Download as pdf or txt
Download as pdf or txt
You are on page 1of 26

Subscriber access provided by Thompson Rivers University | Library

Article
Self-Assembled Peptide-Lanthanide Nanoclusters for Safe Tumor Therapy:
Overcoming and Utilizing Biological Barriers to Peptide Drug Delivery
Jin Yan, Wangxiao He, Siqi Yan, Fan Niu, Tianya Liu, Bohan Ma, Yongping
Shao, Yuwei Yan, Guang Yang, Wuyuan Lu, Yaping Du, Bo Lei, and Peter X Ma
ACS Nano, Just Accepted Manuscript • DOI: 10.1021/acsnano.8b00081 • Publication Date (Web): 27 Jan 2018
Downloaded from http://pubs.acs.org on January 28, 2018

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted
online prior to technical editing, formatting for publication and author proofing. The American Chemical
Society provides “Just Accepted” as a free service to the research community to expedite the
dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts
appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been
fully peer reviewed, but should not be considered the official version of record. They are accessible to all
readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered
to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published
in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just
Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor
changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers
and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors
or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Nano is published by the American Chemical Society. 1155 Sixteenth Street
N.W., Washington, DC 20036
Published by American Chemical Society. Copyright © American Chemical Society.
However, no copyright claim is made to original U.S. Government works, or works
produced by employees of any Commonwealth realm Crown government in the course
of their duties.
Page 1 of 25 ACS Nano

1
2
3
4
5 TOC
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 2 of 25

1
2
3
4
5 Self-Assembled Peptide-Lanthanide Nanoclusters for Safe Tumor Therapy:
6
7 Overcoming and Utilizing Biological Barriers to Peptide Drug Delivery
8
9
10
11
12 Jin Yan 1†, Wangxiao He 2,5,* †, Siqi Yan 3†, Fan Niu 2, Tianya Liu 2, Bohan Ma 2, Yongping Shao 2, Yuwei
13
14
15 Yan 2, Guang Yang 4, Wuyuan Lu 2, 5, , Yaping Du 1, Bo Lei 1,*, Peter X Ma 1, 6
16
17
18
19 1
Frontier Institute of Science and Technology, State Key Laboratory for Mechanical Behavior of Materials, Xi’an
20
21
22 Jiaotong University, Xi’an 710049, China.
23
24 2
Center for Translational Medicine, Key Laboratory of Biomedical Information Engineering of Ministry of Education,
25
26
27 School of Life Science and Technology and Frontier Institute of Science and Technology, Xi’an Jiaotong University,
28
29 Xi’an 710049, China.
30
31 3
32 Department of Ophthalmology, the First Affiliated Hospital, Health Science Center of Xi’an Jiaotong University,
33
34 Xi’an 710049, China. 4 Department of oncology, BenQ Medical Center, Nanjing Medical University, Nanjing 210029,
35
36
China.
37
38
39 5
Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School
40
41
of Medicine, Baltimore, MD 21201, USA.
42
43
6
44 Department of Biologic and Materials Sciences, Department of Biomedical Engineering, Macromolecular Science and
45
46 Engineering Center, Department of Materials Science and Engineering, University of Michigan, Ann Arbor 48109,
47
48
49 USA
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 3 of 25 ACS Nano

1
2
3
4
5
6
7
8
9
10 ABSTRACT: Developing sophisticated nanomedicine platform to deliver therapeutics effectively and safely
11
12 into tumor/cancer cells remains challenging in nanomedicine field. In particular, reliable peptide drug delivery
13
14
15 systems capable of overcoming biological barriers are still lacking. Here, we developed a simple, rapid and robust
16
17 strategy to manufacture nanoclusters of ~90 nm in diameter that are self-assembled from lanthanide-doped
18
19
nanoparticles (5nm), two anti-cancer peptides with different targets (BIM and PMI) and one cyclicpeptide iNGR
20
21
22 targeted to cancer cells. The peptide-lanthanide nanoclusters (LDC-PMI-BIM-iNGR) enhanced the resistance of
23
24 peptide drugs to proteolysis, disassembled in response to reductive conditions that are present in the tumor
25
26
27 microenvironment and inhibited cancer cell growth in vitro and in vivo. Notably, LDC-PMI-BIM-iNGR exhibited
28
29 extremely low systemic toxicity and side effect in vivo. Thus, the peptide-lanthanide nanocluster may serve as an
30
31
32 ideal multifunctional platform for safe, targeted and efficient peptide drug delivery in cancer therapy.
33
34 KEYWORDS: multifunctional nanocluster, stimuli-responsive, peptides, targeted delivery, p53 activator, safe
35
36
37 cancer therapy
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 4 of 25

1
2
3
4
5
6
7
8
9
10 Over the past few decades, nanotechnology has emerged as a promising means to deliver anticancer
11
12 therapeutics for its preferential and selective accumulation at tumor sites.1 However, the clinical applications of
13
14
15 most nanomedicines are hampered by the lack of specific localization of the therapeutic agents at sites of interest.2
16
17 Chemotherapeutic drugs escaped from target site often distribute freely and kill healthy cells, resulting in
18
19
undesirable side effects and limited therapeutic efficacies. The lack of efficacy and clinical safety for
20
21
22 nanotherapeutic systems remain the principal causes of failure in clinical application.
23
24 An ideal nanotherapeutic system should not only deliver therapeutic agents to desired sites effectively, but
25
26
27 also manage to obliterate the off-target side effect. Such requirements have promoted the recent development of
28
29 stimuli-responsive nanoparticles, which can release the drugs at the target site under the trigger of tumor
30
31
32 microenvironment.3-5 Although conceptually impressive, those systems are still at an early stage of development
33
34 due to their narrow therapeutic windows attributed to the toxic side effects of the small molecules. Peptide drugs,
35
36
37 with higher specificity and less side effects, offer ideal alternatives to small molecules as therapeutics in
38
39 nanotherapeutic system.6 However, peptide drugs encounter a series of biological obstacles including rapid
40
41
degradation by enzymolysis, lack of accumulation at the tumor sites, and weak internalization by cancer cells.7-9
42
43
44 Although various strategies have been proposed to overcome these obstacles to peptide application, to our
45
46 knowledge, none of them was able to solve all these problems at the same time.
47
48
49 With the enhanced permeability and retention effect (EPR) in tumor tissues as well as effective cellular
50
51 internalization ability,10,11 nanoparticles provide an ideal solution to targeted delivery and stimuli-responsive
52
53
54 release of peptide drugs. Besides, in the delivery process, nanoplatform can protect peptides from enzymolysis.
55
56 Importantly, while low proteolytic stability is one of the biggest obstacles for peptide drug application, this
57
58
59
60 ACS Paragon Plus Environment
Page 5 of 25 ACS Nano

1
2 property can be exploited for rapid clearance of peptide drugs after their release from nanoplatform to prevent
3
4
5 extended off-target toxicity. Thus, nanoplatform-based peptide delivery system holds the promise of overcoming
6
7 the aforementioned biological barriers all at once.
8
9
10 The p53 tumor suppressor pathway is perturbed in the majority of human cancers.12,13 Although p53 gene is
11
12 frequently mutated or deleted, many cancer cells have WT p53 gene except that its expression is inhibited by
13
14
15 overexpression of the negative regulator MDM2.14,15 MDM2 is an E3 ubiquitin ligase that targets p53 for
16
17 poly-ubiquitination which leads to proteasomal degradation of p53.15,16 Therefore, disrupting the interaction
18
19
between MDM2 and p53 to restore p53 expression and its tumor suppressor activity is a promising strategy to
20
21
22 treat cancers.17 PMI is a potent inhibitor of MDM2-p53 interaction discovered by phage display screening and
23
24 subsequent mutational optimization process.18 PMI can activate p53 efficiently, and activate the downstream
25
26
27 BH3-only family protein (such as Bim, PUMA and NOXA) to promote apoptosis by sequestering anti-apoptotic
28
29 Bcl-2 family proteins from BAX and BAK.18-20 However, the tumor-inhibiting activity of PMI is greatly limited
30
31
32 by its low proteolytic stability and poor cell-member penetration.18 Therefore, it is meaningful to overcome these
33
34 biological obstacles of PMI by nanoplatform-based peptide delivery system. What’ more, BIM peptide is the BH3
35
36
37 domain of the pro-apoptotic protein, Bim.21-23 Hence, it is rationale to combine BIM with PMI to get a better
38
39 anti-tumor efficacy.
40
41
Herein, we report the development of a peptide-lanthanide clustered nanosystem, named
42
43
44 LDC-PMI-BIM-iNGR, which effectively overcomes barriers to peptide application in cancer therapy, and
45
46 achieves safe and reliable antitumor efficacy in vitro and in vivo (Scheme 1). LDC-PMI-BIM-iNGR was
47
48
49 constructed through a thiol-poly(ethyleneglycol)-thiol (SH-PEG-SH) polymer-induced molecular assembly of
50
51 three peptides (PMI, BIM, and a cyclicpeptide iNGR targeted to cancer cells) with biocompatible
52
53
54 lanthanide-doped fluorescent nanoparticles (LDN, GdOF:45%Ce,15%Tb) (Scheme 1).17, 24, 25 For peptide delivery,
55
56 LDC-PMI-BIM-iNGR was designed to protect the peptide cargos (PMI and BIM) from enzymolysis, accumulate
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 6 of 25

1
2 in tumor tissues with the guidance of iNGR peptide, get internalized by the tumor cells (Scheme 1), and then
3
4
5 stimuli-responsively release potent peptide drugs for robust antitumor efficacy (Scheme 1).
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28 Scheme 1. Schematic diagram of LDC-PMI-BIM-iNGR serving as a smart and safe peptide delivery platform for targeted
29
30
cancer therapy and labeling. The lanthanide-doped nanoclusters (LDC) were constructed through a
31
32
33 thiol-poly(ethyleneglycol)-thiol (SH-PEG-SH) (II) induced molecular assembly of anti-cancer peptides (PMI&BIM (I)) and
34
35 targeting peptide (iNGR (III)) with ligand-free lanthanide-doped fluorescent nanoparticles (LDN). These nanoclusters can
36
37
38 protect the peptides from enzymolysis, target tumor cells specifically and get internalized by tumor cells. In reductive cytosol,
39
40 LDC can be reduced to release potent peptide drugs to kill cancer cells.
41
42
43 RESULTS AND DISCUSSION
44
45 Preparation and characterization of stimuli-responsive LDC-PMI-BIM-iNGR nanoclusters. LDN
46
47
48 capable of conjugating free sulfhydryl group was synthesized as previously described,17 and reacted with
49
50 hydrochloric acid (pH 4.0) to expose the surface.18 The three peptides, cysteine modified PMI (PMI-Cys) and
51
52
BIM (BIM-Cys) as well as sulfhydryl PEG modified iNGR (iNGR-PEG-SH) were synthesized by total chemical
53
54
55 synthesis and site-directed modification (Scheme 1). The molecular weights of peptides and pharmacological
56
57 features were characterized by electrospray ionization-mass spectrometry (ESI-MS), high-performance liquid
58
59
60 ACS Paragon Plus Environment
Page 7 of 25 ACS Nano

1
2 chromatography (HPLC) and isothermal titration calorimetry (ITC) (Fig. S1).
3
4
5 For the synthesis of LDC-PMI-BIM-iNGR, the anti-cancer peptides, PMI-Cys and BIM-Cys, were firstly
6
7 conjugated with LDN to obtain LDN-PMI-BIM (Scheme 1). Briefly, 5 mg of LDN was dissolved in 10 ml of
8
9
10 reaction buffer (20% acetonitrile and 80% standard PBS) containing 1 mM PMI-Cys and 1 mM BIM-Cys. After a
11
12 20-min incubation with stirring,1 mg of HS-PEG-SH was added to crosslink LDN-PMI-BIM (about 4 nm) into
13
14
15 nanoclusters with a size of around 90 nm in diameter. Finally, 1 mg of sulfhydryl-PEGylated targeting peptide
16
17 iNGR-PEG-SH was added to the mixture to saturate the free sulfhydryl groups at the surface of the
18
19
LDC-PMI-BIM (Scheme 1). After 40-min incubation at 35ºC with stirring,~7mg LDC-PMI-BIM-iNGR can be
20
21
22 collected by centrifugation (the yield of ~60%).
23
24 The successful conjugations of peptides with LDN were confirmed by Fourier transform infrared spectra
25
26
27 (FTIS, Fig. 2B) and ultraviolet–visible spectra (UVS, Fig.S2A). In FTIS, a new peak at 1650 cm-1 were detected
28
29 in LDN-PMI-BIM and LDN-PMI-BIM-iNGR samples, which were attributed to the stretching vibration C=O
30
31
32 groups from peptides (Fig.2B). Meanwhile, more sharp-angled peak at 3400cm-1 (characteristic peak of N-H
33
34 stretching vibration) can be founded in LDN-PMI-BIM and LDN-PMI-BIM-iNGR samples, which was further
35
36
37 evidence of the successful conjugations of peptides with LDN. In UVS, after peptide conjugation, two typical
38
39 UV-vis absorption peaks of aromatic structures and peptides at 325 nm and 280 nm can be observed (Fig. S2A).
40
41
To further determine the efficiency of peptide loading in the nanoclusters,LDC-PMI-BIM-iNGR was dissolved in
42
43
44 PBS buffer containing 6 M guanidine hydrochloride and 1 M dithiothreitol to break the conjunction between
45
46 peptide and LDN, and then the amount of released peptides were quantified by high-performance liquid
47
48
49 chromatography (HPLC, Fig. S2B). Our results showed that the loading quantities of the PMI-Cys and BIM-Cys
50
51 were 25.2 mmol/mg and 12 mmol/mg, respectively. (mmol/mg=peptide molarity/LDN mass).
52
53
54 High-resolution TEM (HRTEM, Fig.1C) revealed that the LDN cores in LDC-PMI-BIM-iNGR remained as
55
56 uniform nanocrystals with a mean diameter of 5.6 ± 0.4 nm (Fig. 1C1). The clear-cut lattice fringes shown in the
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 8 of 25

1
2 Fig.1C2 HRTEM images and the symmetric faculaes shown in the Fig.1 C3 fast Fourier transform (FFT) patterns,
3
4
5 verified that the LDN core kept a high nature of crystallization with single crystalline structure after peptide and
6
7 PEG modification. Additionally, LDC-PMI-BIM-iNGR is well dispersed in water, and emits strong green light
8
9
10 and weak red light under excitation at 365 nm (Fig.1D, Fig.S3A), a feature that can be utilized for nuclear and
11
12 cytoplasmic imaging (Fig.S3B).
13
14
15 The molecular assembly and stimuli-responsive properties (Fig.1A) of smart LDC-PMI-BIM-iNGR
16
17 nanoclusters were characterized by monitoring changes in their morphologies and in the hydrodynamic diameter
18
19
distribution, using transmission electron microscopy (TEM) and dynamic light scattering (DLS), respectively
20
21
22 (Fig.1E&F). LDN showed a spherical nanostructure with a hydrodynamic diameter of about 4.1 nm (Fig.1E&F).
23
24 In sharp contrast, as shown in the TEM image of LDC-PMI-BIM-iNGR in Fig. 1E, the self-assembled
25
26
27 nanoclusters had a bigger raspberry-like structure, which presented a monodispersed nanocluster morphology
28
29 composed of several LDN cores. The mean hydrodynamic diameter of LDC-PMI-BIM-iNGR was around 89.3 nm
30
31
32 (Fig. 1F), which was consistent with TEM observation and was an advantageous size for improved
33
34 pharmacokinetics and vascular extravasation.26, 27 More importantly, LDC-PMI-BIM-iNGR showed a sensitive
35
36
37 redox-responsive behavior in an intracellular-mimic reducing environment28 (10mM dithiothreitol, DTT, in ph7.4
38
39 PBS). After reduction for 4 h, the LDC-PMI-BIM-iNGR presented a monodispersed nanocrytal morphology with
40
41
a significantly decreased hydrodynamic diameter (about 5.0 nm), demonstrating the stimuli-responsive
42
43
44 disassembly process (Figs.2E&F).
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 9 of 25 ACS Nano

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
Figure 1. Synthesis and physicochemical properties of smart LDC-PMI-BIM-iNGR nanoclusters. (A) Schematic illustration
31
32
33 showing the self-assembly of LDN into the redox-sensitive LDC-PMI-BIM-iNGR nanocluster, and the disintegration of the
34
35 nanocluster into small particles in the reducing intracellular environment; (B) FTIR spectra of LDN, LDC-PMI-BIM and
36
37
38 LDC-PMI-BIM-iNGR, demonstrating their surface chemical structures before and after self-assembly; (C) HRTEM images
39
40 (C1 and C2) and corresponding FFT pattern (C3) of an individual particle of LDC-PMI-BIM-iNGR, exhibiting single crystal
41
42
43 structures; (D) Fluorescence spectra and quantum yield of LDN, LDC-PMI-BIM-iNGR before and after responding to
44
45 reducing milieu, significant stimuli-responsive fluorescence-switch property was observed; (E) TEM images and (F)
46
47
hydrodynamic distributions of ligand-free LDN, LDC-PMI-BIM-iNGR showing the self-assembly and disassembly process of
48
49
50 nanocluster system responding to reducing milieu.
51
52
Excellent performance on proteolysis resistance and stimuli-responsive release of peptide cargos. As
53
54
55 our design indicates, LDC-PMI-BIM-iNGR is capable of overcoming enzymolysis during peptides delivery by
56
57 providing steric hindrance against the enzyme binding (Fig. 2A). For proteolysis resistance test, all samples were
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 10 of 25

1
2 incubated in an extracellular-milieu mimicking phosphate buffer solution with oxidized glutathione and
3
4
5 chymotrypsin, a protease with dual specificities for both basic and bulky hydrophobic residues (Both PMI and
6
7 BIM peptides have many hydrophobic residues). Compared with non-clustered LDN-PMI-BIM-iNGR (half-time,
8
9
10 22.1h), LDC-PMI-BIM-iNGR showed a significantly higher resistance against enzymolysis (half-time>48h),
11
12 whereas the free peptides had a half-life less than 6 min (Fig.2B). Another designed function of
13
14
15 LDN-PMI-BIM-iNGR is to release peptide drugs in response to the reductive intracellular environment. To assess
16
17 the reduction-induced release of peptides from LDC-PMI-BIM-iNGR, nanoclusters were incubated with PBS
18
19
buffer containing 10 mM DTT (mimicking intracellular reductive condition).28 As shown in Fig. 2C, PMI and
20
21
22 BIM peptides can be released from the nanoclusters in a redox-dependent manner. These results demonstrated the
23
24 superior performance of LDC-PMI-BIM-iNGR on proteolytic stability and stimuli-responsive release of peptide
25
26
27 cargos.
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56 Figure 2. Excellent performance on proteolysis resistance and stimuli-responsive release of peptides. (A) Schemata of the
57
58
59
60 ACS Paragon Plus Environment
Page 11 of 25 ACS Nano

1
2 superior functions of LDC-PMI-BIM-iNGR on proteolysis resistance and peptide drug delivery (B) LDC-PMI-BIM-iNGR protects
3
4
5 peptides from proteolysis by chymotrypsin; (C) Redox-dependent release of peptides from LDC-PMI-BIM-iNGR at pH 7.4.
6
7 LDC-PMI-BIM-iNGR inhibits the viability of cancer cells in a p53 pathway-dependent manner. As
8
9
10 expected, LDC-PMI-BIM-iNGR, LDC-PMI-BIM, LDC-PMI-iNGR dose dependently inhibited the growth of the
11
12 p53 wild type cancer cell line HCT116 p53+/+ with IC50 values of 81 nM, 166 nM and 125 nM (PMI concentration)
13
14
15 respectively. By contrast, the positive control drug Nutlin-3 showed an IC50 of 5.5 µM while free PMI exhibited
16
17 no inhibitory effect (Fig. 3A). Moreover, according to the western blotting analysis (normalized against Actin
18
19
protein), after treatment with LDC-PMI-BIM-iNGR, LDC-PMI-iNGR or Nutlin-3, the expressions of p53, MDM2,
20
21
22 p21, NOXA and PUMA proteins in HCT116p53+/+ cells were significantly increased, indicating the activation of
23
24 p53 pathway (Fig. 3B&Fig. S5A-D). Interestingly, an isogenetic p53 null cell line, HCT116 p53-/- were highly
25
26
27 resistant to the treatment of LDC-PMI-BIM-iNGR (Fig. 3C), suggesting that the tumor-inhibiting activity of our
28
29 nanoclusters is p53-dependent. Additionally, Annexin V- PI staining further revealed that LDC-PMI-BIM-iNGR
30
31
32 induced apoptotic rather than necrotic cell death in HCT116 p53+/+ cells (Fig.3D). Together, these findings strongly
33
34 support that LDC-PMI-BIM-iNGR can reduce the viability of HCT116p53+/+ cells by reactivating the p53 pathway
35
36
37 and addition of the BIM peptide can potentiate the anti-tumor efficacy of PMI.22, 23
38
39 Real-time Monitoring of morphological changes of nucleus during LDC-PMI-BIM-iNGR induced
40
41
apoptosis. We have previously shown that LDN had a distinctive capability to emit two different fluorescences,
42
43
44 and it can be used as a probe to simultaneously label the nucleolus yellow and the cytoplasm green.29 Therefore,
45
46 LDC-PMI-BIM-iNGR is endowed with a capability to monitor the pharmacological effects of peptide drugs on
47
48
49 the morphology of subcellular structures in real time (Fig. 3E&Fig. S6). After 24 h treatment of
50
51 LDC-PMI-BIM-iNGR, a small population of cancer cells (~12.8%) underwent early apoptosis (Fig.3F&S7),
52
53
54 which was characterized by the collapse of the chromatin against the nuclear periphery and fusion into a few large
55
56 clumps (circle 1 in Fig.3E1).30, 31 After 48 h, some nucleuses were condensed into a single dense sphere (circle 2
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 12 of 25

1
2 in Fig.3E2), and other chromatins budded outward into smaller sphere surrounded by nuclear envelope (circle 3 in
3
4
5 Fig.3E2), both of which were consistent with the increased apoptosis level (41.4%, Fig.3F&S7).30, 31 After 72 h,
6
7 96.2% of cells were dead or in apoptosis (Fig.S7) and the cell nucleus showed important features of the late
8
9
10 apoptosis: diffused and granular apoptotic bodies (circle 4 in Fig.3E3) or ring-like structures (circle 5 in
11
12 Fig.3E3).32, 33 Of note, these morphology changes of nucleus were also confirmed by a Hoechst staining in situ
13
14
15 (Fig. S6). This imaging capability of LDC-PMI-BIM-iNGR may have promising applications for cell morphology
16
17 test and drug efficacy evaluation in the development of anti-cancer drugs as well as in the clinical settings.
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55 Figure 3. In vitro anti-cancer efficacy and therapy monitoring of LDC-PMI-BIM-iNGR. (A) Dose-dependent growth
56
57 inhibition of HCT116p53+/+ cells upon various treatments as determined by the Amar blue cell viability assay; (B) Protein
58
59
60 ACS Paragon Plus Environment
Page 13 of 25 ACS Nano

1
2 expression analysis of p53, p21, MDM2, NOXA and PUMA in HCT116p53+/+ cells after 48 h treatment with 100 nM
3
4
5 LDC-PMI-BIM-iNGR and LDC-PMI-iNGR by western blot, normalized against Actin; (C) Growth inhibition of
6
7 HCT116p53+/+ and HCT116p53-/- cells 72 h after treatment with 100 nM LDC-PMI-BIM-iNGR and LDC-PMI-iNGR; (D)
8
9
10 Effects of Nutlin3a and LDC-PMI-BIM-iNGR on HCT116 p53+/+ cells as determined by Annexin V-PI staining and flow
11
12 cytometric analysis; (E) CLSM images of HCT116p53+/+ cells at 24 h (E1), 48 h (E2) and 72 h (E3) after 100 nM
13
14
LDC-iNGR-PMI-BIM treatment, scale bars: 60 µm; (F) Apoptosis levels measured by FACS in HCT116 p53+/+ cells treated
15
16
17 with 100 nM LDC-iNGR-PMI-BIM or 6µM Nutlin-3a for 24-72 hours.
18
19
Targeted antitumor activity of LDC-PMI-BIM-iNGR in vivo. The excellent anti-cancer and
20
21
22 stimuli-responsive performance of LDC-PMI-BIM-iNGR in vitro further compelled us to study its in vivo activity.
23
24 Mice bearing HCT116p53+/+ tumors were used to investigate the tumor targeting ability and therapeutic efficacy of
25
26
27 our nanoclusters. The targeting ability was determined by tracking the LDN fluorescent signals, which can be
28
29 used to gauge the quantity and distribution of LDC-PMI-BIM-iNGR in tumor and normal tissues (Figs. 4A-B).
30
31
32 Facilitated by the tumor-targeting iNGR peptide and the EPR effect of nanoclusters, LDC-PMI-BIM-iNGR is
33
34 expected to be primarily taken by tumor tissues but much less by the normal tissues (Fig.4A). 10, 11, 25, 26 Indeed, a
35
36
37 strong fluorescent signal was detected at the tumor site after 4 h post-injection, while only a low (kidney and liver)
38
39 or background (brain, lung, spleen and heart) level of fluorescence was observed in normal tissues, confirming the
40
41
tumor-targeting ability of LDC-PMI-BIM-iNGR nanoclusters (Fig. 4B).
42
43
44 To test the therapeutic efficacy of LDC-PMI-BIM-iNGR, xenograft tumors were established and randomly
45
46 divided into 7 groups (n = 5/group) to respectively receive a 2-week treatment regimen as follows: PBS,
47
48
49 doxorubicin, PMI peptide, PMI&BIM, LDC-PMI-iNGR, LDN-PMI-BIM-iNGR, or LDC-PMI-BIM-iNGR (The
50
51 dosage of all groups were 2.5 mg/kg). The volumes of tumors were measured every day. As shown in Fig. 4C, the
52
53
54 free PMI peptide or the combination of free PMI and BIM peptides displayed hardly any therapeutic effects when
55
56 compared to the PBS control, whereas LDC-PMI-iNGR and LDN-PMI-BIM-iNGR inhibited tumor growth by
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 14 of 25

1
2 76.8% and 75.3% respectively on day 12, close to the inhibition level of the clinical chemotherapeutics
3
4
5 Doxorubicin (DOX), 76.8%. LDC-PMI-BIM-iNGR exhibited the strongest suppression of tumor growth (90.1%).
6
7 The average weights of the tumors excised at the end of the experiment also demonstrated the same trend (Fig. 4D
8
9
10 & Fig. S8). The geometric mean of the tumor weights in the LDC-PMI-BIM-iNGR group (19.4mg) were just 21%
11
12 of that in the LDN-PMI-BIM-iNGR group (90.5 mg, P <0.01),suggesting the cluster structure significantly
13
14
15 improve the therapeutic efficacy. Meanwhile, as we expected, the mean tumor weights of the LDC-PMI-iNGR
16
17 group (82.2 mg) were 4.2-fold higher than that of the LDC-PMI-BIM-iNGR group, indicating that BIM peptide
18
19
can enhance the therapeutic efficacy of PMI. To further characterize the anti-tumor efficacy in vivo at pathological
20
21
22 level, histological analyses, including hematoxylin and eosin (H&E, Fig. 4E) staining, and terminal
23
24 deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end (TUNEL, Fig4F) staining were carried
25
26
27 out. As shown in Fig. 4E, the dense texture and morphologic integrity of tumor tissues can be observed in control,
28
29 PMI and PMI/BIM-combo groups. By contrast, necrotic tumor tissues were observed in DOX,
30
31
32 LDN-PMI-BIM-iNGR and LDC-PMI-BIM groups (Fig.4E). As expected, LDC-PMI-BIM-iNGR treated groups
33
34 showed large areas of necrotic tumor with numerous apoptotic cancer cells (Fig.4E). Furthermore, TUNEL
35
36
37 staining (brown) was performed to determine the level of apoptosis in tumor tissues (Fig.4F).34 Compared with the
38
39 other groups, the LDC-PMI-BIM-iNGR group exhibited apparently stronger staining in the tumor regions,
40
41
demonstrating that much more cancer cells entered the apoptotic process. These results indicated that
42
43
44 LDC-PMI-BIM-iNGR markedly impedes the growth of the tumors by promoting apoptosis in vivo.
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 15 of 25 ACS Nano

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38 Figure 4. Targeted in vivo antitumor activity of LDC-PMI-BIM-iNGR in human colon tumor model. (A) Schematic diagram
39
40 of targeted accumulation process of LDC-PMI-BIM-iNGR in tumor region; (B) Ex vivo fluorescent images of tumors and
41
42
43 major organs from LDC-PMI-BIM-iNGR treated mice; (C) Tumor growth curves of indicated treatment groups; (D) The
44
45 average weights of the tumors excised at the end of the experiment, ** stand for P<0.01; (E) H & E ( ×200) and (F) TUNEL
46
47
(×200) staining of HCT116p53+/+ solid tumors tissues after various treatments for 12 days
48
49
50 Therapeutic safety evaluation on LDC-PMI-BIM-iNGR in vitro. Systemic toxicity from the freely
51
52
distributed off-target drugs poses a major challenge to cancer chemotherapy. To minimize the off-target effects, a
53
54
55 dual-targeting strategy was adopted in our design of LDC-PMI-BIM-iNGR nanocluster (Fig. 5A). Firstly, the
56
57 Asn–Gly–Arg (NGR) sequence in iNGR peptide can specifically bind to CD13, which are highly expressed in
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 16 of 25

1
2 diverse cancer cell lines.24 Then, the internalization process of iNGR can be triggered when the C-end rule motif
3
4
5 sequence tNGR (CRNGR) is exposed through a proteolytic process that specifically occurs in cancer cells.16 This
6
7 strategy allows enrichment of LDC-PMI-BIM-iNGR specifically in CD13-over-expressed cancer cells. Indeed,
8
9
10 we observed that LDC-PMI-BIM-iNGR preferentially labeled the cancer cells (HCT116) while not the normal
11
12 cells (BMSCs and HUVECs) with bright fluorescence (Fig.5B). The second level of specificity of our
13
14
15 nanoclusters results from the PMI peptide, whose target, MDM2 is only overexpressed in cancer cells but not in
16
17 the normal cells. In accordance with this, LDC-PMI-BIM-iNGR had no inhibitory activity for the normal cells
18
19
BMSC and HUVEC, as well as the untargeted p53 null cell line, SW480, whereas DOX induced growth inhibition
20
21
22 for all cell lines (Figs. 5C-D).
23
24 The low proteolytic stability of peptides advantageously offers another level of protection against off-target
25
26
27 effects. Peptide drugs can be rapidly degraded before they diffuse and bind to unwanted targets. For instance, 100
28
29 nM LDC-PMI-BIM-iNGR effectively induced a time-dependent growth inhibition in various cancer cells, such as
30
31
32 HCT116p53+/+, SJSA (a cell line from primitive multipotential sarcoma of femur) and MCF-7 (a breast cancer cell
33
34 line), whereas free PMI exhibited no inhibitory activity even at 6 µM (Fig.5E). Meanwhile, according to previous
35
36
37 reports,21 the free BIM peptide has no inhibitory activity for cancer cells at a concentration within the nM range.
38
39 These results suggested that PMI or BIM peptides extracellulary leaked from the nanoclusters would not have any
40
41
effects to cells.
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 17 of 25 ACS Nano

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46 Figure 5. Therapeutic safety evaluation of LDC-PMI-BIM-iNGR in vitro. (A) Schematic diagram of tumor therapeutic safety
47
48
49 benefitted from LDC-PMI-BIM-iNGR; (B) Confocal laser scanning microscope (CLSM) images of HCT116, BMSC and
50
51 HUVEC cells after incubation with LDC-PMI-BIM-iNGR (20 µg/mL), all images were taken under the same exciting light
52
53
54 and detector gain (scale bars 60 µm); (C) Selective cancer cell cytotoxicity of LDC-PMI-BIM-iNGR determined by
55
56 LIVE/DEAD staining (×600); (D) Selective cytotoxicity of LDC-PMI-BIM-iNGR against HCT116p53+/+, SW480, HMSC and
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 18 of 25

1
2 HUVEC, using DOX as a positive control; (E) Time-dependent cytotoxic activity of 6 µM nutlin-3, 6 µM free PMI and 100
3
4
5 nM LDC-PMI-BIM-iNGR against HCT116p53+/+ (E1), SJSA (E2) and MCF-7 (E3) cell lines.
6
7 Therapeutic safety evaluation on LDC-PMI-BIM-iNGR in vivo. To further verify the biosafety of
8
9
10 LDC-PMI-BIM-iNGR in vivo, drug toxicity tests were carried out on three randomly divided mice groups (n =
11
12 6/group) by treating each individual group with PBS, doxorubicin or LDC-PMI-BIM-iNGR (5 mg/kg for all
13
14
15 groups) respectively for one week. Physical appearance and body weight were monitored every two days (Figs.
16
17 6A-C), and major organs were excised at the end of the experiment. After 1 W administration, the body weight of
18
19
mice in the LDC-PMI-BIM-iNGR group showed little difference from the control group (PBS), whereas the
20
21
22 DOX-treated group lost weight significantly (Fig.6B). Meanwhile, mice in the DOX group showed symptoms of
23
24 adynamia, dehydration and fading of eye color, but the LDC-PMI-BIM-iNGR group of mice remained as healthy
25
26
27 as the control group (Fig. 6C). Histologically, no morphological or pathological changes were observed in livers,
28
29 kidneys and lungs of the mice from the control or LDC-PMI-BIM-iNGR group (Fig.6D), whereas some
30
31
32 significant pathological changes, such as necrosis and fatty degeneration in liver, necrosis and inflammation in
33
34 kidney as well as thickening of alveolar walls in lung, were discovered in the DOX group (Fig.6A&D). These
35
36
37 results further demonstrated the biosafety of our LDC-PMI-BIM-iNGR nanocluster as a cancer therapeutic
38
39 regime.
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment
Page 19 of 25 ACS Nano

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32 Figure 6. Biosafety evaluation of LDC-PMI-BIM-iNGR in vivo. (A) Schematic diagram of tumor therapeutic safety benefitted
33
34 from LDC-PMI-BIM-iNGR; (B) Changes in body weight of nude mice with tumors upon various treatments; (C) Photos of
35
36
mice with tumors after 8 days of various treatments; (D) the representative histological H&E staining images of liver (×200),
37
38
39 kidney (×200) and lung (×200) tissue sections, demonstrating significantly lower tissue necrosis levels in
40
41
LDC-PMI-BIM-iNGR treated group.
42
43
44 CONCLUSIONS
45
46 In summary, a kind of peptide-lanthanide nanocluster (LDC-PMI-BIM-iNGR) with effective anti-cancer efficacy
47
48
49 in vitro and in vivo was fabricated facilely through self-assembly of LDN-Peptides ultra-small nanocrystals. These
50
51 nanoclusters demonstrated excellent capacity for targeted and safe cancer therapy while overcoming all biological
52
53
54 barriers to peptide drug delivery. LDC-PMI-BIM-iNGR platform could efficiently protect peptide drugs from
55
56 enzymolysis, selectively deliver them into the tumor cells and gradually release peptide drugs in response to
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 20 of 25

1
2 reductive intracellular environment. With the high tumor-targeting ability and short half-life of peptide drugs,
3
4
5 LDC-PMI-BIM-iNGR greatly mitigated systemic toxicity and side effects, thereby inhibiting tumor growth with
6
7 therapeutic safety. In brief, LDC-PMI-BIM-iNGR could serve as an ideal multifunctional platform for targeted
8
9
10 peptide drugs delivery with real-time tracking ability and superior therapeutic efficacy and safety.
11
12 EXPERIMENTAL SECTION
13
14
15 Fabrication of LDC-PMI-BIM-iNGR. The as-prepared LDN were re-dispersed in PBS (pH 7.4) buffered
16
17 saline (containing 20% acetonitrile). PMI, BIM or iNGR-PEG (Cys-Peptide or -SH) was conjugated with LDN by
18
19
the the sulfhydryl groups of cysteine of peptide using ligand chemistry. In detail, 5 mg of LDN was dissolved in
20
21
22 10 ml of reaction buffer (20% acetonitrile and 80% standard PBS) containing 1 mM PMI-Cys and 1 mM
23
24 BIM-Cys. After a 20-min incubation with stirring at 30℃,1 ml HS-PEG-SH solution (1 mg/ml in deionized water)
25
26
27 were added for further functionalized reaction at 30℃ for 1h. Finally, 1 mg of sulfhydryl-PEGylated targeting
28
29 peptide iNGR-PEG-SH was added to the mixture to saturate the free sulfhydryl groups at the surface of the
30
31
32 LDC-PMI-BIM. After 40-min incubation at 35ºC with stirring,~7mg LDC-PMI-BIM-iNGR can be collected by
33
34 10000g centrifugation (the yield of ~60%).
35
36
37 Physicochemical properties of the LDN and its conjugates. The nanocrystals morphology and lattice
38
39 structure were observed on a high-resolution transmission electron microscope (HRTEM, F20, FEI) operated at
40
41
200 kV. The hydrodynamic size distribution was obtained from the dynamic light scattering (DLS) measurement
42
43
44 (Malvern Zetasizer Nano ZS system). The surface chemical structure of modified nanocrystals was evaluated by
45
46 Fourier transform infrared (FT-IR) spectroscopy (Nicolet 6700) and UV−vis absorption spectra (Shimadzu 3000
47
48
49 spectrophotometer).
50
51 Proteolytic stability. Proteolytic resistance of free PMI, LDC-PMI-BIM-iNGR and LDN-PMI-BIM-iNGR
52
53
54 were incubated at 100 µM each in PBS with 10 µg/ml chymotrypsin – an intracellular protease with dual
55
56 specificities for both basic and bulky hydrophobic residues. RP-HPLC was used to monitor and quantify
57
58
59
60 ACS Paragon Plus Environment
Page 21 of 25 ACS Nano

1
2 time-dependent peptide hydrolysis.
3
4
5 Animal handling and tumor xenografts. Animal procedures were in agreement with the guidelines of the
6
7 Institutional Animal Care and Use Committee. Tumor cells were harvested when they reached near confluence by
8
9
10 incubation with 0.05% trypsin-EDTA. Cells were pelleted by centrifugation and resuspended in sterile PBS.
11
12 HCT116 cells (4 × 106cells/site) were implanted subcutaneously into bilateral hip of four- to five-weeks-old male
13
14
15 athymic nude mice. Treatment of three groups of mice (six mice per group) was initiated after the tumor had been
16
17 established in 2 weeks as a palpable mass (a volume of ~ 50mm3). Tumor length and width were measured with
18
19
calipers, and the tumor volume was calculated using the following equation: tumor volume(V)=length × width
20
21
22 × width / 2. For histological examination, the tumor, liver, kidney and lung tissues were fixed with
23
24 paraformaldehyde, dehydrated, sliced into 5.0µm sections and subjected to H&E or immune-histochemical
25
26
27 staining assay.
28
29 In vivo imaging and biodistribution analysis. Tumor bearing mice were injected with 200µl
30
31
32 LDC-PMI-BIM-iNGR (1mg/ml). IVIS Spectrum In Vivo Imaging System was used to take the ex vivo images of
33
34 mice. Light with a wavelength at 500 nm was used as the excitation source. Furthermore, mice were humanely
35
36
37 killed at 4 h post-injection, and then the heart, liver, spleen, lung, kidney and tumor were collected from each
38
39 mouse without delay. The fluorescence intensity in all organs was further analyzed by the IVIS Spectrum In Vivo
40
41
Imaging System.
42
43
44 ASSOCIATED CONTENT
45
46 Supporting Information
47
48
49 General information, synthetic procedures of LDN, synthesis and modification of the peptide, protocols of the
50
51 necessary cell experiments and 8 supplementary figures were included in Supporting Information . This material is
52
53
54 available free of charge via the Internet at http://pubs.acs.org
55
56 AUTHOR INFORMATION
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 22 of 25

1
2 Corresponding authors
3
4
5 * Email (B. Lei) rayboo@xjtu.edu.cn
6
7 * Email (W. He): WHe@ihv.umaryland.edu
8
9
10 Author Contributions
11
12 † J. Yan, W. He and S. Yan contributed equally to this work.
13
14
15 ACKNOWLEDGMENTS
16
17 The authors acknowledge the valuable comments of potential reviewers. This research was supported by State
18
19
Key Laboratory for Mechanical Behavior of Materials (No. 20161801), the Natural Science Basic Research Plan
20
21
22 in Shaanxi Province of China (No. 2015JQ5165) and National Natural Science Foundation of China (No.
23
24 51502237).
25
26
27 REFERENCES
28
29 (1) Peer, D.; Karp, J. M.; Hong, S.; Farokhzad, O. C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for
30
31
32 Cancer Therapy. Nat. Nanotechnol. 2007, 2, 751-760.
33
34 (2) Blanco, E.; Shen, H.; Ferrari, M. Principles of Nanoparticle Design for Overcoming Biological Barriers to Drug
35
36
Delivery. Nat. Biotechnol. 2015, 33, 941-951.
37
38
39 (3) Wong, C.; Stylianopoulos, T.; Cui, J.; Martin, J.; Chauhan, V. P.; Jiang, W.; Popović, Z.; Jain, R. K.; Bawendi, M. G.;
40
41
Fukumura, D. Multistage Nanoparticle Delivery System for Deep Penetration into Tumor Tissue. Proc. Natl. Acad. Sci. U.
42
43
44 S. A. 2011, 108, 2426-2431.
45
46 (4) Decuzzi, P.; Godin, B.; Tanaka, T.; Lee, S. Y.; Chiappini, C.; Liu, X.; Ferrari, M. Size and Shape Effects in the
47
48
49 Biodistribution of Intravascularly Injected Particles. J. Control. Release 2010, 141, 320-327.
50
51 (5) Zhang, P.; Cheng, F.; Zhou, R.; Cao, J.; Li, J.; Burda, C.; Min, Q.; Zhu, J.-J. DNA-Hybrid-Gated Multifunctional
52
53
54 Mesoporous Silica Nanocarriers for Dual-Targeted and MicroRNA-Responsive Controlled Drug Delivery. Angew. Chem.
55
56 Int. Edit. 2014, 53, 2371-2375.
57
58
59
60 ACS Paragon Plus Environment
Page 23 of 25 ACS Nano

1
2 (6) Bamrungsap, S.; Zhao, Z.; Chen, T.; Wang, L.; Li, C.; Fu, T.; Tan, W. Nanotechnology in therapeutics: a focus on
3
4
5 nanoparticles as a drug delivery system. Nanomedicine 2012, 7, 1253-1271.
6
7 (7) Morris, M. C.; Depollier, J.; Mery, J.; Heitz, F.; Divita, G. A Peptide Carrier for the Delivery of Biologically Active
8
9
10 Proteins into Mammalian Cells. Nat. Biotechnol. 2001, 19, 1173-1176.
11
12 (8) Fosgerau, K.; Hoffmann, T. Peptide Therapeutics: Current Status and Future Directions. Drug Discov. Today 2015, 20,
13
14
122-128.
15
16
17 (9) Almeida, A. J.; Souto, E. Solid Lipid Nanoparticles as a Drug Delivery System for Peptides and Proteins. Adv. Drug
18
19
Deliver Rev. 2007, 59, 478-490.
20
21
22 (10) Zhou, L.; Qu, X.; Guo, Y.; Wang, M.; Lei, B.; Ma, P.X. A Branched Glycerol-based Copolymer with Ultrahigh p65
23
24 siRNA Delivery Efficiency for Enhanced Cancer Therapy. ACS Appl. Mater. Inter. 2018, DOI: 10.1021/acsami.7b17109.
25
26
27 (11) Meng, H.; Xue, M.; Xia, T.; Ji, Z.; Tarn, D. Y.; Zink, J. I.; Nel, A. E. Use of Size and a Copolymer Design Feature to
28
29 Improve the Biodistribution and the Enhanced Permeability and Retention Effect of Doxorubicin-loaded Mesoporous
30
31
32 Silica Nanoparticles in a Murine Xenograft Tumor Model. ACS Nano 2011, 5, 4131-4144.
33
34 (12) Vogelstein, B.; Lane, D.; Levine, A. J. Surfing the p53 Network. Nature 2000, 408, 307-310.
35
36
(13) Levine, A. J.; Momand, J.; Finlay, C. A. The p53 Tumour Suppressor Gene. Nature 1991, 351, 453.
37
38
39 (14) Haupt, Y.; Maya, R.; Kazaz, A.; Oren, M. Mdm2 Promotes the Rapid Degradation of p53. Nature 1997, 387, 296.
40
41
(15) Kubbutat, M. H.; Jones, S. N.; Vousden, K. H. Regulation of p53 Stability by Mdm2. Nature 1997, 387, 299.
42
43
44 (16) Honda, R.; Tanaka, H.; Yasuda, H. Oncoprotein MDM2 is a Ubiquitin Ligase E3 for Tumor Suppressor p53. Febs Lett.
45
46 1997, 420, 25-27.
47
48
49 (17) Prives, C. Signaling to p53: Breaking the MDM2–p53 Circuit. Cell 1998, 95, 5-8.
50
51 (18) Pazgier, M.; Liu, M.; Zou, G.; Yuan, W.; Li, C.; Li, C.; Li, J.; Monbo, J.; Zella, D.; Tarasov, S. G. Structural Basis for
52
53
54 High-affinity Peptide Inhibition of p53 Interactions with MDM2 and MDMX. Proc. Natl. Acad. Sci. U. S. A. 2009, 106,
55
56 4665-4670.
57
58
59
60 ACS Paragon Plus Environment
ACS Nano Page 24 of 25

1
2 (19) Puthalakath, H.; Huang, D. C.; O’Reilly, L. A.; King, S. M.; Strasser, A. The Proapoptotic Activity of the Bcl-2 Family
3
4
5 Member Bim is Regulated by Interaction with the Dynein Motor Complex. Mol. Cell 1999, 3, 287-296.
6
7 (20) Bouillet, P.; Metcalf, D.; Huang, D. C.; Tarlinton, D. M.; Kay, T. W.; Köntgen, F.; Adams, J. M.; Strasser, A.
8
9
10 Proapoptotic Bcl-2 Relative Bim Required for Certain Apoptotic Responses, Leukocyte Homeostasis, and to Preclude
11
12 Autoimmunity. Science 1999, 286, 1735-1738.
13
14
(21) LaBelle, J. L.; Katz, S. G.; Bird, G. H.; Gavathiotis, E.; Stewart, M. L.; Lawrence, C.; Fisher, J. K.; Godes, M.; Pitter, K.;
15
16
17 Kung, A. L.; Walensky, L. D. A Stapled BIM Peptide Overcomes Apoptotic Resistance in Hematologic Cancers. J. Clin.
18
19
Invest. 2012, 122, 2018-2031.
20
21
22 (22) Tomita, Y.; Marchenko, N.; Erster, S.; Nemajerova, A.; Dehner, A.; Klein, C.; Pan, H.; Kessler, H.; Pancoska, P.; Moll, U.
23
24 M. WT p53, But Not Tumor-derived Mutants, Bind to Bcl2 via the DNA Binding Domain and Induce Mitochondrial
25
26
27 Permeabilization. J. Biol. Chem. 2006, 281, 8600-8606.
28
29 (23) Carter, B. Z.; Mak, P. Y.; Mak, D. H.; Ruvolo, V. R.; Schober, W.; McQueen, T.; Cortes, J.; Kantarjian, H. M.; Champlin,
30
31
32 R. E.; Konopleva, M.; Andreeff, M. Synergistic Effects of p53 Activation via MDM2 Inhibition in Combination with
33
34 Inhibition of Bcl-2 or Bcr-Abl in CD34(+) Proliferating and Quiescent Chronic Myeloid Leukemia Blast Crisis Cells.
35
36
Oncotarget 2015, 6, 30487-30499.
37
38
39 (24) Alberici, L.; Roth, L.; Sugahara, K.N.; Agemy, L.; Kotamraju, V.R.; Teesalu, T.; Bordignon, C.; Traversari, C.; Rizzardi,
40
41
G.P.; Ruoslahti, E. De Novo Design of a Tumor-Penetrating Peptide. Cancer Res. 2013, 73, 804-812.
42
43
44 (25) Sugahara, K. N.; Teesalu, T.; Karmali, P. P.; Kotamraju, V. R.; Agemy, L.; Girard, O. M.; Hanahan, D.; Mattrey, R. F.;
45
46 Ruoslahti, E. Tissue-Penetrating Delivery of Compounds and Nanoparticles into Tumors. Cancer Cell 2009, 16, 510-520.
47
48
49 (26) Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.; Kimura, M.; Terada, Y.; Kano, M.; Miyazono, K.;
50
51 Uesaka, M. Accumulation of Sub-100 nm Polymeric Micelles in Poorly Permeable Tumours Depends on Size. Nat.
52
53
54 Nanotechnol. 2011, 6, 815-823.
55
56 (27) Wang, L.; Huang, J.; Chen, H.; Wu, H.; Xu, Y.; Li, Y.; Yi, H.; Wang, Y. A.; Yang, L.; Mao, H. Exerting Enhanced
57
58
59
60 ACS Paragon Plus Environment
Page 25 of 25 ACS Nano

1
2 Permeability and Retention Effect Driven Delivery by Ultrafine Iron Oxide Nanoparticles with T1–T2 Switchable
3
4
5 Magnetic Resonance Imaging Contrast. ACS Nano 2017, 11, 4582-4592.
6
7 (28) Cheng, R.; Meng, F.; Ma, S.; Xu, H.; Liu, H.; Jing, X.; Zhong, Z. Reduction and Temperature Dual-responsive
8
9
10 Crosslinked Polymersomes for Targeted Intracellular Protein Delivery. J. Mater. Chem. 2011, 21, 19013-19020.
11
12 (29) Yan, J.; He, W.; Li, N.; Yu, M.; Du, Y.; Lei, B.; Ma, P. X. Simultaneously Targeted Imaging Cytoplasm and Nucleus in
13
14
Living Cell by Biomolecules Capped Ultra-small GdOF Nanocrystals. Biomaterials 2015, 59, 21-29.
15
16
17 (30) Wyllie, A. H.; Kerr, J. R.; & Currie, A. R. Cell death: the significance of apoptosis. Int. Rev. Cytol. 1980, 68, 251-306.
18
19
(31) Earnshaw, W. C. Nuclear Changes in Apoptosis. Curr. Opin. Cell Biol. 1995, 7, 337-343.
20
21
22 (32) Woo, M.; Hakem, R.; Soengas, M. S.; Duncan, G. S.; Shahinian, A.; Kägi, D.; Hakem, A.; McCurrach, M.; Khoo, W.;
23
24 Kaufman, S. A.; Senaldi, G.; Howard, T.; Lowe, S. W.; Mak, T. W. Essential Contribution of Caspase 3/CPP32 to Apoptosis
25
26
27 and Its Associated Nuclear Changes. Gene. Dev. 1998, 12, 806-819.
28
29 (33) Wadia, J. S.; Chalmers-Redman, R. M. E.; Ju, W. J. H.; Carlile, G. W.; Phillips, J. L.; Fraser, A. D.; Tatton, W. G.
30
31
32 Mitochondrial Membrane Potential and Nuclear Changes in Apoptosis Caused by Serum and Nerve Growth Factor
33
34 Withdrawal: Time Course and Modification by (−)-Deprenyl. J. Neurosci. 1998, 18, 932-947.
35
36
(34) Deng, X.; Wang, Y.; Chou, J.; Cadet, J. L. Methamphetamine Causes Widespread Apoptosis in the Mouse Brain:
37
38
39 Evidence from Using an Improved TUNEL Histochemical Method. Mol. Brain Res. 2001, 93, 64-69.
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 ACS Paragon Plus Environment

You might also like