ArticlePDF Available

Magnetoferritin nanoparticles for targeting and visualizing tumour tissues

Authors:

Abstract and Figures

Engineered nanoparticles have been used to provide diagnostic, therapeutic and prognostic information about the status of disease. Nanoparticles developed for these purposes are typically modified with targeting ligands (such as antibodies, peptides or small molecules) or contrast agents using complicated processes and expensive reagents. Moreover, this approach can lead to an excess of ligands on the nanoparticle surface, and this causes non-specific binding and aggregation of nanoparticles, which decreases detection sensitivity. Here, we show that magnetoferritin nanoparticles (M-HFn) can be used to target and visualize tumour tissues without the use of any targeting ligands or contrast agents. Iron oxide nanoparticles are encapsulated inside a recombinant human heavy-chain ferritin (HFn) protein shell, which binds to tumour cells that overexpress transferrin receptor 1 (TfR1). The iron oxide core catalyses the oxidation of peroxidase substrates in the presence of hydrogen peroxide to produce a colour reaction that is used to visualize tumour tissues. We examined 474 clinical specimens from patients with nine types of cancer and verified that these nanoparticles can distinguish cancerous cells from normal cells with a sensitivity of 98% and specificity of 95%.
Content may be subject to copyright.
Magnetoferritin nanoparticles for targeting and
visualizing tumour tissues
Kelong Fan
1
, Changqian Cao
2
, Yongxin Pan
2
,DiLu
1
,DonglingYang
1
, Jing Feng
1
,LinaSong
1
,
Minmin Liang
1
*
and Xiyun Yan
1
*
Engineered nanoparticles have been used to provide diagnos-
tic
1–3
, therapeutic
4,5
and prognostic information
6,7
about the
status of disease. Nanoparticles developed for these purposes
are typically modified with targeting ligands (such as anti-
bodies
8–10
, peptides
11,12
or small molecules
13
) or contrast
agents
14–16
using complicated processes and expensive
reagents. Moreover, this approach can lead to an excess of
ligands on the nanoparticle surface, and this causes non-
specific binding
17–20
and aggregation of nanoparticles
18–20
,
which decreases detection sensitivity
17–20
. Here, we show that
magnetoferritin nanoparticles (M-HFn) can be used to target
and visualize tumour tissues without the use of any targeting
ligands or contrast agents. Iron oxide nanoparticles are encap-
sulated inside a recombinant human heavy-chain ferritin (HFn)
protein shell, which binds to tumour cells that overexpress
transferrin receptor 1 (TfR1). The iron oxide core catalyses
the oxidation of peroxidase substrates in the presence of hydro-
gen peroxide to produce a colour reaction that is used to visu-
alize tumour tissues. We examined 474 clinical specimens from
patients with nine types of cancer and verified that these nano-
particles can distinguish cancerous cells from normal cells with
a sensitivity of 98% and specificity of 95%.
Ferritin is an iron storage protein composed of 24 subunits made
up of the heavy-chain ferritin (HFn) and the light-chain ferritin.
Ferritin is spherical, with an outer diameter of 12 nm and interior
cavity diameter of 8 nm (ref. 21). The cavity has been used as a reac-
tion chamber to synthesize highly crystalline and monodisperse
nanoparticles through biomimetic mineralization within the
protein shell
22–24
. Recently, it was shown that HFn binds to
human cells via transferrin receptor 1 (TfR1)
25
. Because TfR1 is
overexpressed in tumour cells, this receptor has been used as a tar-
geting marker for tumour diagnosis and therapy
26–29
.
Current HFn-based tumour detection methods rely on functiona-
lization of HFn with recognition ligands
30,31
and signal molecules
31,32
.
Previously, we have shown that iron oxide nanoparticles can catalyse
the oxidation of peroxidase substrates in the presence of hydrogen
peroxide to produce a colour reaction similar to that of natural per-
oxidases
33
. We hypothesize that magnetoferritin (M-HFn) nanopar-
ticles generated by encapsulating iron oxide nanoparticles inside a
HFn shell should be able to target TfR1 without any additional rec-
ognition ligands on their surface, and visualize tumour tissues
through the peroxidase activity of the iron oxide core (Fig. 1a).
We expressed recombinant human HFn in Escherichia coli. After
purification, the proteins were analysed by transmission electron
microscopy (TEM). The HFn protein shells had a well-defined mor-
phology and were monodisperse in size (Fig. 1b,c). After iron
loading and oxidation, a well-defined iron oxide core with an
average diameter of 4.7 nm was synthesized within the HFn
protein shell (Fig. 1d,e). The iron mineral core of M-HFn is com-
posed of magnetite or maghemite, as characterized in our previous
publication
34
. Cryoelectron transmission microscopy (cryoTEM)
analysis showed that the mineral cores were clearly encapsulated
within the HFn protein shells (Fig. 1b). Dynamic light scattering
(DLS) and size-exclusion chromatography (SEC) results further
confirmed that the M-HFn nanoparticles were monodispersed
with an outer diameter of 12–16 nm (Supplementary Fig. S1a),
and the iron loading did not significantly perturb the overall
protein cage architecture of HFn (Supplementary Fig. S1b,c).
M-HFn nanoparticles catalyses the oxidation of peroxidase sub-
strates 3,3,5,5-tetramethylbenzidine (TMB) and di-azo-aminoben-
zene (DAB) in the presence of H
2
O
2
to give a blue colour
(Fig. 1f ) and brown colour (Fig. 1g), respectively, confirming that
M-HFn nanoparticles have peroxidase activity towards typical per-
oxidase substrates. The mineral phase composition of the iron core
determines the peroxidase activity of the M-HFn, as is evident from
Supplementary Fig. S2. M-HFn with mineral cores consisting of
magnetite or maghemite
34
exhibited a much higher peroxidase
activity when compared with natural holoferritin. The natural
cores in holoferritin consist mainly of the hydrated iron oxide
mineral ferrihydrite (5Fe
2
O
3
.
9H
2
O)
35
, which exhibits little peroxi-
dase activity. Apoferritin, without a mineral core, exhibited no
peroxidase activity.
The specificity of HFn binding to living cancer cells was investi-
gated using human breast, colon and liver cancer cell lines and
their corresponding xenograft tumours. HT-29 human colon
cancer cells and SMMC-7721 human liver cancer cells express TfR1
at high levels, but MX-1 human breast cancer cells do not express
this receptor (Fig. 2a). HFn bound to TfR1-positive HT-29 and
SMMC-7721 cells, and their xenograft tumours, but not to TfR1-
negative MX-1 or its xenograft tumour (Fig. 2b,c). The binding of
HFn to TfR1-positive cells was saturable, and could be inhibited by
adding an excess of unconjugated HFn (Supplementary Fig. S3a),
showing that HFn binding is specific. The saturation binding curve
and Scatchard analysis demonstrate that the K
d
value for HFn is
50 nM (Supplementary Fig. S3b), indicating that HFn has a high affi-
nity for TfR1. In addition, HFn showed significant binding to A375
melanoma cells, MDA-MB-231 breast cancer cells, K562 erythroleu-
kemia cells, HeLa cervical cancer cells, SKOV-3 ovarian cancer cells,
PC-3 prostate cancer cells, U251 glioblastoma cells, U937 histiocytic
lymphoma cells, SW1990 pancreatic cancer cells and Jurkat T-cell
leukemia cells (Supplementary Fig. S4), indicating that HFn has a
universal capability for recognizing cancer cells.
1
Key Laboratory of Protein and Peptide Pharmaceutical, National Laboratory of Biomacromolecules, CAS-University of Tokyo Joint Laboratory of Structural
Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China,
2
Paleomagnetism and
Geochronology Laboratory, Key Laboratory of the Earth’s Deep Interior, Institute of Geology and Geophysics, Chinese Academy of Sciences,
Beijing 100029, China.
*
e-mail: mmliang@moon.ibp.ac.cn; yanxy@sun5.ibp.ac.cn
LETTERS
PUBLISHED ONLINE: 17 JUNE 2012 | DOI: 10.1038/NNANO.2012.90
NATURE NANOTECHNOLOGY | VOL 7 | JULY 2012 | www.nature.com/naturenanotechnology 459
© 2012 Macmillan Publishers Limited. All rights reserved.
To investigate whether TfR1 mediates the specific binding of
HFn to cancer cells, TfR1 was immunoprecipitated from HT-29
cell lysates. HFn reacted with the precipitated TfR1 (Fig. 2d), indi-
cating that HFn binds to cancer cells via TfR1. The next far-
Western blotting analysis showed that HFn bound to TfR1 in
HT-29 and SMMC-7721 cells, but not in MX-1 cells (Fig. 2e), con-
sistent with their TfR1 expression patterns (Fig. 2a) and confirming
that TfR1 is the receptor of HFn and mediates its specific binding to
cancer cells. Further flow cytometry and confocal results showed
that anti-TfR1 monoclonal antibody (mAb) could completely
inhibit the binding of FITC-labelled HFn to TfR1 on SMMC-
7721 cancer cells (Supplementary Fig. S5), indicating that TfR1 is
the only receptor for HFn present on these cancer cells. In addition,
although transferrin competed with HFn for binding to TfR1, excess
transferrin only partially inhibited HFn binding (Supplementary
Fig. S6), consistent with observations reported by Li and
Seaman
25
. These results suggest that HFn and transferrin share
receptor TfR1, but may bind to different epitopes on TfR1.
To establish the validity of the M-HFn nanoparticle-based cancer
diagnostic method, we carried out the following histological staining
experiments in xenograft tumours. FITC-conjugated HFn showed
strong fluorescence staining in HT-29, SKOV-3 and SMMC-7721
xenograft tumours (Fig. 3, top row), confirming the tumour-
binding reactivity of the HFn protein. After iron loading and
oxidation, M-HFn nanoparticles displayed an intensive brown per-
oxidase activity that visualized the tumour cells after adding DAB
substrate and H
2
O
2
(Fig. 3, middle row), verifying the feasibility
of our M-HFn nanoparticle-based cancer diagnostic method. The
fluorescence staining co-localized with mineral-peroxidase staining
in tumour cells (Supplementary Fig. S7), indicating that iron loading
and fluorescence labelling do not affect the tumour-binding activity
of the HFn protein. This again shows the feasibility of our M-HFn-
based assay for tumour detection.
Traditional immunohistochemical staining using anti-TfR1
antibodies (Abs) was next performed to compare its tumour-
binding specificity and staining quality with our M-HFn nanoparti-
cle-based method in xenograft tumour tissues. The intensity and the
pattern of M-HFn nanoparticle-based staining were almost the
same as that of immunohistochemical staining (Fig. 3, bottom
row), demonstrating the accuracy of tumour detection by the M-
HFn nanoparticles. TfR1-negative MX-1 tumour xenograft tissues
consistently showed negative staining for M-HFn nanoparticles,
FITC-conjugated HFn and TfR1 Abs (Fig. 3, right column),
further confirming that HFn targets tumour cells via TfR1.
To evaluate the potential clinical application of M-HFn nanopar-
ticles as a diagnostic agent for tumours in tissue specimens, we
screened 247 clinical tumour tissue samples and 227 corresponding
normal tissue samples by histological staining. Staining was con-
sidered positive when 10% or more of the tumour cells were
stained (cutoff, 10%). M-HFn did not stain, or only slightly
stained, normal or lesion tissues, with a staining frequency of only
4.8% (11/227, Table 1, Fig. 4). In tumour tissues, M-HFn
c
d
b
TMB
2
0
40
80
120
160
200
3456
n
= 700
4.7
± 0.8 nm
78
Diameter (nm)
Number of particles
TMB/H
2
O
2
/M-HFn
f
g
DAB DAB/H
2
O
2
/M-HFn
e
20 nm
20 nm
20 nm
a
HFn
protein shell
H
2
O
2
Fe
2+
Oxidation
M-HFn
nanoparticle
Iron
core
Figure 1 | Preparation and characterization of M-HFn nanoparticles. a, Schematic showing the preparation of M-HFn nanoparticles and their structure.
b, CryoTEM image of M-HFn nanoparticles. c,d, TEM images of HFn protein shells (c) and iron oxide cores (d). HF n pr otein shells wer e negativ ely stained
with uranyl acetate for TEM observations and iron oxide cores in HFn were unstained. e, Size distribution of iron oxide cores, with a median diameter of
4.7+0.8 nm. f,g, Char a cterization of peroxidase activity of M-HF n nanoparticles. M-HF n cataly sed the oxidation of peroxidase substrates TMB (f)andDAB
(g) in the presence of H
2
O
2
to give a coloured product.
LETTERS
NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2012.90
NATURE NANOTECHNOLOGY | VOL 7 | JULY 2012 | www.nature.com/naturenanotechnology460
© 2012 Macmillan Publishers Limited. All rights reserved.
nanoparticles strongly stained tumour cells, and a clear distinction
was seen between cancerous cells and adjacent normal cells in repre-
sentative sections (Fig. 4). M-HFn staining showed a sensitivity of
98% across nine types of cancer represented by 247 tumour
tissues (Table 1). These clinical tissue specimens were further
stained with FITC-conjugated HFn protein shells. As expected, con-
sistent staining patterns were observed (Fig. 4), verifying the diagno-
sis based on M-HFn nanoparticles. Importantly, HFn showed a
distinct staining reaction in different grades and growth patterns
of hepatocellular carcinoma, lung squamous cell carcinoma, cervical
squamous cell carcinoma, prostate adenocarcinoma, ovarian serous
papillary carcinoma and colonic adenocarcinoma (Supplementary
Fig. S8–S13), demonstrating that HFn has an impressive ability to
discriminate tumour cells from normal cells, and thus has clinical
potential in cancer diagnosis.
To understand the mechanism of the M-HFn-based peroxidase-
like reaction, the formation of OH
during the reaction was
measured using electron spin resonance. OH
was produced
during the peroxidase-like reaction in the presence of both
M-HFn nanoparticles and H
2
O
2
(Supplementary Fig. S14a,b).
With the addition of an OH
scavenger (ethanol), the formed
OH
disappeared (Supplementary Fig. S14 c) and the peroxidase
activity of the M-HFn nanoparticles decreased to 20% of the original
activity (Supplementary Fig. S14d,e), indicating that the OH
formed during the peroxidase-like reaction is responsible for the
catalytic oxidation of peroxidase substrate that gives the coloured
precipitate at the site of its target.
Based on these results, we propose the following reaction mech-
anism. With the addition of H
2
O
2
and peroxidase substrate to the
M-HFn reaction solution, H
2
O
2
diffuses into the ferritin cavity
through its hydrophilic channels and interacts with the iron oxide
core of M-HFn to generate OH
on the surface of the iron core.
The generated OH
then oxidizes nearby peroxidase substrates
(for example, DAB) to form an insoluble coloured precipitate at
the site of the M-HFn, which is targeted to cancer cells. The
coloured precipitates are only formed at the site of the M-HFn
because OH
radicals are highly reactive and short-lived, and can
only oxidize nearby substrates. The clear boundary between
tumour and normal tissues on M-HFn-stained tissue slides
(Supplementary Fig. S15) also demonstrates that the coloured pre-
cipitates are generated right at the site of the M-HFn-targeted
cancer cells and do not diffuse away from their targets.
Achieving rapid, low-cost and sensitive cancer diagnosis remains
a challenge due to the complexities of this disease. Our studies show
that one-step tumour targeting and visualization with low-cost and
mass-produced M-HFn nanoparticles is feasible for convenient and
sensitive monitoring and analysis of tumour cells in tissue speci-
mens. The recombinant human HFn protein shell has tumour-
specific binding properties, and the encapsulated iron oxide core
has strong peroxidase activity, which allows us to combine effective
tumour cell recognition in clinical tissues with highly sensitive stain-
ing of the targeted cells. Staining results for 247 clinical tumour
tissue samples from patients with ovarian, liver, prostate, lung,
breast, cervical, thymus colorectal or oesophageal cancers, as well
as for 227 normal and lesion tissue control samples (Table 1,
Fig. 4), clearly demonstrate the capacity of M-HFn nanoparticles
to distinguish cancer cells from normal ones in tissue specimens.
Compared with conventional antibody-based histological
methods for cancer detection in clinics, our novel M-HFn nanopar-
ticle-based method has the following advantages. First, it has high
a
TfR1
β-actin
IP mIgGα-TfR1
TfR1
HFn
WB
d
c
b
Counts
HT-29
Counts
SMMC-7721
SMMC-7721
HT-29
MX-1
SMMC-7721
HT-29
MX-1
Counts
MX-1
150
120
90
60
30
0
10
0
150
120
90
60
30
0
10
1
10
2
10
3
10
4
10
0
10
1
10
2
10
3
10
4
150
120
90
60
30
0
10
0
10
1
10
2
10
3
10
4
MX-1
HT-29 SMMC-7721
Fluorescence intensity Fluorescence intensity
Fluorescence intensity
95 kDa
17
kDa
55
kDa
43 kDa
130 kDa
170
kDa
72
kDa
e
TfR1
Figure 2 | HFn binds specifically to TfR1 in cancer cells. a, Western blot of TfR1 expression in HT-29 colon cancer cells, SMMC-7721 liver cancer cells and
MX-1 breast cancer cells. b-actin was used as a loading control. b, Flow cytometric analysis of the specific binding of HFn to HT-29, SMMC-7721 and MX-1
cancer cells. c, Fluorescence staining of HT -29, SMMC-7721 and MX -1 xenogr a ft tumours incubated with FITC-conjugated HFn (scale bars, 50
m
m). d,TfR1
immunoprecipitated from HT-29 cell lysates by anti-TfR1 mAbs (a-TfR1) was recognized by HFn, detected by mouse anti-HFn mAbs and visualized with
HRP-coupled anti–mouse IgG. e, F ar -Western blotting of HT-29, SMMC-7721 and MX-1 cell line lysates was performed using HF n, detected by mouse
anti-HFn mAbs and visualized with HRP-coupled anti-mouse IgG. T fR1 repr esented an 95 kDa band.
NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2012.90
LETTERS
NATURE NANOTECHNOLOGY | VOL 7 | JULY 2012 | www.nature.com/naturenanotechnology 461
© 2012 Macmillan Publishers Limited. All rights reserved.
sensitivity and specificity. Screening 474 clinical specimens from
patients with nine types of cancer shows that the M-HFn nanopar-
ticle-based method has a sensitivity of 98% and a specificity of 95%
(Table 1), which is much better than most antibody-based
histological detection methods reported in the literature
(Supplementary Table S1). In addition, a side-by-side comparison
of the standard antibody-based immunohistochemistry and our
M-HFn-based approach in two hepatocellular carcinoma cases ident-
ified by pathologists also shows that our M-HFn-based approach
performs much better than anti-TfR1 antibody-based immunohisto-
chemistry when distinguishing tumours from normal tissues
(Supplementary Fig. S16). The second advantage of our method is
its high accuracy, credibility and repeatability. Traditional immuno-
histochemistry involves multiple manipulation steps. The results are
easily affected by the proficiency and subjectivity of the manipulators.
By using a one-step incubation of one reagent in our M-HFn nano-
particle-based method, the accuracy, credibility and repeatability of
the results are improved greatly. Our method also has the advantage
of a rapid examination time, taking 1 h, rather than the 4 h required
for immunohistochemistry, which generally involves multistep incu-
bation of primary antibody, secondary antibody or enzyme-labelled
third antibody. Finally, it is low in cost, avoiding the use of expensive
and unstable antibodies, and the HFn can be produced in Escherichia
coli at high yield. M-HFn nanoparticles can be low in cost and mass-
produced by simply oxidizing Fe
2þ
within HFn by H
2
O
2
.
M-HFn-based peroxidase staining and FITC-HFn-based fluor-
escence staining target tumour cells via the same HFn protein, but
visualize them using chromogenic and fluorescence signals, respect-
ively. Peroxidase staining is clearly better for clinical diagnostics
because it is compatible with haematoxylin counterstains, which
allows visualization of the context of a tumour’s expression
pattern and provides more detailed histopathological information,
allowing diagnostic features to be easily discerned. Fluorescence
staining, as with most fluorescence techniques, has two significant
FITC-conjugated
HFn
HT-29 SKOV-3
M-HFn
nanoparticles
Anti-TfR1 Abs
SMMC-7721 MX-1
Figure 3 | M-HFn nanoparticle staining of tumour tissues. FITC -conjugated HF n-based fluorescence staining (top row), M-HFn nanoparticle-based
peroxidase staining (middle row) and anti-T fR1 Abs-based immunohistochemical staining (bottom row) of paraffin-embedded HT-29 colon cancer, SK OV-3
ovarian cancer, SMMC-7721 liver cancer and MX-1 breast cancer xenograft tumours. TfR1-positive xenograft tumours sho w ed str ong positive staining for
FITC-conjugated HFn (green fluorescence), M-HF n nanoparticles (brown) and anti-TfR1 Abs (bro wn), whereas T fR1-nega tiv e xenogr aft tumours show ed no
staining for FITC-conjugated HFn, M-HFn nanoparticles and anti-TfR1 Abs (scale bars, 100
m
m).Abs,antibodies.
Table 1 | Histological analysis of M-HFn nanoparticle staining
of tumours in clinical tissue specimens.
Tumour tissues Positive/cases
(sensitivity)
Normal
tissues
Negative/cases
(specificity)
Hepatocellular
carcinoma
54/55 (98%) Liver 39/45 (87%)
Lung squamous
cell carcinoma
50/52 (96%) Lung 55/56 (98%)
Colonic
adenocarcinoma
23/23 (100%) Colon 40/41 (98%)
Cervical
squamous cell
carcinoma
28/28 (100%) Cervix 52/52 (100%)
Prostate
adenocarcinoma
22/22 (100%) Prostate 14/16 (88%)
Ovarian serous
papillary
carcinoma
55/56 (98%) Ovary 4/4(100%)
Breast ductal
carcinoma
3/4 (75%) Breast 4/4(75%)
Thymic carcinoma 4/4 (100%) Thymus 4/4(100%)
Oesophagus
squamous cell
carcinoma
2/3 (67%) Oesophagus 5/5(100%)
To t a l 2 4 1 /247 (98%) Total 216/227 (95%)
LETTERS
NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2012.90
NATURE NANOTECHNOLOGY | VOL 7 | JULY 2012 | www.nature.com/naturenanotechnology462
© 2012 Macmillan Publishers Limited. All rights reserved.
problems—photobleaching and autofluorescence—which severely
limits the detection sensitivity of FITC-HFn-based fluorescence
staining. This study suggests that the easily synthesized M-HFn
nanoparticles have the potential to become a diagnostic tool for
rapid, low-cost and universal assessment of cell cancerization.
Methods
Conjugation of HFn and M-HFn. HFn and M-HFn were labelled by fluorescein
isothiocyanate (FITC, Sigma) using the following procedure. FITC was dissolved in
dry dimethylformamide and added to an HFn or M-HFn solution in 0.1 M
NaHCO
3
, pH 8.5, at an FITC to HFn or M-HFn molar ratio of 10:1. The reaction
solution was gently stirred for 3 h at room temperature in the dark and then purified
on a polyacrylamide column (Thermo Scientific, MWCO 6000) using 0.1 M PBS,
pH 7.5, as an eluant to remove free dyes. The labelled FITC concentration was
determined by measuring the absorbance at 492 nm, and the HFn or M-HFn
concentration was determined using a BCA protein assay reagent kit.
Cell bind ing studies. The reactivity of HFn with cancer cell lines was assessed by
flow cytometry. Briefly, 100 ml detached cell suspensions (1 × 10
6
cells per ml) were
stained with 20 mgml
21
of FITC-conjugated HFn for 2 h at 4 8C in PBS containing
0.3% bovine serum albumin. After three washes in cold PBS, cells were analysed
immediately using a FACSCalibur flow cytometry system (Becton Dickinson).
Western blotting, far-Western blotting and immunopreciptation. TfR1
expression was assessed by Western blotting. Cell lysates of each type were run on a
10% SDS–polyacrylamide gel and transferred to a nitrocellulose membrane blocked
with 5% non-fat milk, 0.1% Tween 20 in PBS for 30 min, and then incubated
overnight at 4 8C with a 1:2,000 dilution of mouse anti-human TfR1 monoclonal
antibody (mAbs, BD Bioscience). The TfR1 mAbs was detected using a 1:6,000
dilution of goat anti-mouse IgG conjugated to HRP (Pierce), and developed with
ECL substrate (Pierce).
Immunopreciptation was performed to confirm that TfR1 is the binding
receptor of HFn. Cell lysates were pre-cleared by incubation with 15 ml protein A/G
agarose (Santa Cruz Biotech), centrifuged to remove the beads, and then incubated
at 4 8C overnight with 4 ml of 0.5 mg ml
21
mouse anti-human TfR1 mAbs or 1 mlof
2mgml
21
normal mouse IgG (Sigma) as a control, followed by incubation with
15 ml protein A/G agarose for 1 h at room temperature. The precipitated complexes
were boiled for 15 min, analysed by 10% SDS–polyacrylamide gel and then
transferred to a nitrocellulose membrane. After blocking with non-fat dry milk, the
nitrocellulose membranes were probed with a 1:2,000 dilution of anti-TfR1 mAbs or
with 3.8 mgml
21
HFn protein followed by a 1:2,000 dilution of mouse anti-HFn
mAbs, and developed with HRP-conjugated anti-mouse IgG.
Far-Western blotting was performed to analyse the binding pattern of HFn to
cancer cells. Lysates from the HT-29, SMMC-7721 and MX-1 cell lines were run on a
10% SDS–polyacrylamide gel and transferred to a nitrocellulose membrane, blocked
in non-fat dry milk for 30 min, and then incubated overnight at 4 8C with HFn
(3.8 mgml
21
). HFn was detected using a 1:2,000 dilution of mouse anti-HFn mAbs
(Santa Cruz Biotech), and developed with HRP-conjugated anti-mouse IgG.
Staining of tumour xenografts and clinical specimens. Paraffin-embedded tissue
sections were deparaffinized by washing twice in xylene for 10 min and then
hydrated progressively using an ethanol gradient. Endogenous peroxidase activity
was quenched by incubation with 0.3% H
2
O
2
in methanol for 30 min. After rinsing,
the tissue sections were boiled in 10 mM citrate buffer (pH 6.0) at 100 8 C for 30 min,
cooled to room temperature, blocked with 5% goat serum in PBS for 1 h at 37 8C,
washed and then incubated with M-HFn nanoparticles (1.8 mM) for 1 h at 37 8C,
and then rinsed in PBS. Freshly prepared DAB was added for colour development.
All samples were counterstained with haematoxylin (blue stain). The stained
sections were analysed under a microscope and the results were expressed in terms of
the percentage of stained tumour cells (0 to 100%). If 10% or more of the tumour
cells were stained, the slide was scored as positive. Two independent pathologists
who were blind to all clinical information scored all specimens.
Fluorescence staining of tissue sections was performed to confirm the binding
specificity of HFn to cells. After blocking in serum, tissue sections were incubated
with FITC-conjugated HFn (1 mM) at 4 8C overnight. The stained tissues were
examined under a confocal laser scanning microscope (Olympus).
Immunohistochemical staining of tissue sections by anti-TfR1 Abs was
performed to compare tumour-binding specificity and staining quality with M-HFn
nanoparticles. Briefly, after blocking in serum, tissue sections were incubated at 4 8C
overnight with a 1:300 dilution of polyclonal rabbit anti-TfR1 antibody (Abcam).
The bound antibody was detected by incubating the tissues with a 1:1,000 dilution of
biotinylated anti-rabbit antibody (Santa Cruz Biotech) at 37 8C for 1 h, then with a
1:200 dilution of HRP-conjugated streptavidin (Pierce) for 40 min.
Received 27 February 2012; accepted 7 May 2012;
published online 17 June 2012
References
1. Galanzha, E. I. et al. In vivo magnetic enrichment and multiplex photoacoustic
detection of circulating tumour cells. Nature Nanotech. 4, 855–860 (2009).
2. Winter, P. M. et al. Molecular imaging of angiogenesis in nascent Vx-2 rabbit
tumors using a novel a
n
b
3
-targeted nanoparticle and 1.5 tesla magnetic
resonance imaging. Cancer Res. 63, 5838–5843 (2003).
3. Stoeva, S. I., Lee, J. S., Smith, J. E., Rosen, S. T. & Mirkin, C. A. Multiplexed
detection of protein cancer markers with biobarcoded nanoparticle probes.
J. Am. Chem. Soc. 128, 8378–8379 (2006).
4. Janib, S. M., Moses, A. S. & Mackay, J. A. Imaging and drug delivery using
theranostic nanoparticles. Adv. Drug Del. Rev. 3, 1–2 (2010).
5. Zrazhevskiy, P., Sena, M. & Gao, X. H. Designing multifunctional quantum dots
for bioimaging, detection, and drug delivery. Chem. Soc. Rev. 3, 1–2 (2010).
6. Li, R. L. et al. Prognostic value of Akt-1 in human prostate cancer: a
computerized quantitative assessment with quantum dot technology. Clin.
Cancer Res. 15, 3568–3573 (2009).
Liver Colon Breast Ovary Prostate Lung Oesophagus Cervix
FITC-conjugated
HFn
M-HFn
nanoparticles
Tumour tissue
Non-tumour
tissue
Tumour tissue
Non-tumour
tissue
Figure 4 | Cancer diagnosis in clinical specimens using M-HFn nanoparticles. P ara ffin-embedded clinical tumour tissues, and their corr esponding normal
and lesion tissues, were stained by FITC-conjugated HF n protein shells and M-HFn nanoparticles. Tumour tissues showed strong positive staining for M-HFn
nanoparticles (brown) and FITC-conjuga ted HF n protein shells (green fluorescence), whereas the normal and lesion tissue controls were negativ e for M-HFn
nanoparticles and FITC-conjugated HF n. Scale bars, 100
m
m.
NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2012.90
LETTERS
NATURE NANOTECHNOLOGY | VOL 7 | JULY 2012 | www.nature.com/naturenanotechnology 463
© 2012 Macmillan Publishers Limited. All rights reserved.
7. Karathanasis, E. et al. Multifunctional nanocarriers for mammographic
quantification of tumor dosing and prognosis of breast cancer therapy.
Biomaterials 29, 4815–4822 (2008).
8. Jaetao, J. E. et al. Enhanced leukemia cell detection using a novel magnetic
needle and nanoparticles. Cancer Res. 69, 8310–8316 (2009).
9. Corsi, F. et al. HER2 expression in breast cancer cells is downregulated upon
active targeting by antibody-engineered multifunctional nanoparticles in mice.
ACS Nano 5, 6383–6393 (2011).
10. Xu, H. Y. et al. Antibody conjugated magnetic iron oxide nanoparticles for
cancer cell separation in fresh whole blood. Biomaterials 32, 9758–9765 (2011).
11. Park, Y. et al. A new atherosclerotic lesion probe based on hydrophobically
modified chitosan nanoparticles functionalized by the atherosclerotic plaque
targeted peptides. J. Control. Rel. 128, 217–223 (2008).
12. Kumar, A. et al. Gold nanoparticles functionalized with therapeutic and targeted
peptides for cancer treatment. Biomaterials 33, 1180–1189 (2012).
13. Weissleder, R., Kelly, K., Sun, E. Y., Shtatland, T. & Josephson, L. Cell-specific
targeting of nanoparticles by multivalent attachment of small molecules. Nature
Biotechnol. 23, 1418–1423 (2005).
14. Gao, X., Cui, Y., Levenson, R. M., Chung, L. W. & Nie, S. In vivo cancer targeting
and imaging with semiconductor quantum dots. Nature Biotechnol. 22,
969–976 (2004).
15. Michalet, X. et al. Quantum dots for live cells, in vivo imaging, and diagnostics.
Science 307, 538–544 (2005).
16. Jin, Y. D., Jia, C. X., Huang, S. W., O’Donnell, M. & Gao, X. H. Multifunctional
nanoparticles as coupled contrast agents. Nature Commun. 1, 1–8 (2010).
17. Chen, H. W. et al. Reducing non-specific binding and uptake of nanoparticles
and improving cell targeting with an antifouling PEO-b-PgMPS copolymer
coating. Biomaterials 31, 5397–5407 (2010).
18. Bagwe, R. P., Hilliard, L. R. & Tan, W. Surface modification of silica
nanoparticles to reduce aggregation and nonspecific binding. Langmuir 22,
4357–4362 (2006).
19. Grubbs, R. B. Roles of polymer ligands in nanoparticle stabilization. Polym. Rev.
47, 197–215 (2007).
20. Doshi, N. & Mitragotri, S. Designer biomaterials for nanomedicine. Adv. Funct.
Mater. 19, 3843–3854 (2009).
21. Theil, E. C. Ferritin-structure, gene-regulation, and cellular function in animals,
plants, and microorganisms. Annu. Rev. Biochem. 56, 289–315 (1987).
22. Meldrum, F. C., Heywood, B. R. & Mann, S. Magnetoferritin: in vitro s
ynthesis of
a novel magnetic protein. Science 257, 522–523 (1992).
23. Douglas, T. & Young, M. Host–guest encapsulation of materials by assembled
virus protein cages. Nature 393, 152–155 (1998).
24. Allen, M., Willits, D., Mosolf, J., Young, M. & Douglas, T. Protein cage
constrained synthesis of ferrimagnetic iron oxide nanoparticles. Adv. Mater. 14,
1562–1565 (2002).
25. Li, L. et al. Binding and uptake of H-ferritin are mediated by human transferrin
receptor-1. Proc. Natl Acad. Sci. USA 107, 3505–3510 (2010).
26. Sutherland, R. et al. Ubiquitous cell-surface glycoprotein on tumor-cells is
proliferation-associated receptor for transferrin. Proc. Natl Acad. Sci. USA 78,
4515–4519 (1981).
27. Ryschich, E. et al. Transferrin receptor is a marker of malignant phenotype in
human pancreatic cancer and in neuroendocrine carcinoma of the pancreas.
Eur. J. Cancer 40, 1418–1422 (2004).
28. Walker, R. A. & Day, S. J. Transferrin receptor expression in non-malignant and
malignant human breast tissue. J. Pathol. 148, 217–224 (1986).
29. Daniels, T. R., Delgado, T., Rodriguez, J. A., Helguera, G. & Penichet, M. L. The
transferrin receptor part I: biology and targeting with cytotoxic antibodies for
the treatment of cancer. Clin. Immunol. 121, 144–158 (2006).
30. Uchida, M. et al. Targeting of cancer cells with ferrimagnetic ferritin cage
nanoparticles. J. Am. Chem. Soc. 128, 16626–16633 (2006).
31. Lin, X. et al. Chimeric ferritin nanocages for multiple function loading and
multimodal imaging. Nano Lett. 11, 814–819 (2011).
32. Lin, X. et al. Hybrid ferritin nanoparticles as activatable probes for tumor
imaging. Angew. Chem. Int. Ed. 50, 1569–1572 (2011).
33. Gao, L. Z. et al. Intrinsic peroxidase-like activity of ferromagnetic nanoparticles.
Nature Nanotech. 2, 577–583 (2007).
34. Cao, C. Q. et al. Magnetic characterization of noninteracting, randomly
oriented, nanometer-scale ferrimagnetic particles. J. Geophys. Res. 115,
B07103 (2010).
35. Mann, S., Bannister, J. V. & Williams, R. J. Structure and composition of ferritin
cores isolated from human spleen, limpet (Patella vulgata) hemolymph and
bacterial ( Pseudomonas aeruginosa) cells. J. Mol. Biol. 188, 225–232 (1986).
Acknowledgements
This work was partially supported by grants from the National Science and Technology
Major Project (2012ZX10002009-016), the Knowledge Innovation Program of the Chinese
Academy of Sciences (KJCX2-YW-M15), 973 Program (2011CB933500, 2012CB934003),
and the National Defense Science and Technology Innovation Fund of Chinese Academy of
Sciences (CXJJ-11-M61).
Author contributions
M.L. conceived and designed the experiments. K.F. and M.L. performed the experiments.
M.L. and X.Y. reviewed, analysed and inte rpreted the data. C.C. and Y.P. synthesized the
nanoparticles. D.L., D.Y., J.F. and L.S. cultured the cancer cells. M.L. wrote the paper.
All authors discussed the results and commented on the manuscript.
Additional information
The authors declare no comp eting financial interests. Supplementary information
accompanies this paper at www.nature.com/naturenanotechnology.
Reprints and
permission information is available online at http://www.nature.com/reprints. Correspondence
and requests for materials should be addressed to M.L. and X.Y.
LETTERS
NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2012.90
NATURE NANOTECHNOLOGY | VOL 7 | JULY 2012 | www.nature.com/naturenanotechnology464
© 2012 Macmillan Publishers Limited. All rights reserved.
... Ferritin can catalyze color reactions for disease visualizing. Fan et al. [113] constructed magnetoferritin nanoparticles (M-HFn) by diffusion of Fe 2+ into ferritin inner cavity, and Fe 2+ was then oxidated by H 2 O 2 , generating an iron oxide (Fe 3 O 4 ) core in ferritin. The M-HFn could catalyze the oxidation of peroxidase substrates 3,3,5,5-tetramethylbenzidine (TMB) or di-azo-aminobenzene (DAB) to produce a color reaction, for quickly distinguishing and visualizing tumor cells from normal tissues in tissue section analysis. ...
... Moreover, ferritin could load multiple agents in cavity and modify multiple modules on outer shell, which combines many imaging techniques to become a multimodal imaging probe, or co-delivery of diagnostic and therapeutic modules to become a multifunctional and integrated nanomedicine. Zhao et al. [113] conjugated 125 I radionuclide to the outer shell of the M-HFn mentioned above to combine single photon emission tomography (SPECT) and MRI. This study broke through apparent sensitivity limitations between MRI and SPECT, enabling single-dose MRI/nuclear tumor-targeted imaging. ...
... Ferritin has been extensively researched for tumortargeted delivery of small molecule drugs. Ferritin has high intrinsic affinity for TfR1 over expressed tumors, including liver, lung and breast cancer, etc. [113]. Liang et al. [47] encapsulated Dox in ferritin, which exhibited more than tenfold higher tumoral Dox concentration than free Dox treatment in mice, and even showed longer survival and lower toxicity than clinically approved liposomal Dox (DOXIL)-treated mice. ...
Article
Full-text available
Ferritin is an endogenous protein which is self-assembled by 24 subunits into a highly uniform nanocage structure. Due to the drug-encapsulating ability in the hollow inner cavity and abundant modification sites on the outer surface, ferritin nanocage has been demonstrated great potential to become a multi-functional nanomedicine platform. Its good biocompatibility, low toxicity and immunogenicity, intrinsic tumor-targeting ability, high stability, low cost and massive production, together make ferritin nanocage stand out from other nanocarriers. In this review, we summarized ferritin-based nanomedicine in field of disease diagnosis, treatment and prevention. The different types of drugs to be loaded in ferritin, as well as drug-loading methods were classified. The strategies for site-specific and non-specific functional modification of ferritin were investigated, then the application of ferritin for disease imaging, drug delivery and vaccine development were discussed. Finally, the challenges restricting the clinical translation of ferritin-based nanomedicines were analyzed.
... Horseradish peroxidase was replaced by iron oxide NPs in ELISA. Simultaneously, the same group utilized the NPs of iron oxide that were enclosed in ferritin to determine the tumors (transferrin receptor positive) in the sections of tissue [165]. These NPs could specifically detect the transferrin receptor (present in excess amounts in the tumors) because of the presence of a coating of ferritin. ...
Article
Full-text available
Nanotechnology has become one of the most extensive fields of research. Nanoparticles (NPs) form the base for nanotechnology. Recently, nanomaterials (NMs) are widely used due to flexible chemical, biological, and physical characteristics with improved efficacy in comparison to bulk counterparts. The significance of each class of NMs is enhanced by identifying their properties. Day by day, there is an emergence of various applications of NMs, but the toxic effects associated with them cannot be avoided. NMs demonstrate therapeutic abilities by enhancing the drug delivery system, diagnosis, and therapeutic effects of numerous agents, but determining the benefits of NMs over other clinical applications (disease-specific) or substances is an ongoing investigation. This review is aimed at defining NMs and NPs and their types, synthesis, and pharmaceutical, biomedical, and clinical applications.
... As a replacement for regular enzymes, metal-organic frameworks (MOFs) and their generated products with enzyme-like activity could be used as antimicrobial medications, and in detecting and treating cancer. Their role as biosensing material, antibacterial compounds, cancer treatment, and similar simulation enzymes has been emphasized in recent studies Fan et al. 2012). ...
Book
The research activities on nanozymes have witnessed boom over the past decade due to their high catalytic activity, low cost and high stability. The burgeoning field of nanozymes is still in its infancy as research is underway on various aspects of nanozymes such as their underlying mechanism of action, engineering efficient nanozymes, and exploring their wide applications. Nanozymes are being used in various spheres of science such as diagnostics, therapeutics, and electronics, however their applicability in different ways to monitor and remediate the environmental pollutants is naïve and bears immense future potential. In recent years, applications of nanozymes are being explored for environmental monitoring. They are being used to develop new methods for sensitive detection of environmental pollutants, removal/extraction of these pollutants from soil or water, and their degradation to relatively safer by products. However, their effective implementation in environmental field to support sustainable growth is yet challenging as it needs deeper understanding of the fundamental principles of their catalytic mechanism, and subsequently design new nanozymes with high selectivity and reusability
... As a replacement for regular enzymes, metal-organic frameworks (MOFs) and their generated products with enzyme-like activity could be used as antimicrobial medications, and in detecting and treating cancer. Their role as biosensing material, antibacterial compounds, cancer treatment, and similar simulation enzymes has been emphasized in recent studies Fan et al. 2012). ...
Book
The book is about the applications of Nanozymes in monitoring Environmental pollutants and the removal/extraction of these pollutants. Chapters address the various role of nanozymes in real-time monitoring and developing sensors for the same.
... Iron is necessary for DNA synthesis as well as other biological functions. Malignant cells frequently overexpress TFR1 due to its essential role in the pathophysiology of cancer cells, and this increased expression has been linked to a poor prognosis [22]. TFR1 is an appealing target for antibody-mediated therapy due to the high levels of TFR1 expression on malignant cells, as well as its extracellular accessibility, capacity for internalization, and crucial role in the pathophysiology of cancer cells. ...
Article
Full-text available
Cholangiocarcinoma (CCA) is the second most common primary hepatic malignancy and associated with poor prognosis. Lack of therapeutic methods for CCA and insensitivity of targeted therapy and immunotherapy make its treatment challenging. NUF2, a component of Ndc80 kinetochore complex, is implicated in the initiation and development of multiple cancers. However, the role and mechanism of NUF2 in CCA is still unclear. In this research, we investigated the biological processes and underlying mechanisms of NUF2 in CCA. We discovered that the expression of NUF2 was upregulated in CCA and negatively correlated with prognosis. Changes in NUF2 levels had an impact on cell proliferation and migration. Moreover, NUF2 functioned as an oncogene to promote the progression of CCA through p38/MAPK signaling by inhibiting p62 binding of TFR1 and affecting its autophagic degradation. In addition, TFR1 promoted CCA progression and Kaplan-Meier analyses uncovered patients with high expression of TFR1 was associated with the poor survival. In conclusion, our study demonstrated that NUF2 promoted CCA progression by regulating TFR1 protein degradation, and the NUF2/TFR1/MAPK axis could be an excellent therapeutic target for CCA.
Chapter
Molecular dynamics (MD) simulations enable the description of the physical movement of the system over time based on classical mechanics at various scales depending on the models. Protein cages are a particular group of different-size proteins with hollow, spherical structures and are widely found in nature, which have vast applications in numerous fields. The MD simulation of cage proteins is particularly important as a powerful tool to unveil their structures and dynamics for various properties, assembly behavior, and molecular transport mechanisms. Here, we describe how to conduct MD simulations for cage proteins, especially technical details, and analyze some of the properties of interest using GROMACS/NAMD packages.Key wordsMolecular dynamics (MD) simulationsProtein cagesGROMACSNAMD
Book
This book highlights recent developments related to fabrication and utilization of nanoparticle-engineered metal matrices and their composites linked to the heavy industries, temperature fasteners, high-pressure vessels, and heavy turbines, etc. The mechanical properties of newly developed metallic composites are discussed in terms of tensile modulus, hardness, ductility, crack propagation, elongation, and chemical inertness. This book presents the design, development, and implementation of state-of-the-art methods linked to nanoparticle-reinforced metal nanocomposites for a wide variety of applications. Therefore, in a nutshell, this book provides a unique platform for researchers and professionals in the area of nanoparticle-reinforced metal nanocomposites.
Chapter
Full-text available
Investigation and evolution in the involved sciences at the molecular or atomic level is the demand of the day under the discipline of nanoscience or nanotechnology, with massive impact on almost all regions of human health and conditioning, including various medical domains such as clinical diagnoses, pharmacological investigations, and additional resistant methods. The domain of nano-dentistry has appeared due to the assorted dental applications and consequences of nanotechnology. This chapter first outlines classifications and categories of nanomaterials established on their source, biochemical arrangement, resources, and measurements. The basic effects of value conversion at the nanoscale and the multiple improved and valuable effects of fabricated nanomaterials, such as incarceration effects, texture influences, mechanical impacts, structural developments, thermal developments, optical effects, magnetic effects, and dental application, are also described. In the final section, we looked at in vivo, in vitro, and the effects of nanomaterials.
Chapter
Full-text available
Metallic nanoparticles (NPs) have fascinated scientists all over the world for over a century. They grab this attention because of their huge potential in nanotechnology. Nanoparticles are a very special class of materials, with sizes ranging from 1 to 100 nm. The uniqueness of these nanoparticles is due to the fact that their properties—physical, chemical, optical, and mechanical—are sensitive to their size, shape, and structure. They can be grouped into fullerenes, metal nanoparticles, ceramics, and polymeric-based nanoparticles. The most important application and industrial demand project is the use of these specialized materials in various fields like coatings, nanolubrication, and catalysis. In this chapter, a brief introduction to metal nanoparticles followed by their synthesis methods is presented. The main objective of this chapter is to discuss the principles underlying the mechanical properties of these metal nanoparticles, e.g., hardness, creep, adhesion, and friction. Pursuant to these, several of the main applications, i.e., lubricant additives, nanoparticles in nanomanufacturing, and nanoparticle-reinforced composite coatings because of their special mechanical properties are discussed.
Article
Full-text available
Early noninvasive detection and characterization of solid tumors and their supporting neovasculature is a fundamental prerequisite for effec- tive therapeutic intervention, particularly antiangiogenic treatment regi- mens. Emerging molecular imaging techniques now allow recognition of early biochemical, physiological, and anatomical changes before manifes- tation of gross pathological changes. Although new tumor, vascular, ex- tracellular matrix, and lymphatic biomarkers continue to be discovered, the 3-integrin remains an attractive biochemical epitope that is highly expressed on activated neovascular endothelial cells and essentially absent on mature quiescent cells. In this study, we report the first in vivo use of a magnetic resonance (MR) molecular imaging nanoparticle to sensitively detect and spatially characterize neovascularity induced by implantation of the rabbit Vx-2 tumor using a common clinical field strength (1.5T). New Zealand White rabbits (2 kg) 12 days after implantation of fresh Vx-2 tumors (2 2 2m m 3) were randomized into one of three treatment groups: (a) 3-targeted, paramagnetic formulation; (b) nontargeted, paramagnetic formulation; and (c) 3-targeted nonparamagnetic nano- particles followed by (2 h) the 3-targeted, paramagnetic formulation to competitively block magnetic resonance imaging (MRI) signal enhance- ment. After i.v. systemic injection (0.5 ml of nanoparticles/kg), dynamic T1-weighted MRI was used to spatially and temporally determine nano- particle deposition in the tumor and adjacent tissues, including skeletal muscle. At 2-h postinjection, 3-targeted paramagnetic nanoparticles increased MRI signal by 126% in asymmetrically distributed regions primarily in the periphery of the tumor. Similar increases in MR contrast were also observed within the walls of some vessels proximate to the tumor. Despite their relatively large size, nanoparticles penetrated into the leaky tumor neovasculature but did not appreciably migrate into the interstitium, leading to a 56% increase in MR signal at 2 h. Pretargeting of the 3-integrin with nonparamagnetic nanoparticles competitively blocked the specific binding of 3-targeted paramagnetic nanoparticles, decreasing the MR signal enhancement (50%) to a level attributable to local extravasation. The MR signal of adjacent hindlimb muscle or con- tralateral control tissues was unchanged by either the 3-targeted or control paramagnetic agents. Immunohistochemistry of 3-integrin corroborated the extent and asymmetric distribution of neovascularity observed by MRI. These studies demonstrate the potential of this targeted molecular imaging agent to detect and characterize (both biochemically and morphologically) early angiogenesis induced by minute solid tumors with a clinical 1.5 Tesla MRI scanner, facilitating the localization of nascent cancers or metastases, as well as providing tools to phenotypically categorize and segment patient populations for therapy and to longitudi- nally follow the effectiveness of antitumor treatment regimens.
Article
Full-text available
The spread of cancer cells between organs, a process known as metastasis, is the cause of most cancer deaths1, 2. Detecting circulating tumour cells—a common marker for the development of metastasis3, 4—is difficult because ex vivo methods are not sensitive enough owing to limited blood sample volume and in vivo diagnosis is time-consuming as large volumes of blood must be analysed5, 6, 7. Here, we show a way to magnetically capture circulating tumour cells in the bloodstream of mice followed by rapid photoacoustic detection. Magnetic nanoparticles, which were functionalized to target a receptor commonly found in breast cancer cells, bound and captured circulating tumour cells under a magnet. To improve detection sensitivity and specificity, gold-plated carbon nanotubes conjugated with folic acid were used as a second contrast agent for photoacoustic imaging. By integrating in vivo multiplex targeting, magnetic enrichment, signal amplification and multicolour recognition, our approach allows circulating tumour cells to be concentrated from a large volume of blood in the vessels of tumour-bearing mice, and this could have potential for the early diagnosis of cancer and the prevention of metastasis in humans.
Article
Full-text available
Acute leukemia is a hematopoietic malignancy for which the accurate measurement of minimal residual disease is critical to determining prognosis and treatment. Although bone marrow aspiration and light microscopy remain the current standard of care for detecting residual disease, these approaches cannot reliably discriminate less than 5% lymphoblast cells. To improve the detection of leukemia cells in the marrow, we developed a novel apparatus that utilizes antibodies conjugated to superparamagnetic iron oxide nanoparticles (SPION) and directed against the acute leukemia antigen CD34, coupled with a "magnetic needle" biopsy. Leukemia cell lines expressing high or minimal CD34 were incubated with anti-CD34-conjugated SPIONs. Three separate approaches including microscopy, superconducting quantum interference device magnetometry, and in vitro magnetic needle extraction were then used to assess cell sampling. We found that CD34-conjugated nanoparticles preferentially bind high CD34-expressing cell lines. Furthermore, the magnetic needle enabled identification of both cell line and patient leukemia cells diluted into normal blood at concentrations below those normally found in remission marrow samples. Finally, the magnetic needle enhanced the percentage of lymphoblasts detectable by light microscopy by 10-fold in samples of fresh bone marrow aspirate approximating minimal residual disease. These data suggest that bone marrow biopsy using antigen-targeted magnetic nanoparticles and a magnetic needle for the evaluation of minimal residual disease in CD34-positive acute leukemias can significantly enhance sensitivity compared with the current standard of care. [Cancer Res 2009;69(21):8310-6]
Article
Both inorganic nanoparticles and polymers have become ubiquitous components of nanostructured systems. The development of an understanding of the interactions between these components will be of crucial importance in the further elaboration of these systems. This review touches upon two aspects of control over surface chemistry in inorganic nanoparticles with polymer ligands: (1) the effect of polymer ligands on accessibility of the inorganic nanoparticle surface to other species, such as is necessary in catalysis; and (2) the design of polymer ligands that can be modified to enable compatibility of the entire polymer‐coated nanoparticle with a broad variety of other materials.
Article
Nanotechnology has had tremendous impact on medical science and has resulted in phenomenal progress in the field of drug delivery and diagnostics. A wide spectrum of novel nanomaterials including polymeric particles, liposomes, quantum dots, and iron oxide particles have been developed for applications in therapeutic delivery and diagnostics. This has resulted in control over the rate and period of delivery and targeting of drugs to specific organs in the human body. This feature article focuses on the delivery of drugs using polymeric particles. The size, choice of polymer, surface chemistry, shape, and mechanical properties of the particles are parameters that critically affect particle function. Numerous biomaterials and fabrication techniques have been developed in the last decade that focus on novel design parameters, such as shape and mechanical properties and the interplay of these parameters with the size and surface chemistry of particles. Recent advances with particular focus on the importance of particle shape are highlighted, and the challenges that are yet to be fulfilled are underscored.
Article
The protein cage constrained synthesis of ferrimagnetic iron oxide nanoparticles was presented. The ferritin-like protein (FLP) from the bacteria Listeria innocua was cloned into an inducible E. coli heterologous expression system and was analyzed by size exclusion chromatography (SEC), transmission electron microscopy (TEM) and dynamic light scattering (DLS). The Fe mineralization in the FLP cage under the non-physiological conditions of elevated temperature, pH and controlled oxidation was also investigated.
Article
1] Studying the magnetic properties of ultrafine nanometer‐scale ferrimagnetic particles (<10 nm) is vital to our understanding of superparamagnetism and its applications to environmental magnetism, biogeomagnetism, iron biomineralization, and biomedical technology. However, magnetic properties of the ultrafine nanometer‐sized ferrimagnetic grains are very poorly constrained because of ambiguities caused by particle magnetostatic interactions and unknown size distributions. To resolve these problems, we synthesized magnetoferritins using the recombinant human H chain ferritin (HFn). These ferrimagnetic HFn were further purified through size exclusion chromatography to obtain monodispersed ferrimagnetic HFn. Transmission electron microscopy revealed that the purified ferrimagnetic HFn are monodispersed and each consists of an iron oxide core (magnetite or maghemite) with an average core diameter of 3.9 ± 1.1 nm imbedded in an intact protein shell. The R value of the Wohlfarth‐Cisowski test measured at 5 K is 0.5, indicating no magnetostatic interactions. The saturation isothermal remanent magnetization acquired at 5 K decreased rapidly with increasing temperature with a median unblocking temperature of 8.2 K. The preexponential frequency factor f 0 determined by AC susceptibility is (9.2 ± 7.9) × 10 10 Hz. The extrapolated M rs /M s and B cr /B c at 0 K are 0.5 and 1.12, respectively, suggesting that the ferrimagnetic HFn cores are dominated by uniaxial anisotropy. The calculated effective magnetic anisotropy energy constant K eff = 1.2 × 10 5 J/m 3 , which is larger than previously reported values for bulk magnetite and/or maghemite or magnetoferritin and is attributed to the effect of surface anisotropy. These data provide useful insights into superparamagnetism as well as biomineralization of ultrafine ferrimagnetic particles.
Article
Engineered nanoparticles have been used to provide diagnostic, therapeutic and prognostic information about the status of disease. Nanoparticles developed for these purposes are typically modified with targeting ligands (such as antibodies, peptides or small molecules) or contrast agents using complicated processes and expensive reagents. Moreover, this approach can lead to an excess of ligands on the nanoparticle surface, and this causes non-specific binding and aggregation of nanoparticles, which decreases detection sensitivity. Here, we show that magnetoferritin nanoparticles (M-HFn) can be used to target and visualize tumour tissues without the use of any targeting ligands or contrast agents. Iron oxide nanoparticles are encapsulated inside a recombinant human heavy-chain ferritin (HFn) protein shell, which binds to tumour cells that overexpress transferrin receptor 1 (TfR1). The iron oxide core catalyses the oxidation of peroxidase substrates in the presence of hydrogen peroxide to produce a colour reaction that is used to visualize tumour tissues. We examined 474 clinical specimens from patients with nine types of cancer and verified that these nanoparticles can distinguish cancerous cells from normal cells with a sensitivity of 98% and specificity of 95%.
Article
Nanoscale therapeutic interventions are increasingly important elements in the portfolio of cancer therapeutics. The efficacy of nanotherapeutics is dictated, in part, by the access they have to tumors via the leaky tumor vasculature. Yet, the extent of tumor vessel leakiness in individual tumors varies widely resulting in a correspondingly wide tumor dosing and resulting range of responses to therapy. Here we report the design of a multifunctional nanocarrier that simultaneously encapsulates a chemotherapeutic and a contrast agent which enables a personalized nanotherapeutic approach for breast cancer therapy by permitting tracking of the nanocarrier distribution by mammography, a widely used imaging modality. Following systemic administration in a rat breast tumor model, imaging demonstrated a wide range of intratumoral deposition of the nanocarriers, indicating variable tumor vessel leakiness. Notably, specific tumors that exhibited high uptake of the nanocarrier as visualized by imaging were precisely the animals that responded best to the treatment as quantified by low tumor growth and prolonged survival.