You are on page 1of 16

Biomaterials 145 (2017) 138e153

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Clickable and imageable multiblock polymer micelles with


magnetically guided and PEG-switched targeting and release property
for precise tumor theranosis
Jing Wei a, Xiaoyu Shuai a, Rui Wang a, Xueling He a, b, Yiwen Li a, Mingming Ding a, *,
Jiehua Li a, Hong Tan a, **, Qiang Fu a
a
College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
b
Laboratory Animal Center of Sichuan University, Chengdu, 610041, China

a r t i c l e i n f o a b s t r a c t

Article history: Targeted delivery of therapeutics and diagnostics using nanotechnology holds great promise to minimize
Received 22 February 2017 the side effects of conventional chemotherapy and enable specific and real-time detection of diseases. To
Received in revised form realize this goal, we report a clickable and imageable nanovehicle assembled from multiblock poly-
1 July 2017
urethanes (MPUs). The soft segments of the polymers are based on detachable poly(ethylene glycol)
Accepted 1 August 2017
Available online 18 August 2017
(PEG) and degradable poly(ε-caprolactone) (PCL), and the hard segments are constructed from lysine-
and cystine-derivatives bearing reduction-responsive disulfide linkages and click-active alkynyl moieties,
allowing for post-conjugation of targeting ligands via a click chemistry. It was found that the cleavage of
Keywords:
Click chemistry
PEG corona bearing a pH-sensitive benzoic-imine linkage (BPEG) could act as an on-off switch, which is
Biodegradable multiblock polyurethane capable of activating the clicked targeting ligands under extracellular acidic condition, followed by
Multifunctional polymer micelles triggering the core degradation and payload release within tumor cells. In combination with super-
Magnetic resonance imaging paramagnetic iron oxide nanoparticles (SPION) clustered within the micellar core, the MPUs exhibit
Tumor targeting excellent magnetic resonance imaging (MRI) contrast effects and T2 relaxation in vitro, as well as
Cancer theranosis magnetically guided MR imaging and multimodal targeting of therapeutics to tumor precisely, leading to
significant inhibition of cancer with minimal side effect. This work provides a safe and versatile platform
for the further development of smart theranostic systems for potential magnetically-targeted and
imaging-guided personalized medicine.
© 2017 Elsevier Ltd. All rights reserved.

1. Introduction including poor solubility, low stability, rapid clearance and lack of
selectivity, which have exerted adverse effects on healthy cells and
The effective treatment and early detection of cancers still prevented the dose escalation necessary to eliminate malignant
remain great challenges, since the transport of therapeutic and cells [3]. On the other hand, the encapsulation of diagnostic agents
diagnostic agents to the tumors have been seriously hampered by into polymeric nanovehicles can remedy the inadequate sensitivity
the complicated body environments and various physiological and specificity of most commercialized contrast agents for imaging
barriers [1]. To address these challenges, targeted delivery system (e.g., Endorem, Resovist, etc.) [4], resulting in an improved spatial
based on nanotechnology holds enormous potential to enable revolution and remarkable contrast against surrounding tissues by
engineered nanomedicines to travel through the body in a specific clustering these agents and localizing them at the sites of interest
way [2]. On the one hand, drug-loaded nanoparticle systems can [5].
overcome the problems brought by conventional free drugs, Relying on the so-called enhanced permeability and retention
(EPR) effect ascribable to the presence of leaky vasculature and
impaired lymphatic drainage in most rapidly growing tumors,
nanomedicines show higher accumulation at tumor sites than the
* Corresponding author.
conventional drugs and contrast compounds [6]. However, EPR
** Corresponding author.
E-mail addresses: dmmshx@scu.edu.cn, dmmshx@163.com (M. Ding), hongtan@ effect has been regarded recently as an immensely heterogeneous
scu.edu.cn (H. Tan). phenomenon, which varies with the types and stages of diseases

http://dx.doi.org/10.1016/j.biomaterials.2017.08.005
0142-9612/© 2017 Elsevier Ltd. All rights reserved.
J. Wei et al. / Biomaterials 145 (2017) 138e153 139

and differs from person to person [7]. Therefore, the EPR-based functions into a single macromolecule in a smart and coordinated
passive targeting seems not sufficient to match the changeable way, as demonstrated recently by our group [29]. However, the role
and complicated tumor microenvironment. As a result, it is only of MPUs in the field of imaging and personalized nanomedicine
effective for limited patient subpopulations [8]. The cooperation of remains largely unexplored [30]. To date, the theranostic property
magnetic nanoparticles is expected to offer improved specificity via of MPUs have yet to be demonstrated. In addition, to make MPUs
magnetic drug targeting (MDT) [9,10]. As a physical force, the more clinically approvable, a further improvement of PU versatility
magnetic stimulus is independent of the complicated physiological and optimization of polymer structure is clearly warranted [27].
conditions. Hence, MDT employing an external field gradient to In this study, we report a clickable and imageable multiblock
manipulate magnetically responsive particles can expedite the polyurethane (MPU) system with switchable tumor targeting and
extravasation and transport of nanoparticles into tumors, and triggered drug release properties for precise tumor therapy and
thereby break through the physical barriers and overcome the specific MR imaging (Fig. 1). The soft segments of MPUs comprise
intrinsic limitations of EPR effect [9,11]. Furthermore, magnetic PEG and PCL, and the hard segments contain L-lysine ethyl ester
nanoparticles can also act as excellent contrast agents for magnetic diisocyanate (LDI) (Fig. 1A). To eliminate the need of multiple chain
resonance imaging (MRI), which provides great opportunity to extenders and complicated synthetic procedures for the construc-
construct theranostic platform for specific disease detection, real- tion of multifunctional MPUs [29], an L-cysteine-derived versatile
time monitoring and potential imaging-guided therapy [11,12]. chain extender (Cys-PA) was incorporated to endow polymers with
Although nanomedicines can passively or magnetically target to a number of reduction-cleavable disulfide linkages in the backbone
tumor tissues, the scarcity of cell-specific interaction and insuffi- and clickable alkyne sites on the side chains (Fig. 1). Subsequently,
cient uptake of nanoparticles may decrease the therapeutic efficacy as a proof-of-concept, a post-conjugation of targeting ligand was
and even induce drug expulsion and multiple drug resistance performed using a facile click chemistry after the formation of
(MDR) [13]. To overcome this problem, nanocarriers can be polymer micelles (Fig. 1C). We found lately that FA-clicked polymer
equipped with a good variety of targeting ligands, such as anti- micelles exhibit relatively lower targeting efficacy compared with
bodies, folic acid (FA) and peptides that can actively bind to anti- other FA-based delivery systems [31]. Herein, it is interesting to
gens and receptors highly overexpressed by tumor cells [14]. note that the cleavage of PEG segment bearing a pH-sensitive
Unfortunately, most of the actively targeted delivery systems benzoic-imine linkage (BPEG) could act as a switch, which is
contain ligands on their surface [15]. The exposure of active surface capable of activating cell targeting under extracellular condition,
may promote nonspecific interactions with endothelial and other followed by triggering the cleavage of disulfide and accelerating the
non-cancerous cells and result in opsonization-mediated clearance release of payloads within tumor cells (Fig. 1E). Furthermore, super-
of nanomedicines from the body [16]. To address this dilemma, paramagnetic iron oxide nanoparticles (SPIONs) and doxorubicin
coating nanocarrier with hydrophilic polyethylene glycol (PEG) (DOX) as model contrast and anticancer agents were efficiently co-
corona (known as PEGylation) can increase the circulation time by encapsulated into the micellar core to enable magnetophoretic
reducing interactions with serum proteins and mitigating uptake enhance of targeting and theranostic capability of MPUs. The
by phagocytic cells [17], whereas such a strategy in turn compro- multiple stimuli-responsive property and multimodal targeting
mises the targeting specificity [18]. effect of MPUs were systematically demonstrated, and the
Another approach to targeting is to take advantage of the special magnetically guided cancer therapy and MR imaging were also
microenvironments of tumors to achieve on-demand delivery, evaluated in vivo.
which in principle allow for tailored release profiles with excellent
temporal, spatial and dosage control [19]. It is known that most 2. Materials and methods
solid tumors develop unique microenvironments, such as lowered
interstitial pH, elevated glutathione (GSH) concentration and 2.1. Materials
increased level of certain enzymes [20]. In particular, the GSH
concentration in some tumors is several times higher than that in N,N-Dimethylacetamide (DMAc) was purchased from Adamas
normal tissues, and the intracellular level of GSH (1e11 mM in the Reagent Co., Ltd., China, and dried over CaH2 overnight and vacuum
cytoplasm) is 2e3 orders higher than that outside the cells distilled. Methoxyl-poly(ethylene glycol) (MPEG, MW 1900, Alfa
(2e20 mM) [21,22]. Therefore, bio-responsive formulations con- Aesar) and PCL (MW 2000, Dow Chemical) were dehydrated under
taining disulfide functionality can facilitate tumor-specific and reduced pressure at 90  C for 2 h before use. LDI, BPEG, Cys-PA, and
intracellular transport of therapeutics by the cleavage of disulfide azido modified FA (AzFA) were synthesized according to previous
bond through thiol-disulfide exchange reactions triggered by GSH reports [31,32]. Iron (III) chloride hexahydrate (FeCl3$6H2O) was
[23]. Nonetheless, recent in vivo studies have shown that disulfide purchased from Tianjin Damao Chemical Co., Ltd., China.
could be potentially cleaved under circulation due to the presence CuSO4.5H2O was attained from Tianjin Bodi Chemical Co., Ltd.,
of reducing species in the blood [24], thus decreasing the specificity China. Sodium ascorbate was purchased from Aladdin Chemical Co.
of redox-responsive delivery systems [25]. On the other hand, the Ltd., China. Iron (II) chloride tetrahydrate (FeCl2$4H2O), ammonium
current studied reduction-responsive systems are commonly pre- hydroxide solution (NH3$H2O), oleic acid, propanediol (PDO,
pared by incorporating non-native segments containing disulfide 98.0%) and dimethyl sulfoxide (DMSO) were supplied by Chengdu
linkages, which may raise a safety concern [21,26]. Kelong Chemical Co., Ltd., China. Doxorubicin hydrochloride
As mentioned above, although the conventional targeting and (DOX$HCl) was obtained from Tecoland Co., USA. 3-(4,5-dimethyl
responsive systems taking advantages of EPR effect, targeting li- thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) was pur-
gands and tumor microenvironments allow for specific delivery of chased from Sigma-Aldrich, USA. 2-(4-amidinophenyl)-6-
theranostics, each of these approaches is still suffering from some indolecarbamidine dihydrochloride (DAPI) was obtained from
limitations. Hence, the development of versatile nanomedicines to Roche Diagnostics, Germany. Other reagents were purchased from
overcome all the limitations and realize multimodal targeting and commercial suppliers and used without further purification.
precise therapy is highly desirable. To fulfill this need, multiblock
polyurethane (MPU) appears as a promising platform due to their 2.2. Characterization
good biocompatibility and high molecular tunability [27,28]. MPUs
could be molecularly engineered to integrate various desired Proton nuclear magnetic resonance (1H NMR, 400 MHz)
140 J. Wei et al. / Biomaterials 145 (2017) 138e153

Fig. 1. Design of clickable and imageable multiblock polymer micelles. Schematic chemical structure (A) and molecular architecture (B) of clickable MPUs. The waved lines in (A)
represent PCL soft segments. (C) Self-assembly of MPU micelles and post-conjugation of folic acid via click chemistry. (D) Schematic illustration of FA residues on the interface of
polymer micelles. (E) Illustration of magnetic-guided and PEG-switched targeting and release properties of MPU nanocarriers.

spectroscopy was performed on a Bruker AV II-400 MHz spec- an angle of 90 . Each sample was determined for three times.
trometer using DMSO-d6 as a solvent and TMS as an internal Transmission electron microscopy (TEM, Hitachi model H-600-
standard at room temperature. 4, Japan) was recorded with an accelerating voltage of 75 KV. A drop
Fourier transform infrared (FTIR) spectroscopy was performed of micellar solution was placed on a copper grid with Formvar film,
on a Nicolet 6700 spectrometer (Thermo Electron Corporation, and stained with 1% (w/v) phosphotungstic acid. Then the solution
U.S.A). The solution samples were cast on KBr plates before was absorbed by a filter paper and dried naturally before mea-
measurement. surement. For the observation of magnetic nanoparticles, no
Gel permeation chromatography (GPC) was performed on staining was needed.
Waters-1515 (U.S.A) with DMF as an eluent at 40  C. Samples were High-resolution TEM (HRTEM, Tecnai G2 F20 S-TWIN, FEI, USA)
run at 2.0 mg mL1 with a flow rate of 1.0 mL min1. was operated with an accelerating voltage of 200 KV. A drop of
Size measurement was performed on a Zetasizer Nano ZS dy- SPION solution in hexane was dropped on a copper grid with For-
namic light-scattering (DLS) instrument (Malvern, UK) at 25  C at mvar film and air-dried before measurement.
J. Wei et al. / Biomaterials 145 (2017) 138e153 141

Thermogravimetric analyzer (TGA Q500, TA Instruments, USA) 2.3.4. Preparation of FA-clicked MPU micelles
was used to obtain the TGA curves of samples. The fully dried 50 mL of MPU micellar solution (1 mg mL1) was mixed with
powder was heated from room temperature to 800  C at a rate of 10 mg of AzFA, and 1 mg of sodium ascorbate and 0.5 mg of
10  C min1 under argon atmosphere. CuSO$45H2O were added as catalysts. The solution was stirred for
Crystallographic analysis was carried out on an X-ray diffrac- 24 h at room temperature, then transferred into a dialysis bag
tometer (XRD, Phlips X'Pert PRO, XL-30) using Cu Ka radiation. The (MWCO 3500) and dialyzed against PBS (pH 7.4) for 5 d. Thereafter,
scattering angle (2q) was in the range of 20 e90 . The structure and the solution was centrifugalized at 3000 r min1 for 20 min and
composition of the sample were determined by comparing the filtrated by a 0.45 mm pore-sized syringe filter and stored at 4  C.
spectrogram with standard powder diffraction files from the Joint The grafting ratio of FA on PU micelles was measured using UVevis
Committee for Powder Diffraction Studies (JCPDS). spectroscopy at the wavelength of 280 nm, and calculated ac-
Vibrating sample magnetometer (VSM, Lakeshore 7410, USA) cording to a calibration curve generated from AzFA solutions at
was used at 298 K under an applied magnetic field varying different concentrations.
from 15 to15 kOe.
2.3.5. Synthesis of SPIONs
2.3. Construction and characterization of clickable and imageable SPIONs were synthesized using a modified chemical co-
multiblock polymer micelles precipitation method [30,33]. Briefly, FeCl3$6H2O (4.1 g) and
FeCl2$4H2O (1.8 g) were dissolved in 45 mL of distilled water with
2.3.1. Synthesis of clickable and multistimuli-responsive MPUs stirring under nitrogen. Then 12 mL of NH3$H2O was quickly added
The clickable MPUs were synthesized through a solution poly- for 1 h of reaction at 80  C. Thereafter, 1.8 g of oleic acid was added
merization as described previously [31]. In brief, PCL and LDI were dropwise, and the reaction was allowed to continue for 1 h. When
copolymerized in anhydrous DMAC at 60  C under a dry nitrogen the mixture was cooled to ambient temperature, the SPION fluid
atmosphere for 1 h in the presence of 1‰ stannous octoate as a was collected using a magnet and washed with water and ethanol
catalyst. After cooling to room temperature, multifunctional chain for several times. The obtained product was re-dispersed in hexane
extender Cys-PA was added to react for 1 h at room temperature and storage at 4  C.
and another 2 h at 60  C. Then different ratios of MPEG and BPEG
were added, and the reaction was completed after 6 h. Finally, the 2.3.6. Preparation of SPION and drug-loaded MPU micelles
polymers were precipitated in a mixture of methanol and anhy- To prepare SPION-loaded MPU micelles (SPION@micelles),
drous diethyl ether and dried under vacuum at 60  C. MPUs and SPIONs were co-dissolved in THF/DMAc (1/9, v/v) and
dropped into deionized water under ultrasonic condition. The so-
2.3.2. Preparation of clickable MPU micelles lution was dialyzed against deionized water for 3 d, centrifugalized
MPU micelles were prepared by a dialysis method. Typically, a at 3000 r min1 for 15 min, and filtered through a 0.45 mm pore-
solution of MPU in DMAc (10 mg mL1) was dropped into phos- sized syringe filter and stored at 4  C. To determine the loading
phate buffered saline (PBS, pH 7.4) under ultrasonic condition. Then content of SPIONs in MPU micelles, the SPION@micelle solution was
the solution was transferred into a dialysis bag (MWCO 3500) and lyophilized and characterized with TGA. To assess the dispersibility
dialyzed against PBS for 3 d, refreshing the dialysate every 3 h. of magnetic loaded nanocarriers, the SPION@micelle solution was
Afterward, the micellar solution was diluted with PBS to 1 mg mL1 mixed with an equal volume of hexane. The mixture was vigorously
and centrifugalized at 3000 r min1 for 15 min. Finally, the obtained shaken and allowed to separate into two layers, then imaged with a
micelles were passed through a 0.45 mm pore-sized syringe filter color digital camera.
(Milipore, Carrigtwohill, Co. Cork, Ireland) and stored at 4  C. To load drugs, DOX$HCl was firstly dissolved in DMAc and
desalted in the presence of excess amount of trimethylamine (TEA)
2.3.3. Stability and corona-detachment behavior with stirring for 2 h. The solution was mixed with the above MPU
To evaluate the stability and stimuli-responsiveness of clickable solution in DMAc or MPU/SPIONs solution in THF/DMAc, respec-
and imageable polymer micelles, the samples were incubated in pH tively, for the preparation of drug-loaded micelles (DOX@micelles)
7.4 or pH 6.5 at 37  C with shaking (100 r min1) for 24 h, and then or drug and SPION co-encapsulated micelles (DOX/SPION@mi-
measured by DLS and TEM. celles). Afterward, the mixtures were dropped into PBS (pH 7.4)
To investigate the corona-detachment profiles, 10 mL of SSPHPU under ultrasonic condition, and dialyzed against PBS for 3 d to
micellar solution (2 mg mL1) was transferred into a dialysis bag remove the organic solvents. The micellar solution was cen-
(MWCO 6500) and incubated at pH 6.5 and pH 7.4 for different trifugalized for 15 min at 3000 r min1 and filtered as described
times at 37  C with shaking (110 r min1). At proper time intervals, above except that the whole process was operated in the dark. The
3 mL of release media was taken out and replenished with an equal loading content of DOX in MPU micelles was determined using
volume of fresh media. The release of PEG derivatives was deter- UVevis spectroscopy.
mined by an L5 ultravioletevisible (UVeVis) spectrometer
(Shanghai INESA Analytical Instrument Co., Ltd.) at the wavelength 2.4. PEG-switched and GSH-accelerated drug release
of 256 nm. The concentration of released PEG segments was
calculated according to the calibration curve established from To study the controlled drug release behavior of multifunctional
MPEG-CBA standard solutions in release media at different con- micelles, 2 mL of DOX-loaded micelles prepared in PBS was trans-
centrations. Values were reported as the means for each triplicate ferred into a dialysis bag (MWCO 3500), and dialyzed against 30 mL
samples. For structural characterization, solutions inside and of PBS (pH 7.4 or 6.5) with or without 10 mM of GSH at 37  C with
outside the dialysis bag were collected and lyophilized for 1H NMR shaking. The release media also contains 100 mM of sodium sa-
measurements. licylate to solubilize DOX and maintain a sink condition. At pre-
To determine the change of molecular weight before and after determined frequencies, 3 mL of release media was taken out and
stimuli-responsive degradation, 10 mL of SSPHPU micellar solution replenished with an equal volume of fresh media. The content of
(2 mg mL1) was incubated under different conditions (pH 7.4; pH DOX in the media was measured by UVeVis spectrometer. The
5.0; 100 mM GSH; pH 5.0 and 100 mM GSH) for 48 h with shaking release experiments were conducted in triplicate.
(110 r min1). The solutions were freeze-dried for GPC analysis. To evaluate the release of SPIONs from MPU micelles, 2 mL of
142 J. Wei et al. / Biomaterials 145 (2017) 138e153

SPION@SSPHPU was incubated at pH 5.0 or in the presence of interest (ROI), and the relaxivity values were calculated by linear
100 mM GSH with shaking at 37  C for 48 h. The solution was least-squares fitting of 1/T2 versus the iron concentration (mM Fe).
allowed to stand for 12 h. Then images were recorded with a color
digital camera, and the supernatant was collected for turbidity 2.8. Animals and tumor model
measurement by UVeVis spectrometer at 650 nm.
All animal procedures were approved by the Ethics Committee
2.5. Cellular uptake and intracellular delivery of Sichuan University, in conformity with the Principles of Labo-
ratory Animal Care of the National Institutes of Health for the care
HeLa cells obtained from West China Medical Center of Sichuan and use of laboratory animals. 5e6-week-old female athymic nude
University were cultured in RPMI 1640 medium (HyClone, U.S.A.) mice were purchased from the Laboratory Animal Center of Sichuan
containing 10% fetal bovine serum (FBS, Gibco Life, U.S.A.) and 5% University (Sichuan, China). All the animals were kept under
penicillin-streptomycin (Gibco Life, U.S.A.) in 5% CO2 at 37  C. To pathogen-free conditions according to AAALAC guidelines and
evaluate the effect of corona-detachment behavior on cellular up- acclimatized for 1 week prior to any experiments. Each mouse
take, HeLa cells were cultured in a six-well plate (with a clean received subcutaneous injections of 1  107 HeLa cells in the upper
coverslip placed in each well before use) at a density of 1  105 cells flank of right hind leg. To evaluate magnetic-guided targeting, mice
per well for 24 h. The pH of the culture media was adjusted to 6.5 or were inoculated with two tumors in the left and right back of the
7.4 before treatment. Then 1 mL of DOX-loaded MPU micelles hind leg region.
containing 10 mg mL1 DOX were added and cultured for 4 h. The
supernatant was carefully removed and the cells were washed with 2.9. In vivo MR imaging
PBS for three times. The cells were fixed with 1 mL of 4% formal-
dehyde for 30 min and stained with DAPI for 10 min. Finally, the When the tumor had grown to approximately 100 mm3 in
coverslip was mounted with 50% glycerol solution and observed volume, the nude mice bearing HeLa tumors were intravenously
with a confocal laser scanning microscope (CLSM, Leica TCS CP5). injected with SPION@SSPHPU-FA at a dose of 4.3 mg Fe kg1. Af-
The mean signal intensity (SI) of DOX fluorescence in cytoplasm terward, the right tumor was exposed to an external magnet
and nucleus was calculated and normalized with ImageJ 1.50i (Grade: N38) during the experiment while the left one was not. T2-
(Wayne Rasband, National Institutes of Health, USA). For flow weighted MR images were acquired under 3% isoflurane anesthesia
cytometry, HeLa cells were cultured in a culture dish at a density of before and after micelle administration for 1, 3 and 7 h using a
1  106 cells per dish for 24 h. After pre-adjusting the culture media Bruker BioSpec 7.0 T MRI scanner (Bruker BioSpin, Billerica, MA,
to pH 6.5 or 7.4, 1 mL of DOX-loaded micelles containing 10 mg mL1 USA). The mice were fixed with a surface coil in the MRI scanner.
DOX were added and incubated for 4 h. The suspension was dis- MR images were obtained using a fast spin echo sequence (TURBO-
carded and the cells were washed with PBS for three times. Then RARE) with the following parameters: TR/TE ¼ 1500/33 ms, slice
the cells were digested by trypsin, centrifuged and re-suspended in thickness ¼ 1 mm, flip angle ¼ 180 , matrix size ¼ 256  256 mm2,
0.5 mL of PBS, and analyzed using a flow cytometer (Beckman FOV ¼ 50  50 mm2, NEX ¼ 4. T2-weighted datasets were processed
cytoflex). with ImageJ 1.50i (Wayne Rasband, National Institutes of Health,
To further study the influence of redox-responsiveness on USA). The signal intensity of tumor was normalized to that of
intracellular delivery, DOX-loaded MPU micelles (SSPHPU or PHPU) muscle at each time point. Relative MRI SI was defined as:
were incubated with HeLa cells for 24 h. Then the medium was Normalized SI of tumor (post-injection)/ Normalized SI of tumor
removed, and the cells were washed three times with PBS. The (pre-injection)  100%, and T2 signal contrast effect enhancement
samples were fixed, stained, mounted and tested as described was defined as [Normalized SI of tumor (pre-injection) - Normal-
above. ized SI of tumor (post-injection)]/ Normalized SI of tumor (pre-
injection)  100%. Following MR imaging, mice were sacrificed and
2.6. MTT assay tumors were removed and stained with Prussian blue for histo-
logical examination of iron distribution. Quantitative analysis of the
The cytotoxicity of DOX-loaded micelles and blank micelles Prussian blue staining images was conducted with Image-Pro Plus
against HeLa cells were evaluated in vitro by an MTT assay. HeLa 6.0 software (Media Cybernetics, Silver Spring, US) based on the
cells were incubated in 96-well plates at a density of 1  105 cells mean optical density.
per well in RPMI 1640 media for 24 h, followed by replacing the
media with 100 mL of micellar solutions with different concentra- 2.10. In vivo antitumor effect and safety studies
tions of DOX. The treated cells were incubated for 24 h and 72 h.
Then 20 mL of MTT solution (5 mg mL1) was added into each well. When the average tumor volume approximately reached
After another 4 h of incubation, the medium was replaced by 200 mL 30e50 mm3, nude mice bearing single HeLa tumor models were
of DMSO. The plates were shaken for 10 min to dissolve the for- randomly divided into four groups (n ¼ 5), and treated with free
mazan crystals. The absorption intensity at 490 nm was recorded DOX, DOX/SSPHPU-FA, and DOX/SSPU-FA at a DOX dose of
on a microplate reader (DNM-9602, Nanjing Perlove Medical 4 mg kg1 via tail vein every 3 d for 12 d. Mice injected with saline
Equipment Co., Ltd., China). solution were set as negative controls. To demonstrate magnetic
targeting, mice bearing two HeLa tumors were divided into two
2.7. In vitro MR imaging groups, and each group was injected with DOX/SPION@SSPHPU-FA
and DOX/SPION@PHPU-FA, respectively, with an external magnet
The MRI experiments were carried out at 25  C using a 1.5 T (Grade: N38) placed on the right tumor for 1 h immediately after
clinical MRI instrument (Siemens, Munich, Germany). The every injection. The body weights and tumor growth of the mice
following parameters were set in the measurement: repetition time were monitored every 3 d. The length (A) and width (B) of tumors
(TR) was 5000 ms, multiple echo times (TE) were 10, 20, 30, 40, 50, were measured by a digital calliper, and the tumor volume was
60, 70, 80, 90, 100, 130, 150, 170 or 200 ms, slice thickness was calculated from the following equation: AB2/2. On the day of 15, all
1.5 mm, field of view (FOV) was 150 mm  300 mm. The transverse the mice were sacrificed and the tumors were excised and weighed.
relaxation time (T2) was obtained from signal intensity in regions of The tumor weight inhibition rate (TWI) was calculated through the
J. Wei et al. / Biomaterials 145 (2017) 138e153 143

formula: (the average tumor weight of control group e the average 3.2. Construction of clickable and imageable multiblock polymer
tumor weight of experiment group)/ the average tumor weight of micelles
control group  100%.
Thereafter, the tumor tissues and main organs including heart, The prepared MPUs could self-assemble into a core-shell
liver, spleen, lung and kidney of the mice were harvested and used nanostructure, where insoluble PCL soft segment constitutes a
for histological analysis. To analyze the tumor tissue apoptosis, the hydrophobic core, surrounded by a cleavable corona formed by
terminal deoxynucleotidyl transferased dUTP nick end labeling BPEG. The hard segments comprising LDI and Cys-PA tend to
(TUNEL) assay was conducted. Briefly, the removed tumor tissues distribute on the subsurface of nanoparticles due to their relatively
were fixed in 4% paraformaldehyde solution and embedded and higher polarity compared with PCL and the poor compatibility
sliced, which were then incubated with the TUNEL reaction between the soft segments and hard segments [34]. The nano-
mixture freshly prepared according to the manufacturer's protocol vehicles appear as dispersed spherical particles with diameters
(Roche, Penzberg, Germany) for 60 min at 37  C in a humidified around 57 nm, as determined by DLS and TEM (Fig. 2A and B). The
atmosphere in the dark. The tissue sections were labeled with DAPI formation of micelles was first confirmed by using the fluorescence
and observed with fluorescence microscopy. The apoptotic rate of of pyrene as a probe. As the concentration of MPU increases, the
tumor sections was calculated with Image-Pro Plus 6.0 software fluorescent intensity of pyrene rises, and the (0, 0) band shifts from
(Media Cybernetics, Silver Spring, US) based on the mean optical 331 nm to 335 nm (Fig. S3). The result demonstrates the transfer of
density measured from three random fields per slice. Furthermore, pyrene molecules from a polar environment to a nonpolar micro-
the tumor tissues and the main organs were stained with hema- environment of micellar core [35]. The micellar structure was also
toxylin and eosin (H&E) for histological examination. characterized by 1H NMR. As seen in Fig. S4, the chemical shifts of
To evaluate the safety of MPU formulations, the blood samples all the components are evident in DMSO-d6 (Fig. S4A), while those
(0.8 mL) were collected from each group of mice at the end of the of PCL are significantly weakened and those of BPEG increase
experiment. Aliquots of plasma were obtained by centrifugation at remarkably in D2O (Fig. S4B), suggesting the formation of core-shell
3000 r min1 at 4  C for 10 min. The serum biochemical parameters assemblies. To visually clarify the structure of polymer micelles,
including alanine aminotransferase (ALT), blood urea nitrogen computational simulation was carried out using a dissipative par-
(BUN) and creatinine kinase (CK) were assessed as function ticle dynamics (DPD) model (Supporting Information). The results
markers of liver, kidney and heart, respectively. All these parame- present a spherical assembly structure with a PCL core and a BPEG
ters were measured with an IDEXX Biochemical Analyzer (West- corona, and the hard segments were found distributed mainly on
brook, ME, USA) using the kits according to the manufacturer's the interface (Fig. 2A inset), which is in agreement with our pre-
instructions. vious finding [36]. Such an architecture is attractive for integration
of targeting and stealth properties into a single nanosystem. One
the one hand, the clickable alkyne sites located on the subsurface of
2.11. Statistics
micelles enable post-conjugation of targeting ligands via a facile
click chemistry. On the other hand, the cleavable PEG corona could
The quantitative data collected were expressed as
provide temporary protection for the hard segments bearing di-
means ± standard deviations (SD). Statistical analysis was per-
sulfide and targeting moieties against destabilization and clearance
formed using the Statistical Package for the Social Sciences (IBM
from the body (Fig. 1).
SPSS Statistics software, Version 19, IBM, New York, USA). Student's
To construct imageable multiblock polymer micelles, SPIONs
t-test or one-way analysis of variance (ANOVA) was conducted to
were prepared by a facile and efficient chemical co-precipitation
evaluate the statistical significance within the data at 95% confi-
method and encapsulated into the micellar core through a dial-
dence levels (p < 0.05).
ysis process. Due to the hydrophobic nature of oleic acid coating,
the SPIONs are well dispersed in hexane with spherical morphology
3. Results and discussion and an average size of 16.3 nm, as determined by DLS and TEM
(Fig. 2C and D). HRTEM image presents a single crystallite of SPION
3.1. Synthesis of clickable MPUs (Fig. 2D inset and Fig. S5), where the lattice planes are clearly
visible. The measured d-spacing of 2.53 and 2.96 Å correspond to
A series of clickable MPUs with controlled responsiveness were the (311) and (220) planes of magnetite, respectively. After loading
successfully synthesized from biodegradable PCL, L-lysine derivat- into the core of polymer micelles, the SPIONs are transferred into
ized LDI, cleavable BPEG bearing a pH-sensitive benzoic-imine aqueous phase with high stability due to the surface protection
linkage, and a clickable and multifunctional chain extender from amphiphilic polymers (Fig. 2C). The average hydrodynamic
generated from L-cystine. The structure of the polymers is shown in particle size of SPION@micelles is 95 nm with a polydispersity in-
Fig. 1A. By appropriately varying the feed ratios, the amounts of dex of 0.19 (Fig. 2C), which is nearly twice the diameter of blank
labile disulfides and benzoic-imine linkages in the polymers were micelles, probably because the encapsulation of solid SPIONs in-
well adjusted. Typically, MPU containing both disulfide and creases the volume of micellar core. The morphology of SPION@-
benzoic-imine linkages is named as SSPHPU, while those including micelles observed by TEM is presented in Fig. 2D. The
only benzoic-imine or reductive disulfide bonds are denoted as SPION@micelles display a spherical shape consisting of Fe3O4
PHPU or SSPU, respectively. The polymers possess moderate mo- clusters, which is highly beneficial to the enhancement of imaging
lecular weight (Mw 14600e18800), with relatively narrow molec- contrast [37].
ular weight distributions (Fig. S1). The structure of MPU was To further investigate the structure and composition of
verified by 1H NMR. As depicted in Fig. S2, the characteristic peaks SPION@micelles, FTIR, XRD and TGA analysis were conducted tak-
of PCL (3.94, 2.25, 1.52, 1.27 ppm), PEG (3.51e3.30 ppm) and LDI ing SPION and blank micelles as controls. As shown in Fig. S6, a
(4.05, 1.15 ppm) can be observed for all the samples. The signals of strong absorption at 563 cm1 in the FTIR spectra of SPION is
Cys-PA (3.13, 2.78 ppm) and BPEG (8.30e7.88 ppm) can be found in attributed to FeeO vibration in Fe3O4. This peak can be observed in
the spectra of SSPU and PHPU, respectively, confirming that the the spectra of SPION@micelles and appears greatly weakened,
designed functional moieties have been successfully introduced which may be due to the encapsulation of SPIONs in polymer mi-
into the multiblock polymers. celles. Fig. S7 shows the TGA thermograms of SPIONs and
144 J. Wei et al. / Biomaterials 145 (2017) 138e153

Fig. 2. Construction and characterization of clickable and imageable MPU micelles. Size distributions (A, C, E) and TEM micrographs (B, D, F) of SSPHPU micelles (A, B), SPIONs
(C, D) and SPION@micelles (E, F). The bars in (B, D, F) are 100 nm. The inset in (A) shows a representative computational simulation image of MPU micelles. The insets in (C) and (E)
present SPIONs and magnetic micelles dispersed in hexane and water, respectively. The inset in D indicates HRTEM image of a single crystallite of SPION showing the (311) and (220)
lattice fringe at 2.53 and 2.96 Å, respectively (the bar is 2 nm). A high resolution figure of HRTEM image is depicted in Fig. S5.

SPION@micelles. The weight loss of 12% in the range of 200e400  C 3.3. Corona-detachment profiles of multiblock polymer micelles
is associated with the decomposition of oleic acid on Fe3O4. For
SPION@micelles, 75.7% weight loss in the range of 200e450  C can The MPU micelles exhibit good responsiveness to tumor mi-
be attributed to the degradation of oleic acid and MPUs (Fig. S7). croenvironments. With the change of solution pH from 7.4 to 6.5,
Based on the residual weight percentages, the SPION loading con- the micellar diameter increases from 57 nm to approximately
tent in the magnetic micelles was calculated to be 27.6% 130 nm, and the size distribution is significantly broadened
(250 mg mL1), with a loading efficiency near 60%. The XRD pattern (Fig. 3A). Moreover, the particle surface turns somewhat rough
of SPION and SPION@micelles are depicted in Fig. S8. The SPION (Fig. 3B), which is consistent with a previous report [31]. This
exhibits diffraction peaks at 2q of 30.2 , 35.7, 43.3 , 54.2 , 57.2 , phenomenon is expectable since corona detachment may decrease
62.9 , 74.5 , corresponding to the (220), (311), (400), (422), (511), the density of PEG on micellar surface, and thus reduce the steric
(440), (533) reflection planes of the face-centered cubic (fcc) Fe3O4 repulsive forces against van der Walls interactions between
crystal [38]. The XRD pattern of the magnetic loaded polymer mi- exposed micellar cores [39]. As a result, a secondary aggregation
celles is similar to that of SPIONs, further indicating that the pre- would be favored in order to minimize the interfacial energy [40].
pared micelles are composed of Fe3O4 nanoparticles. The aggregation of polymer micelles and increase of particle size
J. Wei et al. / Biomaterials 145 (2017) 138e153 145

were also visually proved by computational simulation results, resonance peaks corresponding to the protons of PEG (3.51,eCH2e)
where a core-shell structure of micelles is still retained with a and benzene ring (7.80e8.10,eCHe) are clearly observed in the
1
larger diameter after partial detachment of outer corona (Fig. 3A HNMR spectra of release media, while these signals decline or
inset). even disappear for samples inside the dialysis bag (Figs. S4 and
To further demonstrate the impact of corona-detachment on S10). These results demonstrate that the MPU micelles display
micellar structure, the morphology change of SPION-loaded MPU high stability under physiological conditions and pH-triggered
micelles in response to an extracellular acidic environment was shell-detachment behavior in response to tumor extracellular
investigated. It was found that the size and size distribution of environment.
SPION@micelles increase moderately after acid treatment (Fig. 3C).
Interestingly, the magnetic nanoparticles still form stable clusters
and remain well dispersed in an aqueous phase (Fig. 3D), further 3.4. Triggered release of payloads from multiblock polymer micelles
confirming that the MPU micelles could maintain their core-shell
structure to accommodate SPIONs after partial shell-detachment. Since the change of micellar structure can affect the drug release
This result is potentially helpful for nanomedicines to expose behavior, we investigated the release of a model drug, DOX, from
their targeting ligands while avoiding premature drug release at MPU micelles under different stimuli in vitro. The micelles can
tumor sites. encapsulate DOX efficiently, with a loading content up to 23%
To study the kinetics of corona-detachment, SSPHPU micelle (230 mg mL1). The DOX-loaded SSPHPU micelles display a slow and
solution was transferred into a dialysis bag and incubated in a weak sustained drug release rate in PBS solution (pH 7.4), with approx-
acid media (pH 6.5) with shaking. As the labile benzoic-imine imately 40% of the total DOX released within 96 h. While in a weak
bonds are cleaved in weak acid, the yielded BPEG residue, i.e. acid PBS solution (pH 6.5) mimicking the extracellular environ-
MPEG-CBA, is capable of penetration through the dialysis mem- ment, the release of DOX from SSPHPU micelles increases slightly
branes, which shows a characteristic UV absorption at 256 nm and (Fig. 4D), owning to the detachment of PEG corona and increase of
allows for a facile quantitative determination with UVeVis spec- particle diameter (Fig. 3). Nevertheless, the total amounts of
trometer (Fig. 4A and Fig. S9). The determined cumulative PEG released DOX is still less than 50%. A possible reason is that the
concentration in acid release media increases remarkably over micelles may preserve a core-shell architecture to accommodate
time. By contrast, there is no noticeable PEG derivative detected in drugs after partial detachment of outer corona, as has been
neutral media (Fig. 4B). To further verify the corona-detachment, demonstrated above. This phenomenon is advantageous to evasion
the solutions outside and inside the dialysis bag were collected of premature extracellular release of therapeutics [29].
and lyophilized for 1HNMR analysis. As shown in Fig. 4C, the On the other hand, the release of DOX from SSPHPU in the
presence 10 mM of GSH was found accelerated mildly (P ¼ 0.053),

Fig. 3. Corona-detachment property of MPU micelles. Size distributions (A, C) and TEM micrographs (B, D) of SSPHPU micelles (A, B) and SPION@micelles (B, D) after incubation at
pH 6.5 for 24 h. The bars in both TEM images are 100 nm. The inset in (A) shows a representative computational simulation image of MPU micelles after partial corona detachment.
The insets in (C) presents SPION@micelles dispersed in hexane and water after acidic treatment.
146 J. Wei et al. / Biomaterials 145 (2017) 138e153

Fig. 4. Corona-detachment and triggered release profiles of MPU micelles. (A) UVevis spectra of MPEG-CBA in aqueous solutions at different concentrations. (B) Time-
dependent cumulative release of PEG outer corona from SSPHPU micelles at different pH values. (C) 400 MHz 1H NMR spectrum of lyophilized acidic release media in DMSO-
d6. (D) The release profiles of DOX from SSPHPU micelles in different media with or without acidic (pH 6.5) and reductive (10 mM GSH) stimuli.

with nearly 65% of drug released in 96 h (Fig. 4D). Such an possibility of disulfide breakage occurred in the blood circulation
enhancement of drug release should not be as significant as ex- has been noticed elsewhere [24,25], the “PEG switch” strategy is
pected, considering the dramatic increase of micellar diameter in promising for remitting undesirable polymer degradation and drug
response to GSH [31] and relatively higher responsivity reported for leakage on the way of nanomedicines to tumor site, and triggering
other disulfide-based drug delivery systems [41]. From a structural the cleavage of redox-sensitive moieties and release of payloads
perspective, the disulfide bonds are mainly distributed in the hy- within tumor cells. To better understand the pH-activated
drophobic hard segments of PUs, which are further shielded by the responsiveness and release behavior, further work is needed and
soft segments comprising PEG and PCL during assembly. The being carried out in our group.
presence of soft segments may promote a steric repulsion against
the penetration of GSH and inhibit the responsive efficiency of 3.5. Cellular uptake of FA-clicked MPU micelles
polymers [42]. In addition, the steric hindrance of propargyl amide
groups should also be taken in to account [43]. This hypothesis To improve the specificity of clickable MPU micelles, FA as a
could be partially confirmed by GPC test, where SSPHPU does not model targeting ligand was chemically attached onto the micellar
show significant decrease in molecular weight after GSH treatment, interface via a copper catalyzed alkyne-azide cycloaddition
whereas it can be greatly degrade as acid and GSH exist simulta- (CuAAC). The FA-conjugated MPU micelles (SSPHPU-FA) hold an FA
neously (Fig. S11). To further understand the result, the release amount of 3.1 w/w%, as determined by UVeVis spectrometer. The
experiment was conducted in an acid media containing 10 mM of critical micelle concentration (CMC) of MPU increases slightly after
GSH. As anticipated, the release of DOX from SSPHPU was greatly FA conjugation (Fig. S13). To investigate the effect of “PEG-switch”
accelerated (P ¼ 0.003), with almost 100% of the loaded drugs on the interaction between MPU micelles and cancer cells, the DOX-
released (Fig. 4D). loaded targeted and untargeted formulations were incubated with
In addition, the release of SPIONs from MPU micelles was also HeLa cells, and the cellular uptake of nanocarriers was determined
evaluated using a turbidity measurement, considering the poor using CLSM and flow cytometry. As shown in Fig. 5, SSPHPU mi-
permeability of solid Fe3O4 nanoparticles through the dialysis bag. celles exhibit a low degree of internalization into tumor cells
As seen from Fig. S12, the absorption of SPION@SSPHPU decreases cultured at pH 7.4, with weak fluorescence observed predomi-
moderately under acidic or reductive conditions. By contrast, in the nantly in the cytoplasm after 4 h of incubation (Fig. 5A, E). After
presence of both acid and GSH stimuli, the turbidity drops conjugation of FA, the nanocarriers show a mild increase of uptake,
dramatically and visible participates appear at the bottom of vials owning to folate receptor-mediated endocytosis causing more
(Fig. S12). This result is in good agreement with DOX release pro- micelles to be taken up by tumor cells [44]. However, it is note-
files, suggesting a high specificity of SPION release from MPU mi- worthy that such a promotion of cellular uptake is relatively lower
celles in responsive to intracellular microenvironments. Since the than those attained for other FA-based nanomedicines [45]. We
J. Wei et al. / Biomaterials 145 (2017) 138e153 147

previously speculate that the short spacer of FA residues and the (SSPHPU-FA) present significantly increased DOX fluorescence in
long chain PEG corona should be responsible for the decreased tumor cells (Fig. 5D and E). This result could be explained by the
activity [31]. Herein, to validate this hypothesis, the cellular uptake fact that the deshielding of PEG shell can expose FA-decorated
experiments were carried out under a simulated extracellular micellar surface to receptor-enriched cells, thus turning the tar-
acidic conditions (pH 6.5). Remarkably, the targeted MPU micelles geting switch “on” and facilitating subsequent cell internalization

Fig. 5. Cellular uptake of FA-clicked MPU micelles. CLSM images of HeLa cells incubated with SSPHPU-DOX (A, B) and FA-clicked MPU micelles (SSPHPU-FA-DOX) (C, D) at pH 7.4
(A, C), and pH 6.5 (B, D) for 4 h. Nuclei of cells were stained with DAPI. The scale bars are 25 mm. (E) The mean SI of DOX fluorescence in cytoplasm and nucleus calculated and
normalized with an ImageJ Software. Statistical significance: *P < 0.05; **P < 0.01; ***P < 0.005. (F) Flow cytometry histograms of HeLa cells incubated with various formulations,
taking untreated cells as controls. (a), (b), (c) and (d) in (E, F) represent cells treated with SSPHPU-DOX at pH 7.4, SSPHPU-DOX at pH 6.5, SSPHPU-FA-DPX at pH 7.4, and SSPUPU-FA-
DOX at pH 6.5, respectively.
148 J. Wei et al. / Biomaterials 145 (2017) 138e153

(Fig. 1E). On the other hand, unconjugated polymer nanocarriers nanovehicles are excellent candidates for achieving on-demand
(SSPHPU) are also able to remove their corona in acid media, tumor cell targeting and controlled drug delivery.
nonetheless, the lack of targeting groups results in an insufficient
uptake, as evidenced by the negligible enhancement of fluores-
cence detected in tumor cells (Fig. 5B). The same phenomenon was 3.6. Intracellular drug delivery and antitumor activity
also found in flow cytometry result, where the cells treated by
SSPHPU-FA under pH 6.5 display the highest fluorescence intensity It is known that tumor cells develop unique microenvironments
(Fig. 5F). These results demonstrate that the targeting-clicked MPU such as slightly acidic (pH 4e6) [46] and markedly reductive (up to
10 mM of GSH) intracellular space [47]. Therefore, the clickable

Fig. 6. Intracellular drug delivery and antitumor activity in vitro. CLSM images of HeLa cells incubated with DOX-loaded redox-responsive SSPHPU (B) and insensitive PHPU (A)
for 24 h. Nuclei of cells were stained with DAPI. The scale bars are 25 mm. (C) The mean SI of DOX fluorescence in cytoplasm and nucleus calculated and normalized with an ImageJ
Software. Cytotoxicity of redox-responsive SSPHPU-DOX and insensitive MPU micelles against HeLa cells for 24 h (D) and 72 h (E) of incubation. Free DOX was used as a positive
control. (F) Cytotoxicity of SPION@micelles without drug loading at different concentrations.
J. Wei et al. / Biomaterials 145 (2017) 138e153 149

multifunctional PU micelles equipped with a “PEG switch” are significant toxic effect at different concentrations (Fig. 6F). These
promising for triggered breakage of disulfide bonds and maximal results demonstrate that the MPU micelles with switchable release
release of payloads within tumor cells. To prove this potential, the capacity hold great promise in multifunctional intracellular drug
MPU formulations were incubated with HeLa cells for 24 and 72 h. delivery applications.
As seen in Fig. 6AeC, the cells incubated with redox-responsive
SSPHPU show a strong DOX fluorescence located in the nucleus. 3.7. Magnetic property and MRI contrast effects of MPU micelles
In contrast, the signal in cells cultured with insensitive PHPU
formulation was detected mainly in the cytoplasm. This phenom- The superparamagnetic property is important to biomedical
enon can be attributed to the triggered release of DOX in response applications of SPION@micelles. Under an external magnetic field,
to the acidity and GSH inside tumor cells. The free drugs released the SPION-loaded micelles can be manipulated and guided to
from the carriers could be readily transported into the cell nucleus, specific sites (Fig. 7A). On the other hand, the SPION@micelles lose
where it can interfere the process of DNA replication and cause cell magnetism and re-disperse when the magnetic field is turned off.
death [48]. With this in mind, we further assessed the antitumor This phenomenon can be exploited in magnetic targeted drug de-
effects of DOX-loaded MPU micelles using an MTT assay. It was livery, cell sorting and separation, as well as imaging agents for
found that all the formulations display a time and dose dependent early detection of several pathologies. The magnetic hysteresis
cytotoxicity toward HeLa cells (Fig. 6D and E). Apparently, the drug loops of SPIONs and SPION@micelles were determined using a
efficacy of SSPHPU micelles is significantly higher compared with vibrating-sample magnetometer. As shown in Fig. 7A, both
insensitive PHPU and SSPU formulations (P < 0.05), with IC50 (50% SPION@micelles and SPIONs are superparamagnetic at room tem-
inhibitory concentration) values of 2.2 mg mL1 and 1.7 mg mL1 perature without magnetic remnants observed at a small applied
after incubation for 24 h and 72 h, respectively, which are two fold magnetic field. The saturation magnetization value of SPION-
lower than those of PHPU (13.0 and 4.0 mg mL1) and SSPU (4.6 and loaded micelles is 21.0 emu g1, smaller than that of Fe3O4 nano-
3.1 mg mL1). Besides, the blank SPION@micelles do not show particles (80 emu g1). This can be attributed to the nonmagnetic

Fig. 7. Magnetic property and MR imaging of MPU micelles in vitro and in vivo. (A) Magnetic hysteresis loops of SPIONs and SPION@micelles. The insets show magetic loaded
MPU micelles in response to an external magnetic field for 0 h (upper) and 6 h (lower). (B) T2-weight MRI images of SPIONs (I) and SPION@micelles (II) recorded on a 1.5 T clinical
MRI instrument at different Fe concentrations (mM): a, 0; b, 0.05; c, 0.1; d, 0.2; e, 0.3; f, 0.4; g, 0.5. (C) T2 relaxation rates as a function of Fe concentrations. In vivo T2-weighed MR
images of HeLa tumor-bearing mice before (D) and after intravascular injection of MPU micelles loading with SPIONs for (E) 1 h, (F) 3 h, and (G) 7 h. (H) Optical images of a
representative mouse inoculated with two tumors for MRI. The tumors in the left and right flanks are marked with green dots and red circles, respectively. The magnetic force was
applied to the right tumor. (I) Relative T2 signal intensity of tumors as a function of time. The standard deviation of signal intensity is derived from five random ROI in tumor
sections. Statistical significance: *P < 0.05; **P < 0.01; ***P < 0.005. Histological analysis of the right tumor (J) and the left tumor (K) after Prussion blue staining. The scale bars are
10 mm. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
150 J. Wei et al. / Biomaterials 145 (2017) 138e153

polymer coating reducing the total magnetization. However, the greatly enhanced by the introduction of SPIONs, which are known
absence of coercivity and remanence reveals that SPIONs remain to shorten the transverse (spin-spin) relaxation time (T2) of water
their superparamagnetic nature after encapsulation into MPU protons and can exert negative contrast in the interfered area [49].
micelles. To evaluate the MRI contrast effect rendered by SPION@micelles,
MR imaging is one of the most useful clinical diagnostic tech- the transverse relaxation rates (1/T2) and T2-weighted MR images
niques in providing non-invasive and real-time detection of various of SPION@micelles with different iron concentrations were ob-
diseases including tumors. The capabilities of MRI have been tained on a clinical 1.5 T MRI instrument. As presented in Fig. 7B

Fig. 8. Multimodal targeting and antitumor effect in vivo. (A) Optical images of representative mice inoculated with two tumors, where the left and right tumors are marked
with green dots and red circles, respectively. The right tumors were exposed to an external magnetic field for magnetically guided therapy (as shown by red curves). (B) Growth
curves of the left tumors (a, c) and right tumors (b, d) after intravenous injection of DOX/SPION@SSPHPU (a, b) and DOX/SPION@SSPU in HeLa tumor-bearing mice (c, d) at a DOX
dose of 4 mg kg1. (C) The average weights of tumors separated from animals receiving different treatments. The insets show representative images of tumors. Statistical sig-
nificance: *P < 0.05; **P < 0.01; ***P < 0.005. (D) Ex vivo histological analyses of tumor sections after treatment with DOX/SPION@SSPHPU. H&E staining (a, b) and TUNEL
immunofluorescence staining (c, d) of slices from the left tumors (a, c) and right tumors (b, d). In TUNEL analysis, the nuclei were labeled blue, and the apoptotic cells were stained
green. The scale bars are 500 mm. (E) H&E staining analysis of the major organs (heart, liver, spleen, lung, kidney) of the mice after different treatments. The scale bars are 100 mm.
(For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
J. Wei et al. / Biomaterials 145 (2017) 138e153 151

and C, the signal intensity of T2-weighted MR images deceases with To further test magnetic guided targeting and therapy in vivo,
increasing of iron concentration. Such a concentration-dependent SPIONs and DOX were co-encapsulated into SSPHPU-FA and SSPU-
darkening phenomenon demonstrates a negative contrast FA micelles, and injected into the nude mice bearing two tumors. As
enhancement in MRI. Particularly, the magnetic micelles display expected, a much greater suppression on the growth of right tu-
higher efficiency in signal darkening than SPIONs (Fig. 7B). As an mors was observed for both SSPU-FA and SSPHPU-FA formulations,
indicator of the ability of contrast agent to shorten T2, the specific with the aid of an external magnet (Fig. 8A and B). Moreover, the
relaxivity rate (r2) of SPION@micelles (89.5 mM1 s1) is much PEG-switched targeting effect is still obvious in the presence of
higher than that of SPIONs (54.6 mM1 s1) (Fig. 7C), owning to the MDT, as evidenced by the higher therapeutic efficacy of DOX/
fact that the physical encapsulation of clustered SPIONs into SPION@SSPHPU-FA compared with that of SSPU-FA formulation
confined polymeric micelles can increase the T2 relaxivity and (Fig. 8). Excitingly, a synergistic targeting effect could be antici-
contrast effect of MRI [37,50]. This clustering effect has also been pated for PEG-triggered active targeting in combination with
demonstrated in many other carrier systems [51e53]. magnetically-guided targeting, considering the excellent thera-
peutic efficacy of FA-clicked SSPHPU micelles toward the right tu-
3.8. Magnetically guided targeting and MR imaging in vivo mor with magnetic exposure (TWI is up to 98.6%, Fig. 8C).
The enhanced anticancer activity of MPUs was further evaluated
The incorporation of magnetic nanoparticles is beneficial for by histological analysis with H&E and TUNEL staining. It was
magnetically targeted drug delivery as well as excellent contrast observed that the DOX and SSPU-FA groups exhibit moderate
effect for MR imaging [11]. To evaluate the MDT and MRI ability of cancer cell remission in comparison with saline treated groups
MPU formulations, nude mice were inoculated with two tumors in (Fig. S14). The sections from targeted SSPHPU-FA group display
the left and right back of the hind leg region, and administrated greater extent of necrosis, and the area of the damaged regions was
with SPION-loaded SSPHPU-FA micelles through the tail vein at a found much larger for right tumors receiving magnetic treatment
dose of 4.3 mg Fe kg1, with the right tumor exposed to an external (Fig. 8D). Furthermore, the quantitative analysis of apoptotic cells
magnetic field after injection. The T2-weighted axial MR images of by TUNEL staining assay also confirms the above observations. The
the mice were acquired on a 7.0 T MRI instrument before and after percentage of apoptotic cells in the tumor treated by DOX/
treatment at different time points. As shown in Fig. 7DeG, SPION@SSPHPU-FA with magnetic treatment is about 92.1%,
remarkable signal attenuation was observed at both the left and which is much higher than those of other groups without MDT
right tumors after 1 h, and the MRI signal intensity further de- (54.0%) and PEG trigger (26.2%) (Fig. S14). In view of the exciting
creases over time. In particular, compared with the left tumors with delivery efficiency and therapeutic effect, further study using a
passive EPR effect and FA-mediated active targeting for MPU de- single injection is warranted to demonstrate the potential of MPU
livery, the right tumors with additional magnetic exposure exhibit micelles.
much faster and higher degree of signal darkening. The T2 signal It is worth noting that no mice died during the treatment period
contrast effect enhancement at 1 h in the right tumors (60%) is due to the relative low dose (Fig. S15), nonetheless, severe weight
more than two times higher than that in the left tumors (27%) loss was noticed in mice treated with DOX (Fig. S14B), indicating
(Fig. 7I). systemic toxicity of free drug without any targeting ability. To
To confirm the accumulation of MPU formulations and assess determine the potential toxic effect, we further tested the
the concentration of intratumoral Fe, the excised tumor tissues biochemistry parameters ALT, BUN and CK as function markers of
were stained with Prussian blue for observation. As indicated in liver, kidney and heart, respectively. Even though all the parameters
Fig. 7J, numerous dark blue regions are observed in the magneti- measured fell within the normal range (Fig. S16), the level of liver
cally treated tumors, in good agreement with the MR images. By enzyme ALT was found much higher in mice receiving DOX treat-
contrast, the contralateral tumors without magnet exposure ment, compared with other groups (Fig. S16A). Furthermore, cell
display much weaker blue signals (Fig. 7K). Quantitative analysis nuclei shrinkage was observed by H&E staining of liver tissues
reveals that the Prussian signal intensity in the right tumors is (Fig. 8E), revealing moderate hepatotoxicity in the mice treated
approximately 4.5 times higher than that in the left tumors. These with free DOX. In contrast, no significant dropping in body weights,
results demonstrate that magnetically-guided delivery of MPUs change of biochemistry parameters and abnormality of major or-
into tumors could largely complement the targeting efficacy gans was discovered in MPU groups during the experiment (Fig. 8E,
mediated by EPR effect, and show great potential to overcome the Figs. S14 and S16), suggesting that the MPU formulations are
tumor heterogeneity and complicated physiological environment. favorably biocompatible. These results demonstrate that the click-
Further advances in the development of high-strength magnetic able and imageable polymer micelles with smart switches and
field gradients and novel magnetic nanocomposites with multi- multimodal synergistic targeting ability would provide a novel
modal contrast capacity would enable the manipulation of MPU route for improving the specificity, efficacy and safety of cancer
nanomedicines at higher depths with greater efficiency [9,54]. theranosis. To further evaluate the efficacy and safety of MPU mi-
celles in vivo, dose response experiments and drug analysis in
3.9. Multimodal targeting and antitumor effect in vivo different organs are being carried out in our laboratory.

To investigate the targeting effect and antitumor efficacy of MPU 4. Conclusion


formulations in vivo, nude mice bearing HeLa tumors were treated
with DOX loaded SSPHPU-FA and SSPU-FA micelles, taking free In summary, a rationally designed clickable and imageable MPU
DOX and saline as controls. It was found that both DOX@SSPHPU-FA was successfully constructed for on-demand tumor targeting and
and DOX@SSPU-FA show remarkable inhibitory effects on tumor controlled drug release. The polymeric nanovehicles possess
growth in comparison with saline solution after intravenous in- attractive core-shell architecture, appropriate size, high loading
jection (Fig. S14). Particularly, the TWI of DOX@SSPHPU-FA (89.7%) capacity for DOX and SPIONs and clickable sites for functionaliza-
is much higher than that of DOX@SSPU-FA (57.9%). The results tion. Furthermore, they present time-dependent detachment of
confirms the significant role of “PEG switcher” in SSPHPU-FA, outer corona under extracellular acidic condition, which can switch
which enables improved cellular drug targeting and effective on the clicked FA ligands for activated tumor targeting and cellular
anticancer activity (Fig. 1E). uptake, followed by facilitating the cleavage of reducible core
152 J. Wei et al. / Biomaterials 145 (2017) 138e153

segments as well as triggered drug release within tumor cells. In [9] J. Park, N.R. Kadasala, S.A. Abouelmagd, M.A. Castanares, D.S. Collins, A. Wei, et
al., Polymereiron oxide composite nanoparticles for EPR-independent drug
addition, the versatile nanoformulations demonstrate not only
delivery, Biomaterials 101 (2016) 285e295.
potent antitumor activity and effective MRI contrast enhancement [10] H. Jeon, J. Kim, Y.M. Lee, J. Kim, H.W. Choi, J. Lee, et al., Poly-paclitaxel/
in vitro, but also magnetically guided and tumor-activated syner- cyclodextrin-SPION nano-assembly for magnetically guided drug delivery
gistic targeting ability in vivo, thus resulting in precise anticancer system, J. Control. Release 231 (2016) 68e76.
[11] W. Chen, G. Luo, Q. Lei, F. Cao, J. Fan, W. Qiu, et al., Rational design of
therapy and specifically enhanced MR imaging. Our work provides multifunctional magnetic mesoporous silica nanoparticle for tumor-targeted
a promising platform for the development of smart theranostic magnetic resonance imaging and precise therapy, Biomaterials 76 (2016)
systems for potential imaging-guided cancer treatment and real- 87e101.
[12] a) Y. Chen, K. Ai, J. Liu, X. Ren, C. Jiang, L. Lu, Polydopamine-based coordination
time detection of diseases. nanocomplex for T1/T2 dual mode magnetic resonance imaging-guided
chemo-photothermal synergistic therapy, Biomaterials 77 (2016) 198e206;
Acknowledgment b) J. Chen, M. Shi, P. Liu, A. Ko, W. Zhong, W. Liao, et al., Reducible
polyamidoamine-magnetic iron oxide self-assembled nanoparticles for
doxorubicin delivery, Biomaterials 35 (2014) 1240e1248.
This work was supported by the National Natural Science [13] M.M. Gottesman, T. Fojo, S.E. Bates, Multidrug resistance in cancer: role of
Foundation of China (21474064, 51203101); the National Science ATP-dependent transporters, Nat. Rev. Cancer 2 (2002) 48e58.
[14] a) Y. Yan, G.K. Such, A.P.R. Johnston, J.P. Best, F. Caruso, Engineering particles
Fund for Distinguished Young Scholars of China (51425305); the for therapeutic delivery: prospects and challenges, ACS Nano 6 (2012)
Project of State Key Laboratory of Polymer Materials Engineering 3663e3669;
(sklpme2015-3-02); and the Youth Science and Technology Inno- b) S. Lee, H. Chen, T.V. O Halloran, S.T. Nguyen, “Clickable” polymer-caged
nanobins as a modular drug delivery platform, J. Am. Chem. Soc. 131 (2009)
vation Team of Sichuan Province (2015TD0001). 9311e9320.
[15] C. Shi, X. Guo, Q. Qu, Z. Tang, Y. Wang, S. Zhou, Actively targeted delivery of
Appendix A. Supplementary data anticancer drug to tumor cells by redox-responsive star-shaped micelles,
Biomaterials 35 (2014) 8711e8722.
[16] S. Wang, P. Huang, X. Chen, Stimuli-responsive programmed specific targeting
Supplementary data related to this article can be found at http:// in nanomedicine, ACS Nano 10 (2016) 2991e2994.
dx.doi.org/10.1016/j.biomaterials.2017.08.005. [17] a) K. Knop, R. Hoogenboom, D. Fischer, U.S. Schubert, Poly(ethylene glycol) in
drug delivery: pros and cons as well as potential alternatives, Angew. Chem.
Int. Ed. 49 (2010) 6288e6308;
References b) X. Du, J. Wang, W. Liu, J. Yang, C. Sun, R. Sun, et al., Regulating the surface
poly(ethylene glycol) density of polymeric nanoparticles and evaluating its
[1] a) F. Alexis, E. Pridgen, L.K. Molnar, O.C. Farokhzad, Factors affecting the role in drug delivery in vivo, Biomaterials 69 (2015) 1e11.
clearance and biodistribution of polymeric nanoparticles, Mol. Pharm. 5 [18] a) E. Gullotti, Y. Yeo, Extracellularly activated nanocarriers: a new paradigm of
(2008) 505e515; tumor targeted drug delivery, Mol. Pharm. 6 (2009) 1041e1051;
b) F.M. Kievit, M. Zhang, Cancer nanotheranostics: improving imaging and b) X. Yang, J. Du, S. Dou, C. Mao, H. Long, J. Wang, Sheddable ternary nano-
therapy by targeted delivery across biological barriers, Adv. Mater. 23 (2011) particles for tumor acidity-targeted siRNA delivery, ACS Nano 6 (2011)
H217eH247. 771e781.
[2] a) O.C. Farokhzad, R. Langer, Impact of nanotechnology on drug delivery, ACS [19] S. Mura, J. Nicolas, P. Couvreur, Stimuli-responsive nanocarriers for drug de-
Nano 3 (2009) 16e20; livery, Nat. Mater. 12 (2013) 991e1003.
b) J.A. Barreto, W. O Malley, M. Kubeil, B. Graham, H. Stephan, L. Spiccia, [20] a) D. Roy, J.N. Cambre, B.S. Sumerlin, Future perspectives and recent advances
Nanomaterials: applications in cancer imaging and therapy, Adv. Mater. 23 in stimuli-responsive materials, Prog. Polym. Sci. 35 (2010) 278e301;
(2011) H18eH40; b) Y. Lu, A.A. Aimetti, R. Langer, Z. Gu, Bioresponsive materials, Nat. Rev.
c) A. Schroeder, D.A. Heller, M.M. Winslow, J.E. Dahlman, G.W. Pratt, R. Langer, Mater. 1 (2016) 16075.
et al., Treating metastatic cancer with nanotechnology, Nat. Rev. Cancer 12 [21] F. Meng, W.E. Hennink, Z. Zhong, Reduction-sensitive polymers and bio-
(2012) 39e50; conjugates for biomedical applications, Biomaterials 30 (2009) 2180e2198.
d) Y. Zhang, H.F. Chan, K.W. Leong, Advanced materials and processing for [22] H. Wei, R. Zhuo, X. Zhang, Design and development of polymeric micelles with
drug delivery: the past and the future, Adv. Drug Deliv. Rev. 65 (2013) cleavable links for intracellular drug delivery, Prog. Polym. Sci. 38 (2013)
104e120; 503e535.
e) H. Hosoya, A.S. Dobroff, W.H.P. Driessen, V. Cristini, L.M. Brinker, [23] a) J. Li, X. Yu, Y. Wang, Y. Yuan, H. Xiao, D. Cheng, et al., A reduction and pH
F.I. Staquicini, et al., Integrated nanotechnology platform for tumor-targeted dual-sensitive polymeric vector for long-circulating and tumor-targeted
multimodal imaging and therapeutic cargo release, Proc. Nat. Acad. Sci. U. S. siRNA delivery, Adv. Mater. 26 (2014) 8217e8224;
A. 113 (2016) 1877e1882. b) M. Huo, J. Yuan, L. Tao, Y. Wei, Redox-responsive polymers for drug de-
[3] a) C.E. Ashley, E.C. Carnes, G.K. Phillips, D. Padilla, P.N. Durfee, P.A. Brown, et livery: from molecular design to applications, Polym. Chem. 5 (2014)
al., The targeted delivery of multicomponent cargos to cancer cells by nano- 1519e1528.
porous particle-supported lipid bilayers, Nat. Mater. 10 (2011) 389e397; [24] G.K. Such, Y. Yan, A.P.R. Johnston, S.T. Gunawan, F. Caruso, Interfacing mate-
b) X. Guo, X. Wei, Y. Jing, S. Zhou, Size changeable nanocarriers with nuclear rials science and biology for drug carrier design, Adv. Mater. 27 (2015)
targeting for effectively overcoming multidrug resistance in cancer therapy, 2278e2297.
Adv. Mater. 27 (2015) 6450e6456. [25] L. Brülisauer, M.A. Gauthier, J. Leroux, Disulfide-containing parenteral delivery
[4] a) Y.J. Wang, S.M. Hussain, G.P. Krestin, Superparamagnetic iron oxide contrast systems and their redox-biological fate, J. Control. Release 195 (2014)
agents: physicochemical characteristics and applications in MR imaging, Eur. 147e154.
Radiol. 11 (2001) 2319e2331; [26] a) X. Guo, C. Shi, G. Yang, J. Wang, Z. Cai, S. Zhou, Dual-responsive polymer
b) P. Reimer, T. Balzer, Ferucarbotran (Resovist): a new clinically approved micelles for target-cell-specific anticancer drug delivery, Chem. Mater. 26
RES-specific contrast agent for contrast-enhanced MRI of the liver: properties, (2014) 4405e4418;
clinical development, and applications, Eur. Radiol. 13 (2003) 1266e1276. b) X. Wang, J. Zhang, R. Cheng, F. Meng, C. Deng, Z. Zhong, Facile synthesis of
[5] a) J. Xie, S. Lee, X. Chen, Nanoparticle-based theranostic agents, Adv. Drug reductively degradable biopolymers using cystamine diisocyanate as a
Deliv. Rev. 62 (2010) 1064e1079; coupling agent, Biomacromolecules 17 (3) (2016) 882e890.
b) X. Wang, D. Niu, Q. Wu, S. Bao, T. Su, X. Liu, et al., Iron oxide/manganese [27] M. Ding, J. Li, H. Tan, Q. Fu, Self-assembly of biodegradable polyurethanes for
oxide co-loaded hybrid nanogels as pH-responsive magnetic resonance controlled delivery applications, Soft Matter. 8 (2012) 5414e5428.
contrast agents, Biomaterials 53 (2015) 349e357. [28] a) Y. Wang, G. Wu, X. Li, Y. Wang, H. Gao, J. Ma, On-off switchable drug release
[6] H. Maeda, H. Nakamura, J. Fang, The EPR effect for macromolecular drug de- from multi-responsive degradable poly(ether urethane) nanoparticles, Bio-
livery to solid tumors: improvement of tumor uptake, lowering of systemic mater. Sci. 1 (2013) 614e624;
toxicity, and distinct tumor imaging in vivo, Adv. Drug Deliv. Rev. 65 (2013) b) S. Yu, J. Ding, C. He, Y. Cao, W. Xu, X. Chen, Disulfide cross-linked poly-
71e79. urethane micelles as a reduction-triggered drug delivery system for cancer
[7] a) U. Prabhakar, H. Maeda, R.K. Jain, E.M. Sevick-Muraca, W. Zamboni, therapy, Adv. Healthc. Mater. 3 (2014) 752e760;
O.C. Farokhzad, et al., Challenges and key considerations of the enhanced c) F. Huang, R. Cheng, F. Meng, C. Deng, Z. Zhong, Micelles based on acid
permeability and retention effect for nanomedicine drug delivery in oncology, degradable poly(acetal urethane): preparation, pH-sensitivity, and triggered
Cancer Res. 73 (2013) 2412; intracellular drug release, Biomacromolecules 16 (2015) 2228e2236;
b) H. Maeda, Toward a full understanding of the EPR effect in primary and e) M. He, A. Potuck, J.C. Kohn, K. Fung, C.A. Reinhart-King, C. Chu, Self-
metastatic tumors as well as issues related to its heterogeneity, Adv. Drug assembled cationic biodegradable nanoparticles from pH-responsive amino-
Deliv. Rev. 91 (2015) 3e06. acid-based poly(ester urea urethane)s and their application as a drug delivery
[8] Y. Huang, S. He, W. Cao, K. Cai, X. Liang, Biomedical nanomaterials for vehicle, Biomacromolecules 17 (2016) 523e537.
imaging-guided cancer therapy, Nanoscale 4 (2012) 6135e6149. [29] a) M. Ding, N. Song, X. He, J. Li, L. Zhou, H. Tan, et al., Toward the next-
J. Wei et al. / Biomaterials 145 (2017) 138e153 153

generation nanomedicines: design of multifunctional multiblock poly- Chem-Eur J. 17 (2011) 10064e10070;


urethanes for effective cancer treatment, ACS Nano 7 (2013) 1918e1928; b) J. Kerr, J.L. Schlosser, D.R. Griffin, D.Y. Wong, A.M. Kasko, Steric effects in
b) M. Ding, J. Li, X. He, N. Song, H. Tan, Y. Zhang, et al., Molecular engineered peptide and protein exchange with activated disulfides, Biomacromolecules
super-nanodevices: smart and safe delivery of potent drugs into tumors, Adv. 14 (2013) 2822e2829.
Mater. 24 (2012) 3639e3645. [43] D.J. Phillips, M.I. Gibson, Redox-sensitive materials for drug delivery: targeting
[30] a) M. Ding, X. Zeng, X. He, J. Li, H. Tan, Q. Fu, Cell internalizable and intra- the correct intracellular environment, tuning release rates, and appropriate
cellularly degradable cationic polyurethane micelles as a potential platform predictive systems, Antioxid. Redox Sign 21 (2013) 786e803.
for efficient imaging and drug delivery, Biomacromolecules 15 (2014) [44] H.S. Yoo, T.G. Park, Folate receptor targeted biodegradable polymeric doxo-
2896e2906; rubicin micelles, J. Control. Release 96 (2004) 273e283.
b) M. Ding, J. Wei, R. Wang, X. Shuai, J. Li, H. Tan, et al., Multifunctional [45] a) J. Lin, J. Chen, S. Huang, J. Ko, Y. Wang, T. Chen, et al., Folic acidePluronic
cystine-installed clickable micelles for efficient drug delivery and magnetic F127 magnetic nanoparticle clusters for combined targeting, diagnosis, and
resonance imaging, J. Control. Release 259 (2017) e96. therapy applications, Biomaterials 30 (2009) 5114e5124;
[31] N. Song, M. Ding, Z. Pan, J. Li, L. Zhou, H. Tan, et al., Construction of targeting- b) M. Guo, C. Que, C. Wang, X. Liu, H. Yan, K. Liu, Multifunctional super-
clickable and tumor-cleavable polyurethane nanomicelles for multifunctional paramagnetic nanocarriers with folate-mediated and pH-responsive targeting
intracellular drug delivery, Biomacromolecules 14 (2013) 4407e4419. properties for anticancer drug delivery, Biomaterials 32 (2011) 185e194;
[32] M. Ding, J. Li, X. Fu, J. Zhou, H. Tan, Q. Gu, et al., Synthesis, degradation, and c) L. Ao, B. Wang, P. Liu, L. Huang, C. Yue, D. Gao, et al., A folate-integrated
cytotoxicity of multiblock poly(ε-caprolactone urethane)s containing gemini magnetic polymer micelle for MRI and dual targeted drug delivery, Nanoscale
quaternary ammonium cationic groups, Biomacromolecules 10 (2009) 6 (2014) 10710e10716;
2857e2865. d) J. Eliezar, W. Scarano, N.R.B. Boase, K.J. Thurecht, M.H. Stenzel, In vivo
[33] X. Li, H. Li, G. Liu, Z. Deng, S. Wu, P. Li, et al., Magnetite-loaded fluorine- evaluation of folate decorated cross-linked micelles for the delivery of plat-
containing polymeric micelles for magnetic resonance imaging and drug de- inum anticancer drugs, Biomacromolecules 16 (2015) 515e523.
livery, Biomaterials 33 (2012) 3013e3024. [46] Y. Huang, Z. Tang, X. Zhang, H. Yu, H. Sun, X. Pang, et al., pH-triggered charge-
[34] M. Ding, Z. Qian, J. Wang, J. Li, H. Tan, Q. Gu, et al., Effect of PEG content on the reversal polypeptide nanoparticles for cisplatin delivery: preparation and
properties of biodegradable amphiphilic multiblock poly(ε-caprolactone in vitro evaluation, Biomacromolecules 14 (2013) 2023e2032.
urethane)s, Polym. Chem. 2 (2011) 885e891. [47] F. Meng, Z. Zhong, J. Feijen, Stimuli-responsive polymersomes for pro-
[35] I. Astafieva, X.F. Zhong, A. Eisenberg, Critical micellization phenomena in grammed drug delivery, Biomacromolecules 10 (2009) 197e209.
block polyelectrolyte solutions, Macromolecules 26 (1993) 7339e7352. [48] H. Sun, B. Guo, X. Li, R. Cheng, F. Meng, H. Liu, et al., Shell-sheddable micelles
[36] M. Ding, X. He, Z. Wang, J. Li, H. Tan, H. Deng, et al., Cellular uptake of poly- based on dextran-SS-poly(ε-caprolactone) diblock copolymer for efficient
urethane nanocarriers mediated by gemini quaternary ammonium, Bio- intracellular release of doxorubicin, Biomacromolecules 11 (2010) 848e854.
materials 32 (2011) 9515e9524. [49] a) S. Laurent, D. Forge, M. Port, A. Roch, C. Robic, L. Vander Elst, et al., Magnetic
[37] H. Ai, C. Flask, B. Weinberg, X.T. Shuai, M.D. Pagel, D. Farrell, et al., Magnetite- iron oxide nanoparticles: synthesis, stabilization, vectorization, physico-
loaded polymeric micelles as ultrasensitive magnetic-resonance probes, Adv. chemical characterizations, and biological applications, Chem. Rev. 108 (2008)
Mater. 17 (2005) 1949e1952. 2064e2110;
[38] H. Zhu, J. Tao, W. Wang, Y. Zhou, P. Li, Z. Li, et al., Magnetic, fluorescent, and b) H.B. Na, I.C. Song, T. Hyeon, Inorganic nanoparticles for MRI contrast agents,
thermo-responsive Fe3O4/rare earth incorporated poly(St-NIPAM) coreeshell Adv. Mater. 21 (2009) 2133e2148.
colloidal nanoparticles in multimodal optical/magnetic resonance imaging [50] H.Y. Yoon, G. Saravanakumar, R. Heo, S.H. Choi, I.C. Song, M.H. Han, et al.,
probes, Biomaterials 34 (2013) 2296e2306. Hydrotropic magnetic micelles for combined magnetic resonance imaging
[39] a) C. Allen, D. Maysinger, A. Eisenberg, Nano-engineering block copolymer and cancer therapy, J. Control. Release 160 (2012) 692e698.
aggregates for drug delivery, Colloids Surfaces B 16 (1999) 3e27; [51] X. Yang, S. Pilla, J.J. Grailer, D.A. Steeber, S. Gong, Y. Chen, et al., Tumor-tar-
b) M. Ding, L. Zhou, X. Fu, H. Tan, J. Li, Q. Fu, Biodegradable gemini multiblock geting, superparamagnetic polymeric vesicles as highly efficient MRI contrast
poly(ε-caprolactone urethane)s toward controllable micellization, Soft Matter. probes, J. Mater. Chem. 19 (2009) 5812e5817.
6 (2010) 2087e2092. [52] H. Su, Y. Liu, D. Wang, C. Wu, C. Xia, Q. Gong, et al., Amphiphilic starlike
[40] J. Zhang, L. Wang, H. Wang, K. Tu, Micellization phenomena of amphiphilic dextran wrapped superparamagnetic iron oxide nanoparticle clsuters as
block copolymers based on methoxy poly(ethylene glycol) and either crys- effective magnetic resonance imaging probes, Biomaterials 34 (2013)
talline or amorphous poly(caprolactone-b-lactide), Biomacromolecules 7 1193e1203.
(2006) 2492e2500. [53] C.E. Smith, D. Ernenwein, A. Shkumatov, N.E. Clay, J. Lee, M. Melhem, et al.,
[41] a) H. Sun, B. Guo, R. Cheng, F. Meng, H. Liu, Z. Zhong, Biodegradable micelles Hydrophilic packaging of iron oxide nanoclusters for highly sensitive imaging,
with sheddable poly(ethylene glycol) shells for triggered intracellular release Biomaterials 69 (2015) 184e190.
of doxorubicin, Biomaterials 30 (2009) 6358e6366; [54] a) J. Zhu, J. Wang, X. Wang, J. Zhu, Y. Yang, J. Tian, et al., Facile synthesis of
b) W. Lu, X. Wang, R. Cheng, C. Deng, F. Meng, Z. Zhong, Biocompatible and magnetic core-shell nanocomposites for MRI and CT bimodal imaging,
bioreducible micelles fabricated from novel a-amino acid-based poly(disulfide J. Mater. Chem. B 3 (2015) 6905e6910;
urethane)s: design, synthesis and triggered doxorubicin release, Polym. Chem. b) Y. Sarigiannis, A. Kolokithas-Ntoukas, N. Beziere, R. Zboril, E. Papadimitriou,
6 (2015) 6001e6010. K. Avgoustakis, et al., Synthesis and evaluation of condensed magnetic
[42] a) C. Wu, C. Belenda, J. Leroux, M.A. Gauthier, Interplay of chemical micro- nanocrystal clusters with in vivo multispectral optoacoustic tomography for
environment and redox environment on thioledisulfide exchange kinetics, tumour targeting, Biomaterials 91 (2016) 128e139.

You might also like