Subscriber access provided by UNIV OF BARCELONA
Article
Tumor Microenvironmental pH and Enzyme Dual Responsive PolymerLiposomes for Synergistic Treatment of Cancer Immuno-Chemotherapy Ya Liu, Zeng-Ying Qiao, Pei-Pei Yang, Hao Wang, and Xiguang Chen Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.8b01510 • Publication Date (Web): 08 Jan 2019 Downloaded from http://pubs.acs.org on January 9, 2019
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 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 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.
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 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
1
Tumor Microenvironmental pH and Enzyme Dual Responsive
2
Polymer-Liposomes for Synergistic Treatment of Cancer
3
Immuno-Chemotherapy
4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Ya Liu*, Zeng-Ying Qiao, Pei-Pei Yang, Hao Wang and Xi-Guang Chen* Ya Liu and Prof. Xi-Guang Chen College of Marine Life Science Ocean University of China No. 5 Yushan Road, Qingdao (China) Tel/Fax.: 86-0532-82032586; E-mail:
[email protected] (Y. Liu);
[email protected] (X. G. Chen) Pei-Pei Yang, Zeng-Ying Qiao and Prof. Hao Wang CAS Center for Excellence in Nanoscience CAS Laboratory for Biomedical Effects of Nanomaterials and Nanosafety National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing (China)
1
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
20
Abstract
21
Despite recent advances in tumor treatment through cancer immunotherapy, the
22
efficacy of this approach remains to be improved. Looking forward to high rates of
23
objective clinical response, cancer immunotherapy combined with chemotherapy has
24
gained increasing attention recently. Here, we constructed liposomes with matrix
25
metalloproteinases (MMPs) responsive moiety and PD-L1 inhibitor conjugate
26
combine with low dose chemotherapy to achieve enhanced anti-tumor efficacy. Upon
27
introduction of the pH-responsive polymer to LPDp, the co-assembly could be almost
28
stable in physiological condition and tumor microenvironment and release the loaded
29
cargos at the lysosome. MMP-2 enzyme extracellularly secreted by the B16F10 cells
30
could cleave the crosslinker and liberate the PD-L1 inhibitor effectively disrupting the
31
PD-1/PD-L1interaction in vitro. Low dose DOX encapsulated in the LPDp was
32
capable of sensitizing B16F10 cells to CTLs by inducing overexpression of M6PR on
33
tumor cell membranes. In comparison with free PD-L1 inhibitor, LPDp improved the
34
biodistribution and on-demand release of the peptide inhibitor in tumor regions
35
following administration. LPDp achieved the optimal tumor suppression efficiency (~
36
78.7%), which demonstrated the significantly enhanced antitumor effect (P < 0.01)
37
than that of LPp (~57.5%) as well as that of LD (< 40%), attributing to synergistic
38
contribution from the substantial increase in M6PR expression on tumor cells and the
39
blockade of immune checkpoints. This strategy provides a strong rationale for
40
combining standard-of-care chemotherapy with relative non-toxic and high specific
41
immunotherapy.
42
2
ACS Paragon Plus Environment
Page 2 of 39
Page 3 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
43
1. Introduction
44
Cancer immunotherapy by training or stimulating the inherent immunological
45
systems of the body to attack tumor cells, has been progressing rapidly and shown
46
tremendous promises as a powerful cancer treatment strategy1-3. Several
47
well-established cancer immunotherapeutic approaches such as interleukin-2, immune
48
checkpoint inhibitors (CTLA-4 and PD-1 receptors), chimeric antigen receptor T-cell
49
immunotherapy (CAR-T) have improved the survival in cancer patients
50
the exciting clinical results of immunotherapy for the treatment of cancer, the efficacy
51
of the approach remained to be improved
52
from immunotherapy with most patients being insensitive with no or low responses 8, 9.
53
Challenges of cancer immunotherapy included the ability of vaccines to generate
54
effective immune responses with the presence of numerous immunosuppressive
55
factors10,
56
parenchyma
57
immunization correctly
58
especially combination therapy, call for urgent in-depth investigations.
59
11,
6, 7.
4, 5.
Despite
Only a subset of patients can benefit
the ability of cytotoxic T lymphocytes (CTLs) to infiltrate tumor 12,
recognize tumor-associated antigen, and choose antigen for 13, 14.
Strategies to improving the immune response rate,
Chemotherapy was usually associated with immunosuppressive effects and not 14-16.
60
previously considered as an attractive adjunct to cancer immunotherapy
Recent
61
data suggested that prior chemotherapy enhanced the therapeutic outcome of
62
immunotherapy and further reversed chemoresistance after prolonged chemotherapy
63
17-19.
64
anticancer effects and promote an immunogenic tumor phenotype. Several reports
Chemotherapeutic drugs combined with immunotherapy may result in enhanced
3
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 4 of 39
65
indicated low dose chemotherapeutic agents were capable of improving the
66
permeability of tumor cell membranes to granzyme B (GranB) via upregulation of
67
mannose–6–phosphate receptor (M6PR) on tumor cells20,
68
mediator of cell apoptosis by CTLs in cell-mediated immune response22, 23. Hence,
69
noncytotoxic concentrations of antitumor agents were able to increase the
70
susceptibility of tumor cells to CTLs24-26.
21.
GranB was a critical
71
We rationalized that combination of an immunotherapy agent with a
72
chemotherapeutics agent can be enabled using nanoscale vehicles for a maximal
73
antitumor outcome27,
74
kinase-inhibiting supramolecular therapeutics to inhibit tumor growth effectively and
75
prolong survival in tumor-bearing mice in various tumor model systems29. Current
76
cancer immunotherapies employing nanoparticles have paid close attention to the
77
immune checkpoint pathways for the activation of tumor-targeting CTLs
78
al. have combined checkpoint inhibitor aPD1 and 1-MT with nanoplatform and have
79
achieved great success in tumor immunotherapy32. While nanoparticles have recently
80
been used as vaccines to induce an enhanced immune reaction against the cancer
81
cells17, 33, combination regimens of immunotherapy with nanoparticles encapsulated
82
low dose chemotherapeutic agents in cancer are yet to be studied.
28.
Sengupta et al. have combined checkpoint inhibitors with
30, 31.
Gu et
83
Herein, we attempted to build up polymer-liposomes (LPDp) grafted with
84
anti-PD-L1 peptide34 and encapsulated low dose doxorubicin (DOX) for combination
85
of cancer immunotherapy and chemotherapy (Scheme 1). The pH-responsive polymer
86
chains contained anchoring moiety for tight attachment onto the liposome membrane 4
ACS Paragon Plus Environment
Page 5 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
87
surface (LP) and pH-sensitive moiety with capability of converting to protonated state
88
at acidic pH, accompanying the dynamic disordering of membrane. The mixing ratios
89
of lipid compositions with pH responders could be fine-tuned to optimize the
90
responsive sensitivity and specificity of LP. Low dose DOX was encapsulated in the
91
LPD with expect of sensitizing tumor cells to CTLs with negligible systemic side
92
effects. Hydrolysis resistant D-peptide (NYSKPTDRQYHF) as an antagonist to target
93
the PD-1/PD-L1 pathway was introduced into LPp via matrix metalloproteinases
94
(MMP) degradable cross-linkers (GPLGVRG)35, 36 to disrupt the immune-suppressing
95
pathway (Scheme 2). The bio-distribution of polymer-liposomes system in vivo was
96
evaluated via near-infrared imaging. Subsequently, LPDp was used as tumor
97
treatment in B16F10 mouse melanoma model and induced robust immune responses
98
with satisfactory therapeutic effects. We expect the MMPs/pH dual responsive
99
polymer-liposomes to provide an innovative strategy for combination of
100
immunotherapy and chemotherapy and facilitate the clinical treatment of melanoma.
101
2. Materials and methods
102
2.1.Materials
103
L-α-Phosphatidylcholine(PC),1,2-dioleoyl-3-trimethylammonium-propane(DOTAP
104
),cholesterol(CH),1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyet
105
hyleneglycol)-2000](DSPE-PEG),and1,2-distearoyl-sn-glycero-3-phosphoethanolami
106
ne-N-[maleimide(polyethyleneglycol)-2000](DSPE-PEG-mal) were purchased from
107
Avanti Polar Lipids. Dulbecco’s Modified Eagle Medium (DMEM), penicillin,
108
streptomycin, phosphate-buffered saline (PBS), fetal bovine serum (FBS) and trypsin 5
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 6 of 39
109
were obtained from HyClone/Thermo fisher (Beijing, China). B16F10 cell lines were
110
purchased from Cell Culture Center of Institute of Basic Medical Sciences, Chinese
111
Academy
112
N-hydroxysuccinimide (NHS), Hochest 33342 and Cy5 were purchased from
113
Sigma-Aldrich
114
3-(dibutylamino)-1-propylamine
115
glycol)diacrylate (average Mn 2000, PEGDA2000) were purchased from Aldrich
116
Chemical Corporation. Peptides (>90%) were prepared using standard Fmoc
117
solid-phase peptide synthesis methods and were purified by reverse-phase
118
high-performance liquid chromatography. Cell counting kit assay (CCK-8) was
119
obtained from Beyotime institute of biotechnology (Shanghai, China). 1,
120
6-Hexanediol diacrylate (HDDA), 3-(Dibutylamino)-1-propylamine (DBPA), and
121
dodecylamine were purchased from Aldrich Chemical Corporation. Methyl PEG-NH2
122
2000 was purchased from Seebio Biotech, Inc. C57BL/6 mice (B6, H-2b) (Female, 16
123
± 2 g) were purchased from Department of Laboratory Animal Science, Peking
124
University Health Science Center, and all animal experiments were conducted by its
125
center. All the other solvents used in this research were all of analytic grade and were
126
used without further purification.
127
2.2. Synthesis and characterization of polymer-liposomes
128
2.2.1. Synthesis of functional peptide
of
Medical
(Shang,
Sciences
China).
(Beijing,
1,
China). Irinotecan
6-Hexanediol
(DBPA),
dodecylamine
Hydrochloride,
diacrylate and
(HDDA),
poly(ethylene
129
Peptide (CPLGVRGK-GGG-NYSKPTDRQYHF) was obtained using standard
130
Fmoc solid-phase peptide synthesis methods. Cys-Pro-Leu-Gly-Val-Arg-Gly-Lys 6
ACS Paragon Plus Environment
Page 7 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
131
(SH-CPLGVRGK) was metalloproteinase-2 (MMP-2) substrate sequence and the
132
D-peptide Asn-Tyr-Ser-Lys-Pro-Thr-Asp-Arg-Gln-Tyr-His-Phe (NYSKPTDRQYHF)
133
performed an inhibitor targeting programmed death ligand 1 (PD-L1).
134
2.2.2. Responsive cleavage of functional peptide by MMP-2
135
The enzyme mixture was prepared: (2 mg mL-1) peptide, (15 ng mL-1) MMP-2 in
136
buffer (20 mM Tris∙HCl/50 mM NaCl/100 mM CaCl2/0.05% Brij 35, pH 7.4) with a
137
total volume of 0.5 ml. The reaction was followed at room temperature for 1 h. The
138
original peptide and after exposition for 1 h to gelatinase were analysed by HPLC
139
(acetonitrile (0.1% TFA) /water (0.1% TFA) linear gradient from 5%/95% to
140
60%/40% in 30 min flux 1 mL/min, 220 nm detection).
141
2.2.3. Preparation of pH-responsive polymers
142
The pH-responsive polymers were prepared by Michael addition. HDDA, DBPA
143
and mPEG-NH2 2000 were dissolved into DMSO and then bubbled with N2 for 15
144
min under stirring, then heated at 50 °C for 7 days in dark to obtain P1. Then
145
dodecylamine and P1 were added into DMSO and bubbled with N2 for 15 min under
146
stirring. The mixture was allowed to react for 5 days at 50 °C and dialyzed against
147
deionized water (MWCO: 3,500 Da). 1H NMR spectra (400 MHz) of the copolymers
148
were recorded on a Bruker ARX 400 MHz spectrometer.
149
2.2.4. pH titration of copolymers
150
The graft copolymers (P1) were dissolved in HCl solution with a final
151
concentration of 1.0 mg mL-1. The pH was adjusted to be 10.0 with a 0.1 M NaOH
152
solution at an increment of 10 µl. The NaCl aqueous solution was acted as control 7
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
153
group. The exact pH increases of the solution were monitored with a pH meter at
154
room temperature.
155
2.2.5. Preparation of polymer-liposomes complexes
156
Liposomes (L) were prepared by a thin lipid film hydration method followed by
157
extrusion. Briefly, PC: CH: DOTAP: DSPE-PEG: DSPE-PEG-NHS=62 : 30 : 5.5 : 2 :
158
0.5 (mol ratio) were dissolved in a chloroform/methanol mixture (4 : 1) in a
159
round-bottom flask. The pH-responsive polymers dissolved in methanol were added
160
to engineer polymer-liposomes (LP). CPLGVRGK-GGG-NYSKPTDRQYHF peptide
161
(1mg mL-1) was added to the liposomes (LPp) with gentle stirring for 12 h. DOX was
162
encapsulated in the complexes (LPDp) via an active transmembrane pH gradient
163
method. Size and surface charge of liposomes were measured by using Zetasizer
164
Nano ZS (Malvern Instruments Ltd).
165
2.2.6. Morphology study of polymer-liposomes
166
The morphology of polymer-liposomes obtained by hydration with PBS (pH 7.4) or
167
acetate buffer (pH 5.0) was observed by transmission electron microscopy (TEM,
168
Tecnai G2 F20 U-TWIN). The liposomes sample was stained by uranium acetate for
169
30 s and washed by distilled water twice. The stability of all kinds of
170
polymer-liposomes was investigated by storing at 37 °C for a period of 2 weeks
171
separately. The morphological changes of them were evaluated.
172
2.2.7. Turbidity study
173
The respective amount of pH-responsive polymer (0, 0.5, 1, 1.5 mol%) was added
174
to the lipid mixture before formation of the lipid film. Then the lipid film was hydrated 8
ACS Paragon Plus Environment
Page 8 of 39
Page 9 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
175
with different buffer (pH 7.4, 6.8, 5.5). And the changes of turbidity were determined
176
by OD600 nm using multifunctional microplate reader (Tecan infinite M200).
177
2.3 Hemolysis Assay
178
For hemolysis analysis, LPDp was dispersed in PBS and incubated with the
179
erythrocyte stock dispersions to make the final concentration of LPDp at 0.125 to 2
180
mg/ml. Incubation was carried out at 37 °C for 2 h. Then reaction mixtures were
181
centrifugated at 800rpm for 5 min to remove intact erythrocytes. The supernatants
182
were dissolved in a hydrochloric acid andethanol mixture, following an additional
183
centrifugation (800 rpm for 3 min). PBS solution was used as negative control (0%
184
lysis), and distilled water was used as positive control (100% lysis). The absorbance
185
of the supernatant was determined at 545 nm by an ultraviolet spectrophotometer. The
186
hemolysis rate (HR %) was calculated by Eq. (1): Dt ― Dnc
187
HR%=Dpc ― Dnc × 100
(1)
188
Where Dt, Dnc, and Dpc were the absorbances of the tested sample, the negative
189
control, and the positive control, respectively. The experiments were run in triplicate.
190
2.4. Loading efficiency of DOX
191
DOX was encapsulated in the polymer-liposomes via an active transmembrane pH
192
gradient method as described. The encapsulation efficiency (EE%) and loading
193
capacity (LC%) of DOX was calculated by Eq. (2) and Eq. (3), respectively.
194
EE %=(Drugt Drug f ) Drugt × 100
(2)
195
LC %=(Drugt Drug f ) Lt × 100
(3)
9
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 10 of 39
196
Where Drugt is the total amount of Drug; Drugf is the free amount of Drug in
197
supernate; and Lt is the total weight of polymer-liposomes.
198
2.5. Drug leakage
199
DOX leakage was investigated to evaluate the leakage of drug from
200
polymer-liposomes in the physiological environment at 37 °C for a period of 2 weeks.
201
Various polymer-liposomes (5 ml) were placed into a cellulose membrane dialysis
202
tube (molecular weight cutoff = 2000 Da), respectively. The amount of DOX released
203
in the medium was assayed at 480 nm.
204
2.6. pH-responsive drug release
205
LPDp or LD (5 mL) was placed into a cellulose membrane dialysis tube
206
(molecular weight cutoff = 8000-10,000). The tube was then introduced into 15 mL
207
PBS (pH 7.4), PBS (pH 6.8) or acetate buffer (pH 5.5) and stirred at 100 rpm at 37 °C.
208
At predetermined time intervals (between 0.25 h and 24 h), whole medium was
209
removed and replaced by fresh release medium to maintain a sink conditions. The
210
amount of DOX released in the medium was assayed at 480 nm. All procedures were
211
carried out in the dark to protect DOX from light. The accumulative drug release
212
percentage (Q%) of drug in acetate buffer was calculated by Eq. (4) and all
213
experiments were carried out in triplicate:
214
Q%=(C n V
i
n 1 0
C i Vi ) (Drugt EE %)× 100
(4)
215
Where Cn was the Drug concentration at Tn, V was the total volume of release medium,
216
Vi was the sampling volume at Ti, Ci was the Drug concentration at Ti (both V0 and C0
217
were equal to zero), Drugt was the total amount of Drug used for preparation of the 10
ACS Paragon Plus Environment
Page 11 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
218
original mixture, EE% was the encapsulation efficiency of drug loaded into the
219
liposomal systems.
220
2.7. 50% inhibitory concentration (IC50) of DOX
221
B16F10 cells were cultured in DMEM medium supplemented and were kept in an
222
atmosphere of 5% CO2 and 95% air at 37 °C. Considering the toxicity of DOX (0.05
223
µg mL-1 to 25.6 µg mL-1), the cell viability was investigated by the CCK-8 assay. The
224
percentage of viability was expressed as relative growth rate (RGR %) by Eq. (5):
225
Tests were performed in quadruplicate for each sample.
226
Dt
RGR%=DC × 100%
(5)
227
Where Dt and Dc were the absorbances of the tested sample and the negative control.
228
The IC50 was defined as the concentration of DOX produced 50 per cent relative
229
growth rate.
230
2.8.M6PR expression
231
The effect of DOX on mannose 6-phosphate receptor (M6PR) expression was
232
determined by immunofluorescence. B16F10 cells were incubated with 1 mL of
233
freshly prepared LPDp suspension (DOX concentration from 0.25 µg mL-1 to 1.0 µg
234
mL-1). After 12 h incubation, B16F10 cells were fixed with 4% cold
235
paraformaldehyde for 15 min, washed three times with PBS, blocked with 10% BSA
236
for 1 h at room temperature, and then incubated with primary mouse monoclonal
237
M6PR antibody (1:100, Ab8093; Abcam, Cambridge, MA, USA) overnight at 4 ºC.
238
Cells were washed three times and incubated with FITC-conjugated rabbit anti-mouse
239
secondary antibody (1:1000, zsBio, Beijing, China) for 1h at room temperature. 11
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
240
Nuclei were stained with 4’, 6-diaminoindole (DAPI) (S36938, Invitrogen) for 10 min
241
after washing three times with PBS. Cell membranes were labeled with commercially
242
available membrane tracker (DiI, red color). Cells were observed using a confocal
243
microscope (LSM710, Carl Zeiss, Germany). The cell resuspension was finally
244
subjected to flow cytometry (BD Calibur) and quantitatively analyzed M6PR
245
expression in the different treatment groups with CellQuest software through
246
fluorescence channel 1 (FL1).
247
For western blotting, protein lysates from cells were extracted in TNE lysis buffer
248
(0.5% NP-40, 10 mM Tris, 150 mM NaCl, 1mM EDTA and protease inhibitors).
249
Total protein (50 mg) was separated by 8% gel and transferred to a PVDF membrane
250
and treated with primary mouse monoclonal M6PR antibody (1:100, Ab8093; Abcam,
251
Cambridge, MA, USA), and then immunoblotted with peroxidase-conjugated
252
secondary antibody (1:5000, zsBio, Beijing, China).
253
2.9. Blockage of PD-1/PD-L1 interaction
254
Since the polymer-liposomes relied on MMP-2 to degrade the linker for anti-PD-L1
255
peptide, MMP-2 concentration in the original cell media of B16F10 cells was
256
measured using a MMP-2 ELISA Kit. The Responsive cleavage of functional peptide
257
linker by MMP-2 secreted by B16F10 cells after 24 h culture was evaluated using
258
HPLC. After treatment with IFN-γ (20 ng/mL) to upregulate the expression of PD-L1
259
in vitro, B16F10 cells harvested in a logarithmic growth phase were seeded on
260
glass-bottomed dishes. After 24 h, a commercial Cy3-labeled recombinant mouse
261
PD1 protein (ab216248) was added to the cells. Meanwhile, the culture medium was 12
ACS Paragon Plus Environment
Page 12 of 39
Page 13 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
262
replaced by serum-free medium containing LPp, LP, or D-peptide. After incubation
263
at 37 °C for 4 h, the cells were washed three times with PBS and stained with 10 μg
264
mL−1 Hoechst 33342 for 10 min, then viewed by confocal laser scanning microscope.
265
To test the blocking efficiency, we conducted flow cytometry experiments.
266
2.10. Apoptosis analysis
267
The apoptosis of B16F10 cells was further determined by fluorescence-activated
268
cell sorting (FACS) using annexin V-FITC/propidium iodide (PI) double staining
269
after treatment with PBS, GranB, LPDp, or LPDp + GranB for 24 h. Briefly, the
270
treated cells were trypsinized and collected by centrifugation for 5 min. Then the cells
271
were washed and resuspended with 100 μL of PBS. Finally, the cells were first
272
stained with annexin V-FITC (25 μg/mL) for 15 min on ice and then incubated with
273
propidium iodide (50 μg/mL) for 5 min before FACS analysis.
274
2.11. In vivo imaging
275
All animal experiments were performed complying with the NIH guidelines for the
276
care and use of laboratory animals and according to the protocol approved by the
277
Institutional Animal Care. 5×106/mL-1 B16F10 cells, collected in serum free DMEM
278
medium (200 μL), were subcutaneously (s. c.) injected into the right lateral hind hip
279
of the female C57BL/6 mice. The tumor volume was calculated by Eq (6): Tests were
280
performed in quadruplicate for each sample.
281 282 283
V = AB2/2
(6)
Where A was the maximum diameter, B was the minimum diameter. When tumors had developed to approximate 150 - 200 mm3, the mice were 13
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 14 of 39
284
intravenously injected Cy5 labeled LP or LPp (200 μL 1mg mL-1). Near-infrared
285
imaging was carried out after 4 h, 8 h, and 24 h using a Maestro in vivo spectrum
286
imaging system (excitation filter, 649 nm; emission filter, 670 nm). Mice treated with
287
intravascular injection of PBS were acted as control group. Then animals were
288
sacrificed and tumor, heart, liver, spleen, lung and kidney were excised for flourecsent
289
intensity measurement.
290
2.12. In vivo combined therapeutic study
291
2.12.1. Tumor treatment
292
When the tumors reached sizes of 50 –100 mm3, at which time the mice (in groups
293
of six each) received different treatments: (a)LPDp, (b)LPp, (c)LD (1 mg/kg in
294
DOX), (d)free DOX (10 mg/kg in DOX), or (e) PBS. The drugs were intravenously
295
administered to mice on every second day (day 1, 3, 5, 7, 9, 11 and 13). DOX
296
concentration in normal tissues was measured after 24 h injection using HPLC.
297
During the process of the treatment, the tumor volumes and body weight were
298
measured every two days. After in vivo anti-tumor treatment, mice were sacrificed
299
and blood, tumor, heart, liver, spleen, lung, kidney were collected for blood
300
biochemistry and histological analysis. The tumor inhibition ratio (IR%) was
301
calculated by Eq. (7): Tests were performed in quadruplicate for each sample.
302
IR%= (Vc Vt ) Vc 100
(7)
303
Where the Vc was the average tumor volume of control group after treated with
304
intravascular injection of PBS for 17 d, and the Vt was the average tumor volumes of
305
treatments with LPDp, LPp, LD, or free DOX, respectively. 14
ACS Paragon Plus Environment
Page 15 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
306
Biomacromolecules
2.12.2. Immunohistochemical staining
307
M6PR expression in tumor tissues was detected by Immunohistochemistry. Tumors
308
were fixed in 4% paraformaldehyde for 4 h at 4 °C and cut into 6 μm sections for
309
histological analysis. Briefly, sections were deparaffinized, hydrated, and subjected to
310
proteinase K treatment for antigen retrieval. Endogenous peroxidase was blocked by
311
3% hydrogen peroxide/methanol. The slides were blocked with normal goat serum
312
and then incubated with corresponding primary antibody (1:50, Abcam, USA)
313
overnight at 4 °C, respectively. After washing with PBS, the sections were incubated
314
with HRP-conjugated Goat Anti-mouse IgG (1:500, Abcam, USA) and DAB was
315
used for color development. The obtained experiment slices were observed by optical
316
microscopy. All measurements were carried out using Image J.
317
2.12.3. The intratumoral CD8+ T cells
318
The tumor tissues were harvested and the cells separated from tumor tissues were
319
stained with PE-CD8a antibody for 20 min on ice at dark. The cells were washed 3
320
times with PBS buffer, centrifuged and re-suspended in PBS buffer. Cell resuspension
321
was finally subjected to flow cytometry (BD Calibur) and analyzed with CellQuest
322
software.
323
The frozen tumor tissue sections of 10 mm thickness were prepared. CD8 positive
324
cells in tumor sections were stained using anti-rat CD8 antibody (1:500 dilution,
325
eBioscience) and Cy3-anti-rat IgG (1:500 dilution, Jackson immune research) as a
326
secondary antibody (Red). Finally, the sections were washed three times with PBS
327
and the nuclei were labeled with DAPI. The apoptosis in tumors was detected by 15
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
328
the TUNEL assay. Sections were then washed, covered with coverslip and observed
329
using a LSM710 confocal microscope.
330
3. Results and discussions
331
3.1. Characterization of polymer-liposomes complexes
332
The pH-sensitive copolymer P1 was synthesized by Michael addition of
333
hydrophobic monomer HDDA, pH-sensitive monomer DBPA and hydrophilic
334
PEG-NH2 (Supporting Information, Figure S1). For effective insertion into
335
membrane of liposome, dodecylamine was conjugated onto the both ends of P1,
336
gaining copolymer P2. The 1H NMR spectrum of P1 and P2 was shown in
337
Figure S2 and Figure S3, respectively (Supporting Information). The pH
338
sensitivity of P2 was evaluated by a pH titration method. After adding 0.1 M
339
NaOH, a high buffer capacity of copolymers was observed in the range of pH
340
4.0–9.0 compared to NaCl solution, which was in accordance with other
341
copolymers with diethyl amino groups (Figure S4, Supporting Information).
342
PC: CH: DSPE-PEG: DOTAP: DSPE-PEG-MAL= 62: 30: 2: 5.5: 0.5(mol
343
ratio) were dissolved in a chloroform/methanol mixture (4: 1) to prepare
344
liposomes (L)
345
mol%) dissolved in methanol was added to the lipid mixture before formation
346
of the lipid film. The lipid film was hydrated with phosphate buffered saline
347
(PBS, pH 7.4) following evaporating the organic solvents. DOX was
348
encapsulated in LP using the active transmembrane pH gradient method
349
(LPD).The peptide (p) (CPLGVRGK-GGG-NYSKPTDRQYHF) was coupled
37, 38.
For polymer-liposomes (LP) preparetion, P2 (0–2.5
16
ACS Paragon Plus Environment
Page 16 of 39
Page 17 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
350
to
the
maleimide
group
of
DSPE-PEG2000-MAL
351
Responsiveness and selective cleavage site of the linker p in the presence of
352
MMP-2 were validated by HPLC (Figures S5, Supporting Information). DOX
353
was encapsulated in L to prepare LD. Schematic representation of the
354
component details can be found in the Supporting Information (Figure S6).
355
Transmission electron microscope (TEM) images of LP with the respective
356
amount of P2 (0–2.5 mol%) (FigureS7, Supporting Information). The
357
self-aggregations of P2 were detected visually with the amount of P2 up to 2
358
mol% and 2.5 mol%. P2 with the amount ≤ 1.5mol% was able to be entirely
359
incorporated into liposomes. The LP containing 1.5 mol% of P2 exhibited
360
hollow and spherical structures in PBS buffer solution (pH 7.4) (Figure1A).
361
The hydrodynamic diameter of all liposomal systems was around 60-70 nm
362
with PDI ≤ 0.2 and zate potential about 20 mV (Table S1, Supporting
363
Information).The LPp with 1.5 mol% of P2 was re-dissolved in acetate buffer
364
(pH 5.0) and incubated for 30 min. The bilayer membrane of LPp was
365
disrupted, which was clearly observed by TEM. The morphological change of
366
L (containing 0 mol% P2) was unobvious in the same condition. The turbidity
367
of LPp with 0–1.5 mol% of P2 was checked to evaluate the integrality of
368
liposome membrane by monitoring the optical density at 600 nm (OD600
369
(Figure1B). At pH 6.8, the OD600 nm of all liposomal systems was detected with
370
almost no change in comparison with that at pH 7.4. The turbidity of LPp with
371
1.5 mol% P2 at pH 5.5 reduced to 44.1% compared to that at pH 7.4. For L 17
ACS Paragon Plus Environment
to
form
LPDp.
nm)
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
372
without mixing of P2, that acidic condition caused no significant change of the
373
OD600
374
and tumor microenvironment and release the loaded cargos at the lysosome (pH
375
5–6), implying that the LPDp could be applied as pH-responsive drug carriers
376
for intracellular delivery.
nm.
Therefore, the LPDp were almost stable in physiological condition
377
Hemolysis rates of LPDp suspension are shown in Figure S8 (Supporting
378
Information). The HR % of LPDp at the highest concentration (2 mg/ml) was
379
below 3.5%, indicating that LPDp had good hemocompatibility.
380
3.2. Loading and release of DOX
381
The introduction of the P2 into polymer-liposomes
382
The encapsulation efficiencies of the DOX in polymer-liposomes were >60%, at
383
loading capacities of approximately 8–10% (Supporting Information, Table S1). The
384
DOX leakage from the delivery systems was quantitatively determined after storing
385
for 2 weeks at 37 °C. The amount of DOX leakage from the LD was found to be
386
about 18.7%. In contrast, only about 3.6% leakage was obtained from the LPD and
387
LPDp under the same conditions. These results indicate that the pH-responsive
388
polymers helped to stabilize the liposomes and reduce leakage of the payloads in the
389
physiological environment. Furthermore, morphological change after the storage of 2
390
weeks at 37 °C was observed using TEM to monitor the physical stability of the
391
particles. LD showed obvious fusion and severe aggregation (Figure 1C). For LPD
392
and LPDp in the identical conditions, only a slight aggregation could be detected.
393
The drug release profiles were monitored at different pH (7.4, 6.8, 5.0), and the 18
ACS Paragon Plus Environment
Page 18 of 39
Page 19 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
394
amount of released DOX was quantified by HPLC. At pH 7.4 and pH 6.8, only 5%
395
and 10% of the loaded DOX was released from LD, respectively. In acetate buffer
396
(pH 5.0), higher solubility of DOX caused more drug release (about 34% for 24 h).
397
When LPDp was incubated at pH 5.0, the release of DOX was significantly
398
accelerated (>65%), which was attributed to the dissociation of the liposomes. The
399
results demonstrated that the pH-responsive drug release was in accordance with the
400
turbidity assay.
401
The pH-responsive polymer chains incorporated in LPD and LPDp contained
402
anchoring moiety for tight attachment onto the liposome membrane surface and made
403
LPD and LPDp more stable to reduce leakage of the payloads in physiological
404
environment. At acidic pH, the pH-sensitive moiety of the pH-responsive polymer
405
chains was capable of converting to protonated state accompanying the dynamic
406
disordering of membrane. Therefore, the pH-sensitive liposomes could reduce leakage
407
of DOX and protected the inner liposomal core in the physiological environment prior
408
to arrival at the targeted cells.
409
3.3. Up-regulation of the M6PR induced by low-dose DOX
410
We optimized the dosage of DOX by assessing the efficacy of up-regulating M6PR
411
on B16F10 cells. The IC50 of DOX was about 2.8 µg mL − 1(Figure S9, Supporting
412
Information) by evaluating cell viability of B16F10 cells and optimized the low dose
413
of DOX (< 2 µg mL-1) for further experiments with negligible systemic side effects.
414
M6PR on the surface of the B16F10 cells incubation with LPDp (DOX concentration
415
from 0.25 µg mL − 1 to 1 µg mL − 1) was comfirmed to be up-regulated obviously by 19
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 20 of 39
416
immunofluorescence (Figure 2A). The expression of the M6PR was verified
417
quantitatively by flow cytometry. The M6PR expression improved with the DOX
418
concentration increase (Figure 2B). Compared with control group, about 6-fold higher
419
(P < 0.01) mean fluorescence intensity (32.12± 3.21) was detected in B16F10 cells
420
treated with LPDp with DOX concentration of 1 µg mL-1 (Figure S10, Supporting
421
Information). The expression of the M6PR was also verified with respect to protein
422
level (Figure 2C) by Western blotting. These results were consistent with reports that
423
DOX could induce overexpression of M6PR on tumor cell membranes.
424
3.4. Blockade of PD-1/PD-L1 interaction in vitro
425
Matrix metalloproteinases (MMPs), up-regulated in tumor regions
39,
have been 40.
426
extensively studied as target enzymes for drug-delivery systems design
427
concentration MMP-2 in B16F10 cell media was measured using ELISA Kit. As
428
demonstrated in Figure S11 (Supporting Information), MMP-2 was overexpressed in
429
B16F10 cells media (about 0.76 μgmL−1) after 12 h culture, which was about 50-fold
430
higher than the concentration we used to cleave the linker of anti-PD-L1 peptide in
431
section 2.2.2. When the incubation time is extended to 24 h, the MMP-2 concentration
432
was up to about 2-fold higher (P < 0.05) in cell culture supernatants. We confirmed
433
that amount of MMP-2 secreted by B16F10 cells in vitro was sufficient to release the
434
PD-L1 antagonist using HPLC (Figure S12, Supporting Information). As PD-L1 was
435
low-expressed during in vitro culturing
436
upregulated the PD-L1 on B16F10 cells and confirmed by confocal laser scanning
437
microscope and flow cytometry experiment (Figure 3A and Figure S13, Supporting
41, 42,
The
we used interferon-γ (IFN-γ) to
20
ACS Paragon Plus Environment
Page 21 of 39 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Biomacromolecules
438
Information). In vitro PD-1/PD-L1 blockage experiments were conducted by treating
439
B16F10 cells with PBS, LP, LPp, or D-pep (0.1 mg mL−1 in peptide) in the presence
440
of Cy3-labeled recombinant mouse PD1 protein for 4 h at 37 °C. In comparison with
441
control group, LPp and D-pep exhibited high inhibitory effect with significant
442
fluorescence intensity decrease (p < 0.01) as revealed in CLSM image (Figure 3B).
443
On the contrary, negligible blockage was detected when B16F10 cells treated with LP.
444
The blocking rate of PD-1/PD-L1 interaction was also assessed quantitatively by flow
445
cytometry (Figure 3C). It could be seen that LPp blocked PD1/PD-L1 interaction
446
more significantly (Plung>spleen>heart
499
concentration of DOX in liver (0.08 μg/ml) was about approximately 4-fold higher (P
500
< 0.05) than that in heart. The toxicity of the LPDp in vivo was further investigated
501
by blood and histological assays (Table S2 and Figure S18, Supporting Information).
5A),
was
indicating
evaluated
nontoxicity
by
of
using
the
HPLC,
(Supporting
low-dose
indicated
Information,
DOX
the Figure
encapsulated
order S17).
of The
502
The platelet counts, white blood cell and red blood cell of mice in LPDp treated
503
group showed no significant changes compared to that in control group. Moreover, 23
ACS Paragon Plus Environment
Biomacromolecules 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 47 48 49 50 51 52 53 54 55 56 57 58 59 60
504
the levels of ALP, CR, BUN, ALT and AST in LPDp treated group were within the
505
normal ranges, indicating low toxicity to liver and kidney. No noticeable organ
506
damage was detected after pathological assay, which indicated low dose DOX showed
507
low toxicity to normal tissues.
508
The M6PR had been shown to play a role in endocytic uptake of GranB46, since
509
target cells overexpressing M6PR had an increased sensitivity to the cytotoxic effect
510
of CTLs47,
511
Immunohistochemistry (Figure 5B). The M6PR expression levels in the LPDp
512
treatment group were the highest (0.015), which demonstrated a far more significant
513
up-regulation (P < 0.01) than that of LPp and a slightly higher one compared to that
514
of LD (Figure S19, Supporting Information). The cell apoptosis in tumor tissue was
515
measured by TUNEL assay (Figure 5C). The fluorescent intensity in tumor tissue
516
sections indicated the order of LPDp>LPp >LD >PBS. Compared with LPp, about
517
2-fold higher (P