You are on page 1of 8

www.advmat.

de
www.MaterialsViews.com

Self-Assembled Autophagy-Inducing Polymeric

COMMUNICATION
Nanoparticles for Breast Cancer Interference In-Vivo
Yi Wang, Yao-Xin Lin, Zeng-Ying Qiao, Hong-Wei An, Sheng-Lin Qiao, Lei Wang,
R P Yeshan J Rajapaksha, and Hao Wang*

Autophagy is a lysosome-based evolutionarily conserved pro- one-pot synthesis method and effectively delivered drugs.[19,20]
cess that plays an important role in cellular degradation for the Although numerous nanomaterials influence the autophagy
clearance of damaged or superfluous proteins and organelles.[1] process,[21] few examples, to the best of our knowledge, have
Abnormalities in autophagy may directly correlate to various been reported to effectively control autophagic flux and ulti-
pathologic diseases, including neurodegenerative disease,[2] mately achieve therapeutic potential.
cardiac disease,[3] and cancer.[4] Recently, researchers have Herein, we describe the development of autophagy-inducing
attempted to regulate autophagy and prevent further tumor pro- peptides engineered into polymeric nanoparticles for highly
gress.[5,6] Compared to present cancer therapy strategies, such efficient induction of autophagy and interference of breast
as chemo-,[7] immuno-,[8] gene,[9] radiation,[10] photothermal,[11] cancer in vitro and in vivo. Bec1, as an autophagy-inducing pep-
and photodynamic[12] therapies, the autophagy-based concep- tide, covalently grafts onto pH-sensitive polymers (Ps), which
tual pilot studies for cancer therapy are of particular – and self-assemble into micelle-like nanoparticles (P-Bec1) with
growing – interest in autophagy-deficient cancers, for example, poly(ethylene glycol) (PEG) as hydrophilic shells (Scheme 1).
breast, ovarian, and prostate cancers.[6] The P-Bec1 nanoparticles improved the stability and enhanced
Beclin-1 (Bec1) is the peptide encoded by the human gene the cellular uptake of Bec1 in vitro. P-Bec1 can dissociate in the
BECN1, a mammalian homolog of the yeast autophagy- weakly acidic lysosomal environment (pH 4–5), which resulted
related gene (Atg) 6, and participates in the regulation of in the alkalization and impairment of lysosomes.[22] The disso-
autophagy through binding to phosphatidylinositol-4,5-bispho- ciated P-Bec1 can escape from lysosomes and effectively induce
sphate 3-kinases (PI3Ks, signal transducer enzymes), which autophagy, leading to autophagic cell death. Moreover, P-Bec1
are required for initiation of autophagosome formation in is also able to effectively inhibit the growth of tumors and slow
autophagy.[13] BECN1 has been identified as a haploinsufficient down tumor development in vivo.
tumor suppressor, and is commonly deleted in human breast, The amphiphilic poly(β-amino ester) copolymer composed
ovarian, and prostate tumors.[14] Furthermore, upregulation of of the hydrophobic monomer 1,6-hexanediol diacrylate (HDDA,
Bec1 enhances Ras-induced cell death in MCF-7 breast cancer 1.2 mM), the pH-sensitive monomer 3-(dibutylamino)-1-pro-
cells.[15] Therefore, it has been suggested that overexpression of pylamine (DBPA, 0.7 mM), and hydrophilic amino-terminated
Bec1 could inhibit tumor development.[6,16] However, the thera- PEG (PEG-NH2, MW = 2 kDa, 0.3 mM) was synthesized by
peutic Bec1 peptide has two major drawbacks that hamper their Michael addition.[19,20] The structure of the polymer was charac-
in vivo application, namely poor chemical stability in vivo and terized by 1H NMR (Figure S4, in the Supporting Information,
non-specific bio-distribution in tissues. Chemical modification SI). The degree of polymerization (DP) and molecular weight
and/or self-assembly are two useful approaches for overcoming (MW) of copolymers were deduced to be 27 and ca. 16 kDa,
these limitations.[17] pH-sensitive polymers have been devel- respectively.
oped widely to realize the controlled disassembly and release of Furthermore, Bec1 was engineered to have a thiol group
payloads in biological conditions.[18] Our previous studies dem- at the N terminal to conjugate with the poly(β-amino ester)
onstrated that a pH-sensitive poly(β-amino ester) copolymer copolymer (Figures S2–S4, SI). The 1H NMR spectrum of
self-assembled into a micelle-like nanoparticle by means of a P-Bec1 showed the disappearance of the acrylate double bonds
of the polymer P at 5.8–6.3 ppm and the appearance of Bec1
Y. Wang, Y.-X. Lin, Dr. Z.-Y. Qiao, H.-W. An, S.-L. Qiao, peaks at 6.7–8.5 ppm (Figure 1a), indicating that the acrylate
Prof. L. Wang, R. P. Y. J. Rajapaksha, Prof. H. Wang groups on both ends of the polymer had completely reacted
CAS Key Laboratory for Biological Effects of with Bec1. Circular dichroism (CD) spectra of Bec1 and P-Bec1
Nanomaterials and Nanosafety were analyzed using standard CONTINLL algorithms,[23] which
National Center for Nanoscience
and Technology (NCNST) revealed the secondary structure of Bec1 to be 33.8% α-helix,
No. 11 Beiyitiao, Zhongguancun 100190 and P-Bec1, 18.8% α-helix, proving that 56% of the secondary
Beijing, (P. R. China) structure of Bec1 was not destroyed during the process of copol-
E-mail: wanghao@nanoctr.cn ymer synthesis (Figure 1c). The amphiphilic P-Bec1 polymers
Y. Wang, Y.-X. Lin, H.-W. An, S.-L. Qiao simultaneously self-assemble into micelle-like nanoparticles
University of Chinese Academy of Science (UCAS)
No.19A Yuquan Road, 100049 Beijing, (P. R. China)
during dialysis from dimethyl sulfoxide (DMSO) into water
(Scheme 1). The morphologies and hydrodynamic sizes of P
DOI: 10.1002/adma.201405926 and P-Bec1 were observed by transmission electron microscopy

Adv. Mater. 2015, © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 1
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

Scheme 1. Schematic illustration of P-Bec1 nanoparticles for a highly efficient autophagy-inducing process.

(TEM) and dynamic lighting scattering (DLS), respectively. of autophagy we then measured the expression of the protein
Average hydrodynamic sizes of 73.7 ± 26.3 nm and 32.4 ± light chain 3-II (LC3-II), as the conversion of LC3-I to LC3-II is
11.3 nm were obtained for P and P-Bec1, respectively, which widely used to monitor autophagosome formation.[25,26] Using
were in accordance with the diameters obtained from TEM the MCF-7 cell line, which stably expresses green fluorescent
(Figure 1b). Moreover, cryo-TEM and scanning electron micros- protein (GFP)-tagged LC3 (GFP-LC3),[26,27] we found that both
copy (SEM) measurements further validated the self-assembled P and P-Bec1 indeed induced accumulation of GFP-LC3–posi-
spherical morphology of P-Bec1 (Figures S6 and S7, SI). The tive puncta. The quantitative analysis of induce GFP-LC3 dots
zeta potential of the P-Bec1 nanoparticles was about 24.1 ± 4.0 of MCF-7 cells showed that P-Bec1 treatment gave better elimi-
mV, implying favorable interaction with negatively charged cells nation than Bec1 and P treatment (p = 0.008) (Figure 2a), which
(Figure S8, SI). In addition, the diameter of P-Bec1 changed indicated that P-Bec1 induced autophagosome accumulation
from 32.4 ± 11.3 nm at pH 7.4 to 275.2 ± 20.2 nm at pH 5.0 effectively.
(Figure 1b), implying that P-Bec1 disassociates and reorganizes Acridine Orange (AO) is a pH-sensitive dye that can stain
into larger nanoparticles under weakly acidic conditions. acidic vesicular organelles and is used for autophagy detection.
In order to investigate the biological effects of P-Bec1 nano- It is membrane permeable so that DNA and cytoplasm can be
particles, we first evaluated the cytotoxicity of P-Bec1 nano- stained green and acid vesicular organelles red; the ratio of red
particles on MCF-7 cells. Bec1 itself was barely taken up by cells to green can be used to evaluate the effect of autophagy.[26,28]
and therefore exhibited little cytotoxicity even at a high concen- The confocal laser scanning microscopy (CLSM) images indi-
tration (ca. 50 µM). A high dose of P (ca. 284 µg mL−1) showed cated that there were more red spots in P-Bec1–treated cells,
a little cytotoxicity, which could be explained by the pH-sensi- which indicated high accumulation of autophagosomes. The
tive nanoparticles inducing autophagic cell death, as we showed CLSM images also revealed the occurrence of cellular mor-
previously[24] (Figure S9, SI). However, the P-Bec1 nanoparticles phologic changes from spreading to rounded states after
(half maximal inhibitory concentration IC50 = 186.7 µg mL−1) P-Bec1 treatment for 24 h (Figure 2c). In addition, flow cytom-
exhibited much higher cytotoxicity than the polymer P (IC50 = etry analysis showed that the ratio of red to green fluorescent
269.7 µg mL−1), which may be reasonably explained with the intensity increased almost six-fold compared to the control
conjecture that P effectively carried autophagy-inducing peptide group when treated with P-Bec1 (Figure S10, SI). These results
Bec1 into the cells. To prove that cytotoxicity is from induction demonstrated that P-Bec1 nanoparticles strongly induced

2 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015,
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
Figure 1. Characterizations of P-Bec1 nanoparticles. a) 1H NMR spectra of Bec1, P, and P-Bec1 in d6-DMSO. Peaks corresponding to the double
bonds of the acrylate end group are labeled *; the partial Bec1 peptide characteristic peaks are enclosed in dashed boxes. b) TEM image of P-Bec1 in
pH 7.4 aqueous solution (scale bar, 100 nm), which shows spherical nanoparticles. Inset: Hydrodynamic size of P-Bec1 in pH 7.4 and pH 5.0 aqueous
solutions by DLS. c) Circular dichroism (CD) spectra of Bec1, P, and P-Bec1 at the same mass concentration (1 mg mL−1).

autophagy and potentially led to autophagic cell death. Western are taken up through the endocytosis process, as we proposed
blot also confirmed that P-Bec1 treatment increased conver- (Figure S12, SI). To determine whether P-Bec1 could inter-
sion of LC3-I to LC3-II significantly, whereas Bec1 treatment fere with lysosome function and induce lysosome-related
induced practically no accumulation of LC3-II. Moreover, we autophagic cell death, we first labeled P-Bec1 nanoparticles with
found that P-Bec1 treatment did not cause p62 (a substrate that the cyanine dye Cy5 (P-Bec1-Cy5) and measured the subcel-
can be degraded by autophagy) degradation but instead caused lular localization by CLSM. The MCF-7 cells were treated with
accumulation of p62, which indicated possible impairment of P-Bec1-Cy5 for 2 h and then stained with LysoTracker Green
autophagic degradation capacity (Figure 2d and Figure S11, SI). DND-26. CLSM images exhibited that the P-Bec1-Cy5 signal
The lysosome is closely related to the autophagic cellular was co-localized with endosomes/lysosomes (Figure 3a). This
process and is believed to be the key organelle involved in result proved that P-Bec1 nanoparticles entered the cell effec-
autophagic cell death.[29] Mounting evidence strongly implies tively by endocytosis. Second, the MCF-7 cells, which stably
that the nanoparticles influence lysosomes remarkably, owing express GFP-LC3, were treated with P-Bec1 for 24 h and then
to their endocytosis cell uptake pathway and high accumula- incubated with LysoTracker Red DND-99. CLSM images dis-
tion in the lysosome compartment.[30] P-Bec1 nanoparticles playing both wavelengths of fluorescence were also obtained,

Adv. Mater. 2015, © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 3
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

Figure 2. P-Bec1 induces autophagic cell death. a,b) Representative images (a) and the corresponding quantitative analysis (b) of GFP-LC3–positive
puncta of non-treated and treated MCF-7/GFP-LC3 cells (15 µM, 24 h). c) AO staining of MCF-7 cells after the same treatment as in (a). d) Western
blot (and its analysis) of LC3-II/LC3-I and the protein p62 after the same treatment as in (a). All treatment concentrations are referred to Bec1 and an
equal amount of copolymer in P-Bec1; ** p < 0.01, *** p < 0.001; one-way analysis of variance (ANOVA) for indicated comparison.

displaying in large areas of yellow signals where the images effect. In addition, we used ex vivo experiments to further
overlapped (Figure 3c), which means that P-Bec1–induced evaluate the bio-distribution of P-Bec1. The tumor exhibited a
GFP-LC3 puncta almost overlapped with the lysosome signal strong signal, and the ratio of tumor signal to liver and kidney
(LysoTracker Red DND-99). These results confirmed that most signals was 1:1.5 and 1:1.9, respectively (Figure 4a,b). Some
of the autophagosomes induced by P-Bec1 could be trapped in other harvested organs exhibited fluorescence signals likewise,
the lysosomes but not degraded. Therefore, we can conclude especially the liver and kidneys, which are the two main meta-
that P-Bec1 might impair lysosomal degradation capacity. bolic organs of mammals. However, fluorescence signals from
As we reported, pH-sensitive nanoparticles can damage lys- the liver and kidneys became weaker at 24 h post-injection,
osomes by increasing lysosomal pH.[24] To test whether P-Bec1 unlike signals from tumor tissue: at the tumor site stronger
can affect lysosomal pH, the MCF-7 cells were labeled with fluorescence was observed at 24 h than at 4 h post-injection
LysoSensor Green DND-189 dye to monitor lysosome acidity. (Figure S13, SI), suggesting that metabolism of P-Bec1 is rapid
Flow cytometry analysis showed that the LysoTracker Green in the liver and kidneys but that it could accumulate in tumor
DND-189 fluorescent intensity of cells that had been treated tissue. The ex vivo results at 24 h suggest to us that P-Bec1 may
with P-Bec1 was decreased, which determined the alkaliza- have little toxicity to normal tissue but will effectively inhibit a
tion of the lysosomes and corresponding lysosome dysfunction tumor.
(Figure 3b). These results revealed that P-Bec1 induced lyso- In order to further confirm the bio-distribution of P-Bec1
somal dysfunction through lysosome alkalization. nanoparticles, besides the fluorescence method, we further
That P-Bec1 induces autophagic cell death significantly in encapsulated gold nanoparticles into the nanoparticles and uti-
vitro led us to consider that P-Bec1 may serve as a novel can- lized inductively coupled plasma mass spectrometry (ICP-MS)
didate for breast cancer therapy. We used P-Bec1-Cy5, injected to monitor the bio-distribution of P-Bec1 nanoparticles (Figure
intravenously, to monitor the bio-distribution of P-Bec1 in S14, SI). According to the data of ICP-MS, we calculated that
MCF-7 tumor-bearing nude mice. Mice injected with phos- the P-Bec1 accumulated in the tumor had increased by 7.8%
phate-buffered saline (PBS) through the tail vein were used as compared with initially injected amount of P-Bec1. To further
controls. Strong fluorescence illuminations around the tumor confirm the systemic toxicity of P-Bec1 nanoparticles, hematox-
were observed using a multispectral imaging system 4 h after ylin-eosin (H&E) staining was applied to liver, kidney, spleen,
injection of P-Bec1-Cy5, indicating the specific tumor targeting heart, and lung tissues. We observed that there was no lesion in
of P-Bec1 by the enhanced permeability and retention (EPR) these major organs, which was probably attributable to the fact

4 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015,
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com

COMMUNICATION

Figure 3. P-Bec1 destroys lysosomal degradation capacity through alkalization of lysosomes. a) Co-localization of P-Bec1 micelles (red) and lysosomes
(green); cells treated with P-Bec1-Cy5 for 2 h and stained with LysoTracker Green DND-26, BF (bright field). b) Flow cytometry analysis of cells with
control, Bec1, P, or P-Bec1 treatment and staining with LysoSensor Green-189 (15 µM, 2 4h). c) Co-localization of GFP-LC3–positive puncta and lys-
osomes. MCF-7/GFP-LC3 cells treated with P-Bec1 for 24 h and stained with LysoTracker Red DND-99.

that the P-Bec1 was rapidly cleared from these major organs mice model. We compared the tumor size, body weight, and
(Figure S15, SI). The high accumulation of P-Bec1 nanoparticles pathological morphology of tumor tissues treated with Bec1,
at the tumor site indicated the potential anticancer effect in vivo P, or P-Bec1. The process of tumor growth was observed
as a result of its efficient autophagy-inducing effect. and recorded for 13 days. As shown in Figure 4c, the tumor
To evaluate the effect of P-Bec1 on tumor suppression in size of MCF-7 tumor-bearing nude mice treated with P-Bec1
vivo, we used the MCF-7 cancer cells xenografted tumor nude nanoparticles increased significantly more slowly than that of

Adv. Mater. 2015, © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 5
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

the control group. The tumor volume ratio


(V/Vinitial) of the control group increased
more than two times more than that of the
P-Bec1–treated group; the tumor inhibitory
rate was calculated to be 51.6%. It is possible
that nanometer-scale P-Bec1 nanoparticles
can slow down the tumor growth by the
effect ascribed to Bec1, which was protected
and accumulated in the tumor site by the
EPR effect (p = 0.004). The Bec1-treated
group showed almost no difference to the
control group, so it can be deduced that the
free peptide Bec1 was degraded and removed
quickly in vivo. However, the nanometer-
scale polymer P can accumulate in the
tumor site by the EPR effect and exhibited
a minute effect on tumor growth, which can
be explained by the pH-sensitive polymers
with autophagy-inducing function. Further-
more, P-Bec1 nanoparticles slowed down the
tumor growth significantly compared to P
nanoparticles and Bec1, demonstrating that
P-Bec1 can kill tumor cells synergistically
and effectively.
None of the mice groups had significant
variations in body weight throughout the
treatment processes (ca. 16 g), so it can be
inferred that there was no extreme toxicity
with these treatments in vivo (Figure 4d). As
well as the biological effects tumor volume
and body weight, we further investigated the
pathological morphology of all the tumor
tissues. H&E staining showed that P-Bec1
reduced breast tumor growth and invasion in
a breast cancer nude mouse model in com-
parison with a control group (Figures 4e).
However, a Bec1-treated group and a P-treated
group showed that tumor cells were poorly
differentiated (Figure S16, SI). These results
indicate that P-Bec1 reduced breast tumor
growth and invasion.
To determine whether P-Bec1 could induce
autophagy in vivo, we used an immunohis-
tochemical method to detect the expression
of LC3-II in tumor tissues. The tumor tis-
sues were incubated with LC3-II antibody,
and the expression of LC3-II was detected by
a fluorescein isothiocyanate (FITC)-labeled
second antibody. Compared to the control
Figure 4. P-Bec1 effectively inhibits the growth of a tumor by inducing autophagy in vivo. a) Ex
vivo near-infrared (NIR) optical images of tumor tissue and major organs dissected from PBS-
group, there was stronger immunofluores-
and P-Bec1–injected mice with MCF-7 cells xenografted for 4 h. b) The corresponding average cence of LC3-II in the P-Bec1 treatment group
signal intensities. Each column represents the mean ± SD (standard deviation), number of (Figure 4f and Figure S17, SI). These results
samples n = 3. c,d) Tumor volume (c) and body weight (d) as a function of time for PBS, Bec1, suggested that P-Bec1 nanoparticles can effec-
P, P-Bec1 treatment groups after MCF-7 tumor xenograft for 7 days. The concentration of Bec1 tively induce autophagic cell death in vivo.
was 20 mg kg−1 in both the Bec1 and P-Bec1 groups. Each point represents a mean ± SD, n In conclusion, nanomaterials with diversi-
= 5, *p < 0.05, **p < 0.01,***p < 0.001; one-way ANOVA for indicated comparison. e) Patho-
fied physiochemical parameters have shown
logical features of tumor tissues in representative mice treated with PBS or P-Bec1. Tumor
issues were H&E stained. f) Immunohistochemical representative images of tumor tissues the possibility of autophagy regulation.[21]
for PBS or P-Bec1-treated mice. Stained with LC3-II; the entire nucleus was stained with DAPI The mechanism is not clear so far; it remains
(4’,6-diamidino-2-phenylindole). challenge for cancer therapy owing to the

6 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015,
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com

COMMUNICATION
limited work on autophagic networks. We have demonstrated [10] a) A. C. Begg, F. A. Stewart, C. Vens, Nat. Rev. Cancer 2011, 11, 239;
a convenient approach to preparing self-assembled micelle-like b) R. Baskar, K. A. Lee, R. Yeo, K.-W. Yeoh, Int. J. Med. Sci. 2012, 9,
nanoparticles composed of pH-sensitive poly(β-amino ester)s 193.
and an autophagy-inducing peptide (Bec1). The P-Bec1 nano- [11] a) S. Lal, S. E. Clare, N. J. Halas, Acc. Chem. Res. 2008, 41, 1842;
b) J. G. Morton, E. S. Day, N. J. Halas, J. L. West, in Cancer Nano-
particles displayed enhanced autophagy-inducing cytotoxicity to
technology: Methods and Protocols, Methods in Molecular Biology,
MCF-7 cells that are Bec1 deficient, because P-Bec1 can con- Vol. 624 (Eds: S. R. Grobmyer, B. M. Moudgil), Humana Press, New
tribute to efficient internalization of the Bec1 peptide into cells York 2010, pp. 101–117.
by the endocytosis pathway. Moreover, the P-Bec1 nanoparticles [12] a) D. Dolmans, D. Fukumura, R. K. Jain, Nat. Rev. Cancer 2003, 3,
effectively induced autophagy and inhibited tumor growth. 380; b) P. Agostinis, K. Berg, K. A. Cengel, T. H. Foster, A. W. Girotti,
By modulating the Bec1 peptide autophagy-inducing effect S. O. Gollnick, S. M. Hahn, M. R. Hamblin, A. Juzeniene, D. Kessel,
assisted by pH-sensitive polymers, we identified the autophagy M. Korbelik, J. Moan, P. Mroz, D Nowis, J. Piette, B. C. Wilson,
anticancer efficiency of P-Bec1. We believe that the P-Bec1 pep- J. Golab, CA — Cancer J. Clin. 2011, 61, 250.
tide described here provides an important addition to existing [13] a) V. M. Aita, X. H. Liang, V. Murty, D. L. Pincus, W. P. Yu,
efforts in identifying new cancer therapies, and has a prom- E. Cayanis, S. Kalachikov, T. C. Gilliam, B. Levine, Genomics 1999,
59, 59; b) S. Pattingre, A. Tassa, X. P. Qu, R. Garuti, X. H. Liang,
ising effect especially in autophagy-deficiency tumors.
N. Mizushima, M. Packer, M. D. Schneider, B. Levine, Cell 2005,
122, 927.
[14] a) X. H. Liang, S. Jackson, M. Seaman, K. Brown, B. Kempkes,
Supporting Information H. Hibshoosh, B. Levine, Nature 1999, 402, 672; b) X. P. Qu,
J. Yu, G. Bhagat, N. Furuya, H. Hibshoosh, A. Troxel, J. Rosen,
Supporting Information is available from the Wiley Online Library or E. L. Eskelinen, N. Mizushima, Y. Ohsumi, G. Cattoretti, B. Levine,
from the authors. J. Clin. Invest. 2003, 112, 1809; c) Z. Y. Yue, S. K. Jin, C. W. Yang,
A. J. Levine, N. Heintz, Proc. Natl. Acad. Sci. USA 2003, 100, 15077.
[15] M. Elgendy, C. Sheridan, G. Brumatti, S. J. Martin, Mol. Cell 2011,
Acknowledgements 42, 23.
[16] a) Y. Liu, S. Shoji-Kawata, R. M. Sumpter Jr., Y. Wei, V. Ginet,
Y.W. and Y.-X.L. contributed equally to this work. This work was L. Zhang, B. Posner, K. A. Tran, D. R. Green, R. J. Xavier, S. Y. Shaw,
supported by the National Basic Research Program of China (973 P. G. H. Clarke, J. Puyal, B. Levine, Proc. Natl. Acad. Sci. USA
Program, 2013CB932701), the National Natural Science Foundation 2013, 110, 20364; b) S. Shoji-Kawata, R. Sumpter Jr., M. Leveno,
of China (21374026, 21304023, 51102014, and 51303036), the Beijing G. R. Campbell, Z. Zou, L. Kinch, A. D. Wilkins, Q. Sun, K. Pallauf,
Natural Science Foundation (2132053), and the 100-Talent Program of D. MacDuff, C. Huerta, H. W. Virgin, J. B. Helms, R. Eerland,
the Chinese Academy of Science (Y2462911ZX). S. A. Tooze, R. Xavier, D. J. Lenschow, A. Yamamoto, D. King,
O. Lichtarge, N. V. Grishin, S. A. Spector, D. V. Kaloyanova,
Received: December 30, 2014
B. Levine, Nature 2013, 494, 201.
Revised: February 5, 2015
[17] A. Accardo, D. Tesauro, G. Morelli, Polym. J. 2013, 45, 481.
Published online:
[18] a) A. Akinc, D. G. Anderson, D. M. Lynn, R. Langer, Bioconjugate
Chem. 2003, 14, 979; b) D. G. Anderson, W. D. Peng, A. Akinc,
[1] Z. Yang, D. J. Klionsky, Nat. Cell Biol. 2010, 12, 814. N. Hossain, A. Kohn, R. Padera, R. Langer, J. A. Sawicki, Proc.
[2] R. A. Nixon, Nat. Med. 2013, 19, 983. Natl. Acad. Sci. USA 2004, 101, 16028; c) S. Aryal, C.-M. J. Hu,
[3] a) A. Terman, U. T. Brunk, Cardiovasc. Res. 2005, 68, 355; L. Zhang, ACS Nano 2009, 4, 251; d) Z.-Y. Qiao, R. Zhang, F. S. Du,
b) B. Levine, G. Kroemer, Cell 2008, 132, 27. D. H. Liang, Z. C. Li, J. Controlled Release 2011, 152, 57; e) Z.-Y. Qiao,
[4] a) B. Levine, Nature 2007, 446, 745; b) R. Mathew, F. S. Du, R. Zhang, D. H. Liang, Z. C. Li, Macromolecules 2010, 43,
V. Karantza-Wadsworth, E. White, Nat. Rev. Cancer 2007, 7, 961; 6485; f) Z.-Y. Qiao, J. Cheng, R. Ji, F.-S. Du, D.-H. Liang, S.-P. Ji,
c) P. D. Jiang, N. Mizushima, Cell Res. 2014, 24, 69. Z.-C. Li, RSC Adv. 2013, 3, 24345; g) S. Y. Park, H. J. Baik, Y. T. Oh,
[5] a) Z. N. J. Yang, C. E. Chee, S. B. Huang, F. A. Sinicrope, Cancer K. T. Oh, Y. S. Youn, E. S. Lee, Angew. Chem. Int. Ed. 2011, 50,
Biol. Ther. 2011, 11, 169; b) D. C. Rubinsztein, J. E. Gestwicki, 1644; h) E. M. Bachelder, T. T. Beaudette, K. E. Broaders, J. Dashe,
L. O. Murphy, D. J. Klionsky, Nat. Rev. Drug Discovery 2007, 6, 304; J. M. J. Frechet, J. Am. Chem. Soc. 2008, 130, 10494; i) W. Chen,
c) N. Mizushima, B. Levine, A. M. Cuervo, D. J. Klionsky, Nature F. H. Meng, R. Cheng, Z. Y. Zhong, J. Controlled Release 2010, 142,
2008, 451, 1069; d) S. H. van Rijt, P. J. Sadler, Drug Discovery Today 40; j) Y. Zhu, F. Wang, C. Zhang, J. Du, ACS Nano 2014, 8, 6644;
2009, 14, 1089. k) D. Ling, H. Xia, W. Park, M. J. Hackett, C. Song, K. Na, K. M. Hui,
[6] a) D. C. Rubinsztein, P. Codogno, B. Levine, Nat. Rev. Drug Dis- T. Hyeon, ACS Nano 2014, 8, 8027; l) L. Xing, H. Zheng, Y. Cao,
covery 2012, 11, 709; b) K. Jain, K. S. Paranandi, S. Sridharan, S. Che, Adv. Mater. 2012, 24, 6433.
A. Basu, Am. J. Cancer Res. 2013, 3, 251. [19] Z.-Y. Qiao, S.-L. Qiao, G. Fan, Y.-S. Fan, Y. Chen, H. Wang, Polym.
[7] a) N. A. Berger, J. Clin. Invest. 1986, 78, 1131; b) G. Bonadonna, Cell Chem. 2014, 5, 844.
Biophys. 1986, 9, 243. [20] Z. Duan, Y.-J. Gao, Z.-Y. Qiao, G. Fan, Y. Liu, D. Zhang, H. Wang, J.
[8] a) L. K. Aguilar, B. W. Guzik, E. Aguilar-Cordova, J. Cell. Biochem. Mater. Chem. B 2014, 2, 6271.
2011, 112, 1969; b) A. M. Scott, J. D. Wolchok, L. J. Old, Nat. Rev. [21] a) O. Seleverstov, O. Zabirnyk, M. Zscharnack, L. Bulavina,
Cancer 2012, 12, 278. M. Nowicki, J.-M. Heinrich, M. Yezhelyev, F. Emmrich, R. O’Regan,
[9] a) A. El-Aneed, Eur. J. Pharmacol. 2004, 498, 1; b) R. Ortiz, A. Bader, Nano Lett. 2006, 6, 2826; b) S. T. Stern, B. S. Zolnik,
C. Melguizo, J. Prados, P. J. Alvarez, O. Caba, F. Rodriguez-Serrano, C. B. McLeland, J. Clogston, J. Zheng, S. E. McNeil, Toxicol. Sci.
F. Hita, A. Aranega, Recent Pat. Anti-Cancer Drug Discovery 2012, 7, 2008, 106, 140; c) J. J. Li, D. Hartono, C.-N. Ong, B.-H. Bay,
297; c) S. K. Das, M. E. Menezes, S. Bhatia, X.-Y. Wang, L. Emdad, L.-Y. L. Yung, Biomaterials 2010, 31, 5996; d) X. W. Ma, Y. Y. Wu,
D. Sarkar, P. B. Fisher, J. Cell. Physiol. 2015, 230, 259. S. B. Jin, Y. Tian, X. N. Zhang, Y. L. Zhao, L. Yu, X. J. Liang, ACS

Adv. Mater. 2015, © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim wileyonlinelibrary.com 7
DOI: 10.1002/adma.201405926
www.advmat.de
www.MaterialsViews.com
COMMUNICATION

Nano 2011, 5, 8629; e) L. Yu, Y. Lu, N. Man, S.-H. Yu, L.-P. Wen, [26] D. J. Klionsky et al., Autophagy 2012, 8, 445.
Small 2009, 5, 2784; f) Y. Zhang, F. Zheng, T. Yang, W. Zhou, Y. Liu, [27] I. H. Lee, L. Cao, R. Mostoslavsky, D. B. Lombard, J. Liu,
N. Man, L. Zhang, N. Jin, Q. Dou, Y. Zhang, Z. Li, L.-P. Wen, Nat. N. E. Bruns, M. Tsokos, F. W. Alt, T. Finkel, Proc. Natl. Acad. Sci.
Mater. 2012, 11, 817; g) P. Wei, L. Zhang, Y. Lu, N. Man, L. Wen, USA 2008, 105, 3374.
Nanotechnology 2010, 21, 495101; h) Q. Zhang, W. Yang, N. Man, [28] a) E. Tasdemir, L. Galluzzi, M. C. Maiuri, A. Criollo, I. Vitale,
F. Zheng, Y. Shen, K. Sun, Y. Li, L.-P. Wen, Autophagy 2009, 5, 1107; E. Hangen, N. Modjtahedi, G. Kroemer, Methods Mol. Biol.
i) C. Li, H. Liu, Y. Sun, H. Wang, F. Guo, S. Rao, J. Deng, Y. Zhang, 2008, 445, 29; b) S. Paglin, T. Hollister, T. Delohery, N. Hackett,
Y. Miao, C. Guo, J. Meng, X. Chen, L. Li, D. Li, H. Xu, H. Wang, M. McMahill, E. Sphicas, D. Domingo, J. Yahalom, Cancer Res.
B. Li, C. Jiang, J. Mol. Cell Biol. 2009, 1, 37. 2001, 61, 439.
[22] S. M. Henry, M. E. H. El-Sayed, C. M. Pirie, A. S. Hoffman, [29] a) G. Kroemer, M. Jaattela, Nat. Rev. Cancer 2005, 5, 886;
P. S. Stayton, Biomacromolecules 2006, 7, 2407. b) J. P. Luzio, P. R. Pryor, N. A. Bright, Nat. Rev. Mol. Cell Biol.
[23] a) S. W. Provencher, J. Gloeckner, Biochemistry 1981, 20, 33; 2007, 8, 622; c) C. Poues, P. Codogno, Nat. Cell Biol. 2011, 13, 342;
b) I. H. M. van Stokkum, H. J. W. Spoelder, M. Bloemendal, d) C. Settembre, A. Fraldi, D. L. Medina, A. Ballabio, Nat. Rev. Mol.
R. van Grondelle, F. C. A. Groen, Anal. Biochem. 1990, 191, Cell Biol. 2013, 14, 283; e) Z. Xie, D. J. Klionsky, Nat. Cell Biol. 2007,
110. 9, 1102.
[24] Y.-X. Lin, Y. Wang, Z.-Y. Qiao, S.-L. Qiao, H.-W. An, R.-X. Zhang, [30] a) H. Hillaireau, P. Couvreur, Cell. Mol. Life Sci. 2009, 66, 2873;
R. P. Rajapaksha, L. Wang, H. Wang, unpublished. b) G. Sahay, D. Y. Alakhova, A. V. Kabanov, J. Controlled Release
[25] I. Tanida, N. Minematsu-Ikeguchi, T. Ueno, E. Kominami, 2010, 145, 182; c) T.-G. Iversen, T. Skotland, K. Sandvig, Nano Today
Autophagy 2005, 1, 84. 2011, 6, 176.

8 wileyonlinelibrary.com © 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim Adv. Mater. 2015,
DOI: 10.1002/adma.201405926

You might also like