purif. Technol. 72, 147–155. (doi:10.1016/j.seppur. 50. Faraudo J, Camacho J. 2010 Cooperative 64. Tamer U, Cetin D, Suludere Z, Boyaci IH, Temiz HT,
2010.01.017) magnetophoresis of superparamagnetic colloids: Yegenoglu H, Daniel P, Dinçer İ, Elerman Y. 2013
35. Hoffmann C, Franzreb M, Holl WH. 2002 A novel theoretical aspects. Colloid Polym. Sci. 288, Gold-coated iron composite nanospheres targeted
high-gradient magnetic separator (HGMS) design 207 –215. (doi:10.1007/s00396-009-2107-z) the detection of Escherichia coli. Int. J. Mol. Sci. 14,
for biotech applications. IEEE Trans. Appl. Supercond. 51. Faraudo J, Andreu JS, Calero C, Camacho J. 2016 6223– 6240. (doi:10.3390/ijms14036223)
12, 963–966. (doi:10.1109/tasc.2002. Predicting the self-assembly of superparamagnetic 65. Sun W, Khosravi F, Albrechtsen H, Brovko LY,
1018560) colloids under magnetic fields. Adv. Funct. Mater. Griffiths MW. 2002 Comparison of ATP and in vivo
36. Ritter JA, Ebner AD, Daniel KD, Stewart KL. 2004 26, 3837–3858. (doi:10.1002/adfm.201504839) bioluminescence for assessing the efficiency of
Application of high gradient magnetic separation 52. Zborowski M, Ostera GR, Moore LR, Milliron S, immunomagnetic sorbents for live Escherichia coli
principles to magnetic drug targeting. J. Magn. Chalmers JJ, Schechter AN. 2003 Red blood cell O157:H7 cells. J. Appl. Microbiol. 92, 1021–1027.
Magn. Mater. 280, 184–201. (doi:10.1016/j.jmmm. magnetophoresis. Biophys. J. 84, 2638– 2645. (doi:10.1046/j.1365-2672.2002.01639.x)
2004.03.012) (doi:10.1016/S0006-3495(03)75069-3) 66. Kim GY, Josephson L, Langer R, Cima MJ. 2007
37. Forbes ZG, Yellen BB, Halverson DS, Fridman G, 53. Zimmerman PA, Thomson JM, Fujioka H, Collins WE, Magnetic relaxation switch detection of human
Barbee KA, Friedman G. 2008 Validation of high Zborowski M. 2006 Diagnosis of malaria by chorionic gonadotrophin. Bioconjugate Chem. 18,
gradient magnetic field based drug delivery to magnetic deposition microscopy. Am. J. Trop. Med. 2024– 2028. (doi:10.1021/bc070110w)
magnetizable implants under flow. IEEE Trans. Hyg. 74, 568 –572. 67. Nel A, Xia T, Mädler L, Li N. 2006 Toxic potential of
Biomed. Eng. 55, 643 –649. (doi:10.1109/tbme. 54. Song E-Q, Hu J, Wen C-Y, Tian Z-Q, Yu X, Zhang Z-L, materials at the nanolevel. Science 311, 622 –627.
2007.899347) Shi Y-B, Pang D-W. 2011 Fluorescent-magnetic- (doi:10.1126/science.1114397)
38. Stephens JR, Beveridge JS, Williams ME. biotargeting multifunctional nanobioprobes for 68. Kunzmann A et al. 2011 Efficient internalization of
2012 Analytical methods for separating and detecting and isolating multiple types of tumor silica-coated iron oxide nanoparticles of different
isolating magnetic nanoparticles. Phys. Chem. cells. ACS Nano 5, 761 –770. (doi:10.1021/ sizes by primary human macrophages and dendritic
Chem. Phys. 14, 3280–3289. (doi:10.1039/ nn1011336) cells. Toxicol. Appl. Pharmacol. 253, 81 –93. (doi:10.
c2cp22982j) 55. Robert D, Pamme N, Conjeaud H, Gazeau F, Iles A, 1016/j.taap.2011.03.011)
39. Ditsch A, Lindenmann S, Laibinis PE, Wang DIC, Wilhelm C. 2011 Cell sorting by endocytotic capacity 69. Karlsson HL, Gustafsson J, Cronholm P, Möller L.
Hatton TA. 2005 High-gradient magnetic separation in a microfluidic magnetophoresis device. Lab Chip 2009 Size-dependent toxicity of metal oxide
of magnetic nanoclusters. Ind. Eng. Chem. Res. 44, 11, 1902–1910. (doi:10.1039/c0lc00656d) particles—a comparison between nano- and
6824–6836. (doi:10.1021/ie048841s) 56. Moore LR, Williams PS, Nehl F, Abe K, Chalmers JJ, micrometer size. Toxicol. Lett. 188, 112 –118.
40. Gerber R, Birss RR. 1983 High gradient magnetic Zborowski M. 2013 Feasibility study of red (doi:10.1016/j.toxlet.2009.03.014)
separation, vol. 209. Research Studies Press Div. of blood cell debulking by magnetic field-flow 70. Häfeli UO, Riffle JS, Harris-Shekhawat L, Carmichael-
John Wiley & Sons, Ltd. fractionation with step-programmed flow. Anal. Baranauskas A, Mark F, Dailey JP, Bardenstein D.
41. Gómez-Pastora J, Bringas E, Ortiz I. 2014 Bioanal. Chem. 406, 1661 –1670. (doi:10.1007/ 2009 Cell uptake and in vitro toxicity of magnetic
Recent progress and future challenges on the s00216-013-7394-z) nanoparticles suitable for drug delivery. Mol.
Downloaded from http://rsfs.royalsocietypublishing.org/ on July 7, 2018

Pharmaceut. 6, 1417–1428. (doi:10.1021/ internalization in growing neurons. Biomaterials 28, 74. Costo R, Bello V, González-Carreño T, Veintemillas- 16
mp900083m) 2572 –2581. (doi:10.1016/j.biomaterials.2007.01. Verdaguer S, Wang D. 2013 Size sorting of

rsfs.royalsocietypublishing.org
71. Dobson J. 2006 Gene therapy progress and 043) ultrasmall magnetic nanoparticles and their
prospects: magnetic nanoparticle-based gene 73. Yeap SP, Toh PY, Ahmad AL, Low SC, Majetich SA, aggregates behaviour. Mater. Res. Bull. 48, 4294–
delivery. Gene Therapy 13, 283– 287. (doi:10.1038/ Lim J. 2012 Colloidal stability and magnetophoresis 4300. (doi:10.1016/j.materresbull.2013.06.050)
sj.gt.3302720) of gold-coated iron oxide nanorods in biological 75. Bird RB, Stewart WE, Lightfoot EN. 2006 Transport
72. Pisanic Ii TR, Blackwell JD, Shubayev VI, Fiñones RR, media. J. Phys. Chem. C 116, 22 561– 22 569. phenomena, 2nd edn. New York, NY: John Wiley &
Jin S. 2007 Nanotoxicity of iron oxide nanoparticle (doi:10.1021/jp306159a) Sons, Inc.

Interface Focus 6: 20160048

Vous aimerez peut-être aussi