Integrated Hydrogel Platform for Programmed Anti-Tumor Therapy

17 hours ago - Complete tumor regression is a great challenge faced by single therapy of near infrared (NIR)-triggered hyperthermia or vascular disrup...
1 downloads 0 Views 1MB Size
Subscriber access provided by BUFFALO STATE

Biological and Medical Applications of Materials and Interfaces

Integrated Hydrogel Platform for Programmed Anti-Tumor Therapy Based on Near Infrared-Triggered Hyperthermia and Vascular Disruption Yuqing Liang, Yijun Hao, Yingjiao Wu, Zhijun Zhou, Juan Li, Xiaoyi Sun, and You-Nian Liu ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.9b05536 • Publication Date (Web): 29 May 2019 Downloaded from http://pubs.acs.org on May 30, 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 32 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

ACS Applied Materials & Interfaces

Integrated Hydrogel Platform for Programmed AntiTumor Therapy Based on Near Infrared-Triggered Hyperthermia and Vascular Disruption Yuqing Liang,† Yijun Hao,† Yingjiao Wu,† Zhijun Zhou,‡ Juan Li,*,† Xiaoyi Sun,† and You-Nian Liu† †

College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083,

P.R. China ‡

Department of Laboratory Animals, Central South University, Changsha, 410083, P.R. China

KEY WORDS: Prussian blue, injectable hydrogel, vascular disrupting agent, combinational therapy, gellan

ABSTRACT: Complete tumor regression is a great challenge faced by single therapy of near infrared

(NIR)-triggered

hyperthermia

or

vascular

disrupting

agents.

An

injectable

nanocomposite (NC) hydrogel is rationally designed for combined anti-cancer therapy based on NIR-triggered hyperthermia and vascular disruption. The NC hydrogel, co-delivered with Prussian blue (PB) nanoparticles and combretastatin A4 (CA4), has good shear-thinning, selfrecovery and excellent photothermal property. Due to the remarkable tumor-site retention and sustained release of CA4 (about 10% over 12 days), the NC hydrogel has a tumor suppression

ACS Paragon Plus Environment

1

ACS Applied Materials & Interfaces 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 2 of 32

rate of 99.6%. The programmed combinational therapy conveys a concept of “attack + guard”, where PB-based NIR irradiation impose intensive attack on most of cancer cells, and CA4 serves as guard against the tumor growth by cutting off the energy supply. Moreover, the biosafety and eco-friendliness of the hydrogel platform pave the way towards clinical application.

1. INTRODUCTION Recent advances in new anti-cancer agents or strategies, such as photothermal therapy (PTT),1-11 vascular disrupting agents (VDAs),12-15 reactive oxygen species (ROS) therapy16 and immune checkpoint blockade17-19 have brought great opportunities to cancer therapy. However, the outcome of single therapy is far from satisfactory due to the challenges such as tumor heterogeneity, systemic toxicity and drug resistance.20 For instance, near-infrared (NIR)triggered PTT is a non-invasive strategy to ablate tumors, and shows advantages of minimal toxicity and precise local treatment.21 The mechanism of PTT on cell death involves necrosis and/or apoptosis.22 However, PTT alone imposes moderate effect without long-term tumor remission because of the limited penetration depth of NIR laser and upregulation of heat shock protein.20,

23

It is found that the tumor regrew three days after PTT therapy alone.24 Another

example of VDAs could selectively act on the endothelial cells in abnormal tumor vessels, and induce cutoff of tumor blood flow, leading to secondary tumor death as a result of the deprivation of oxygen and nutrients.25-27 Among the existing VDAs, combretastatin A4 (CA4) and its derivatives are the leading agents under clinical evaluation.21, 27 However, the short halflife and side effects of CA4 and prodrug CA4P in normal tissues are still observed.21 Moreover, the blockade of energy supply can only slow down tumor growth without complete tumor ablation.21, 28

ACS Paragon Plus Environment

2

Page 3 of 32 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

ACS Applied Materials & Interfaces

By optimal biomaterial engineering, an integrated delivery system is able to deliver multiple cargoes and control the release behavior, which may overcome the current challenges by maximizing the efficacy while minimizing the side effects. Though various nanoparticles (such as iron oxide nanoparticle, gold nanoparticles, and poly(β-aminoester) nanoparticles) themselves can be employed as drug carriers for synergistic therapy, they still suffer the rapid clearance and low retention efficiency in the body for short periods (less than one week) due to their small size and/or hydrophilic surface.29-31 Thus, it is hard for these nanoparticles to maintain a sustained release of chemotherapeutic drugs and to allow sufficient therapeutic drug concentrations for prolonged period. Injectable hydrogel is very intriguing for combinational therapy, showing advantages of facile incorporation of multiple cargos, huge loading capacity, minimally-invasive administration and sustained release behavior.32-37 A variety of injectable hydrogels have been constructed for combinational anti-tumor therapy, including chemo-PTT,11 chemo-VDA,14 and PTT-PDT.38 For example, injectable hydrogels based on thermosensitive polypeptide have been applied for chemo-VDA therapy with sequential drug release. The final tumors are greatly suppressed, though not fully diminished in the chemo-VDA therapy.13-14 Recently, our lab discovered that gellan can be applied for in-situ synthesis of small CuS nanoparticles with high photothermal conversion efficiency, and anti-cancer hydrophilic cationic drug (DOX) was encapsulated for synergistic chemo-PTT therapy.11 The gellan hydrogel endows a sustained release behavior via the electrostatic interaction between gellan and DOX. In particular, PTT can enhance the tumor perfusion and improve the delivery of chemotherapeutic drugs.39 On the other hand, PTT may bring some antihypoxic effect and cause blood vessel injury.40-41 Hence, it is of interest to find out whether PTT can also cooperate with the VDA effectively. Surprisingly, most studies focused on other combinational therapies, including chemo-VDA,28 photodynamic-

ACS Paragon Plus Environment

3

ACS Applied Materials & Interfaces 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 32

VDA42 and radio-VDA,43 the combination of PTT with VDA, especially integrated into hydrogel platform, is rarely explored.

Scheme 1. Illustration of injectable NC hydrogel for co-delivery of CA4 and PB in synergistic photothermal and vascular disrupting therapy. Herein, an injectable nanocomposite (NC) hydrogel for anti-tumor combinational therapy is rationally designed based on NIR-triggered hyperthermia and vascular disruption (Scheme 1). Prussian blue (PB), approved by US Food and Drug Administration (FDA) for clinical application of detoxification for radioactive metals, is selected as a PTT nanoagent due to its excellent NIR absorption property.5, 44-49 As a proof of concept, this platform provides an antitumor strategy of “attack + guard”, where PB-based NIR irradiation imposes intense attack on most of cancer cells, and CA4 serves as guard against the tumor growth by cutting off the energy supply. The enhanced photothermal efficiency of PB in gellan and disruption of tumor vascular with prolonged released CA4 ensure the therapeutic effect synergistically. Moreover, the NC

ACS Paragon Plus Environment

4

Page 5 of 32 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

ACS Applied Materials & Interfaces

hydrogel could be exploited as a universal drug carrier for local delivery of multiple cargoes with sustained release behavior by single injection. 2. EXPERIMENTAL SECTION 2.1. Materials. All the agents applied in this work were described in the Supporting Information. 2.2. Preparation of PB nanoparticles. PB nanoparticles were prepared according to a reported method. 50 In a typical procedure, PVP (3.0 g) and K3[Fe(CN)6]·3H2O (131.7 mg) were added to HCl solution (0.01 M, 40 mL) under magnetic stirring. After 30 min of stirring, a clear solution was obtained. The sample was then placed into an oil bath and heated at 85 oC for 20 h. After aging, the mixture was then centrifuged at 10000 rpm for 20 min to collect PB nanoparticles, and the precipitate was washed with distilled water until the supernatant was neutral. Finally, the obtained PB nanoparticles were lyophilized and dispersed in deionized water for future use. 2.3. Preparation of PB@gellan and NC hydrogel. PB nanoparticles was dispersed in water (0.1 mg mL-1) under sonication, and gellan (2 wt%) was dissolved in PB dispersion at 85 oC to produce the pre-gel solution. After cooling down to room temperature, PB@gellan hydrogel was obtained. For CA4 loading, PB@gellan pre-gel was placed in 50 oC water bath for 1 hour, after that, CA4 powder was added to PB@gellan pre-gel under vortex mixing (the final concentration of CA4 is about 6.0 mg mL-1). Until CA4 was completely dispersed in the mixed solution, the mixture was cooled down to room temperature and the NC hydrogel was obtained. 2.4. Characterization of PB nanoparticles and PB@gellan hydrogel. The phase and crystallography of PB nanoparticles were characterized by using an X-ray diffractometer

ACS Paragon Plus Environment

5

ACS Applied Materials & Interfaces 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 32

(Axios mAX), with a scanning rate of 10o s-1 applied to record the pattern in the 2θ range of 10 ~ 80o. The morphology of PB nanoparticles was examined by TEM (JEM-2100F) and SEM (TESCAN MIRA3 LMU). The surface charge and dynamic light scattering of PB was measured on a Malvern Zetasizer Nano-ZS Instrument.

The UV-Vis-NIR

spectrum was obtained with a Shimadzu UV-2450 spectrophotometer. The morphology of gellan, PB@gellan and NC hydrogel was uncovered by SEM (TESCAN MIRA3 LMU). The Raman spectroscopy of the lyophilized hydrogels was measured on a Raman microscope (Renishaw, INVIA) with 532 nm laser source in the range of 1800−2400 cm−1. 2.5. Photothermal effects of PB@gellan. PB@gellan hydrogels (1.0 mL), with different concentration of PB nanoparticles, were added into vials, and irradiated with a fibercoupled continuous semiconductor diode laser (808 nm, Beijing Viasho Technology Co., Ltd., China) for 5 min, the temperature was monitored with an infrared thermal imaging camera (Flir C2, USA). The power density is 1.0 W cm-2. The photothermal stability of PB@gellan was investigated as well. PB@gellan (0.1 mg mL-1 of PB) was irradiated with 808 nm laser for 3 min and then cooled down to room temperature for five cycles. The photothermal conversion efficiency was calculated in the Supporting Information according to a reported literature,51 and PB nanoparticles dispersed in water were set as control. 2.6. Rheology test of NC hydrogel. Rheological measurements were performed on a rheometer (Anton Paar, MCR 302) with parallel plate geometry (25 mm diameter). The gap between the probe and the plate was maintained at 1.0 mm. The samples were incubated at 60 oC water bath to keep the sol state of the hydrogel, and then the samples

ACS Paragon Plus Environment

6

Page 7 of 32 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

ACS Applied Materials & Interfaces

(1 mL) were added onto the sample platform and equilibrated at the testing temperature for 30 min before test (except the temperature sweeps). The parallel plate was carefully sealed with silicon oil in case of water evaporation during measurement. Unless otherwise stated, all tests were performed at 37 oC with a strain of 0.1%, and the frequency was set at 1.0 Hz. Strain sweep test of NC hydrogel was conducted within a strain amplitude sweep (0.01 ~ 400%) at 37 oC to uncover the linear viscoelastic region. Shear-thinning property of the NC hydrogel was characterized at 37 oC as well. The viscosity of the sample was measured within a wide range of shear rate (0.01 ~ 10 s-1), and also in the recovery cycle with the shear rate decreased from 10 s-1 to 0.01 s-1. Self-recovery behavior was evaluated by continuous change of strains between 0.1% and 300% for 5 cycles at 37 oC. The sample was damaged under a strain of 300% for 30 s, and then a strain of 0.1% was applied to inspect the recovery of NC hydrogel for 3 min. A temperature sweep was carried out in a temperature range of 25 ~ 55 oC with a temperature variation of 1.0 oC min-1. 2.7. Drug release of CA4 in NC hydrogel. In vitro drug release was carried out according to a previously reported method.14,

52

After CA4 was incorporated into PB@gellan

mixture at 50 oC, NC hydrogel pre-gel solution was added to a vial with the inner diameter of 15 mm, the vial was stored at 4 oC refrigerator overnight. Afterwards, about 0.5 g NC hydrogel was added to PBS (40.0 mL) at pH 7.4. PBS with 0.5% SDS was also used as the release medium to increase the solubility of CA4. The release studies were carried out at 37 oC water bath with continuous shaking at 70 rpm. The release medium (1.0 mL) was collected and replenished with equivalent fresh buffer every other day. The

ACS Paragon Plus Environment

7

ACS Applied Materials & Interfaces 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 8 of 32

concentration of CA4 in the collected medium was quantified by RP-HPLC method.53 The mobile phase consisted of methanol and water (2:1, v/v), and the flow rate was 0.8 mL min-1, the absorbance was detected at 295 nm, and the retention time of CA4 was about 9 min. 2.8. In vitro photothermal study. Cancerous 4T1 cells were seeded into 6 well plate (1 × 106 cells per well) and cultured at 37 oC for 24 h. After attachment, the medium was removed and replaced by fresh culture medium, PB@gellan (20 L) was added to each well. After incubation for 4h, cells were irradiated with 808 nm laser (1.0 W cm-2) for 3 min. After another 30 min incubation, the culture medium and the residual hydrogel were removed and fresh medium containing calcein-AM (5 g mL-1) and PI (10 g mL-1) was added. After cultured for another 10 min, the cells were washed with PBS for three times and imaged on an inverted fluorescence microscope. The invitro photothermal effect of NC hydrogel was investigated according to the same procedures. 2.9. In vivo retention of NC hydrogel by fluorescent imaging. The local retention of NC hydrogel was tracked through fluorescence imaging. Cy5, a hydrophobic molecule, was selected as a fluorescence signal molecule because CA4 has no fluorescence behavior.54 4T1 cells in logarithm growth period were collected and subcutaneously injected into the armpits of nude mice (6 weeks old), after two weeks, the tumor volume reached about 300 mm3 (tumor volume = 1/2 × length × width2), the mice were divided into three groups randomly. The treatment includes: free Cy5 in PBS, Cy5 in PB@gellan without irradiation, Cy5 in PB@gellan with 808 nm laser irradiation. Each mouse was intratumorally injected with 50 μL samples; the irradiation was carried out with 808 nm laser (1.0 W cm-2) for 3 min. The concentration of Cy5 was set as 6 mg mL-1. The

ACS Paragon Plus Environment

8

Page 9 of 32 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

ACS Applied Materials & Interfaces

fluorescent signal was tracked for 12 days. The experiment was conducted under protocols approved by Servicebio Laboratory (Wuhan, China). 2.10. In vivo photothermal stability. Female BALB/c mice (5 ~ 6 weeks old) were used under protocols approved by Department of Laboratory Animals, Central South University (Changsha, China). 4T1 cells in logarithm growth period were collected and subcutaneously injected into the back of the mice. After one week, the tumor volume reached about 150 mm3. Tumor-bearing mice were randomly divided into two groups (n = 3) for the following treatment: free PB nanoparticles with 808 nm laser irradiation, and PB@gellan with 808 nm laser irradiation. Each mouse was intratumorally injected with 100 μL samples, and the irradiation was carried out with 808 nm laser (1.0 W cm-2) for 3 min over three consecutive days. The temperature evolution during the irradiation was monitored by infrared thermal imaging camera, and the thermal images of mice were captured as well. 2.11. In vivo anti-tumor synergistic therapy. Female BALB/c mice (5 ~ 6 weeks old) were used under protocols approved by Department of Laboratory Animals, Central South University (Changsha, China). 4T1 cells in logarithm growth period were collected and subcutaneously injected into the back of the mice. After one week, the tumor volume reached about 150 mm3. Tumor-bearing mice were weighted and randomly divided into six groups (n = 5). The treatment includes: PBS as control (group 1, G1), CA4@gellan (group 2, G2), free CA4 (group 3, G3), PB@gellan with 808 nm laser irradiation (group 4, G4), free PB nanoparticles in PBS with 808 nm laser irradiation (group 5, G5), NC hydrogel with 808 nm laser irradiation (group 6, G6). Samples (100 L) were injected intratumorally only once, with the dosage of 30.0 mg kg-1 for CA4, 0.5 mg kg-1 for PB

ACS Paragon Plus Environment

9

ACS Applied Materials & Interfaces 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 32

nanoparticles. Irradiation (808 nm laser, 1.0 W cm-2, 3 min) was performed at one hour after injection, and infrared thermal imaging camera was used to monitor the local temperature during irradiation. The tumor volume and body weight of mice were measured every other day. The tumor suppression rate (TSR) was obtained according to the following formula: TSR (%) = (Vc – Vx) / Vc × 100%, where Vc represents the tumor volume of control group, and Vx represents that of treatment group.12 After 22 days, the mice were sacrificed and the major organs as well as tumors were harvested for H&E staining, the tumor tissues were cut into sections for immunohistochemical staining as well. 2.12. H&E and immunohistochemical staining. Major organs harvested from the sacrificed mice (after 22 days) were fixed in 10% PBS-buffered formalin, then all the tissues were embedded into paraffin and sliced with 8-micron thickness, stained with hematoxylin and eosin (H&E), and imaged on an inverted fluorescence microscope for histopathological analyses. Immunohistochemical staining was conducted according to a reported immunocytochemistry protocol.14 The sliced tumor tissue was firstly dewaxed and washed with PBS for three times, tumor slices were stained with CD31 antibody for microvessel and DAPI for the nuclei of tumor cells. 2.13. Statistical analysis. All data in this article were presented as mean result ± standard deviation, and the statistical analysis were performed via Origin 8.0 software. The data of antitumor in vivo were analyzed using one-way ANOVA, and asterisks were applied to indicate the significant differences (*p < 0.05, **p < 0.01, ***p < 0.001). 3. RESULTS AND DISCUSSION

ACS Paragon Plus Environment

10

Page 11 of 32 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

ACS Applied Materials & Interfaces

3.1. Characterization of PB nanoparticles and PB@gellan hydrogel. PB nanoparticles were

synthesized

through

a

reduction

method.50

In

the

presence

of

PVP,

K3[Fe(CN)6]·3H2O turns into solid mesocrystals of PB nanoparticles. The prepared PB nanoparticles are monodispersed with a cubic shape and have an average size of ~100 nm from TEM and SEM observations (Figure 1A and B). The average hydrodynamic diameter and zeta potential of PB nanoparticles is 124 nm and −24.6 mV, respectively (Figure S1, Supporting Information). Diffraction peaks at 17.44, 24.74, 35.28 and 39.62 o, corresponding to the 200, 220, 400 and 420 planes, demonstrate the high crystallinity of PB (Figure 1C).

Figure 1. A) TEM, B) SEM and C) XRD patterns of PB nanoparticles. D) SEM image of PB@gellan hydrogel.

ACS Paragon Plus Environment

11

ACS Applied Materials & Interfaces 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 12 of 32

By simply adding gellan powder into PB nanoparticle dispersion at elevated temperature (above 60 oC), the PB@gellan hydrogel was obtained after cooling at room temperature. Owing to the negative surface charge of PB nanoparticles (zeta-potential: −34 mV), they could be well dispersed in the anionic gellan hydrogels. More importantly, PB nanoparticles are stable in gellan without sedimentation even after two months (Figure S2, Supporting Information). By contrast, the pristine PB nanoparticles in water settle to the bottom of the vial after one week. PB@gellan hydrogel preserves a porous structure with an average pore size of 0.97 μm (Figure 1D), and PB nanoparticles themselves are fully embedded in hydrogel matrix in the SEM image owing to the low content of PB in the sample (0.1 mg PB to 20 mg gellan per mL). Nevertheless, HR-TEM image of PB@gellan shows that PB nanoparticle preserves its crystal structure in the gellan matrix (Figure S3, Supporting Information). The measured lattice spacing is about 0.51 nm, representing the plane 200 of PB. In addition, the Raman characteristic bands of PB shift towards a lower frequency (2156 to 2155 cm-1, and 2092 to 2087 cm-1) after encapsulation into gellan matrix,55 indicating the interaction between PB and gellan (Figure S4, Supporting Information). 3.2. Optical and photothermal property of PB@gellan hydrogel. The optical spectrum of PB@gellan exhibits a broad absorption of 500-900 nm with a centered peak at 710 nm (Figure 2A), in accordance with that of pristine PB nanoparticles. The photothermal effect of PB@gellan was investigated under exposure to 808 nm laser irradiation (1.0 W cm-2). A fast increment of temperature is observed for PB@gellan under irradiation, while gellan itself showed slight change (Figure 2B and D). In addition, it turns out that the temperature enhancement also depends on the concentration of PB. And the concentration of PB

ACS Paragon Plus Environment

12

Page 13 of 32 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

ACS Applied Materials & Interfaces

nanoparticles was settled at 0.1 mg mL-1 in the following experiment, which is sufficient to reach the critical temperature to kill cancer cells, in accordance with the in vitro photothermal therapy of PB (Figure S5, Supporting Information). PB@gellan has better photothermal effect than that of pristine PB nanoparticles in water (photothermal conversion efficiency 54.7% vs 44.3%, Figure S6 and S7, Supporting Information) and PB in other literatures.56 Furthermore, the stable photothermal effect of PB@gellan suggests that single injection of PB@gellan can exert multiple photothermal therapy to induce tumor ablation (Figure 2C).

Figure 2. A) Absorbance spectra of PB in different media (Inset was photos of samples). B) Photothermal heating curves of PB@gellan with different PB concentration under 808 nm laser irradiation for 5 min. C) Temperature variations of PB@gellan (0.1 mg mL-1 for PB) under irradiation by the 808 nm for 5 cycles (3 min irradiation and then cooled down to room temperature for each cycle). D) Infrared thermal images of PB@gellan with different PB concentrations under 808 nm irradiation. The power density was set at 1.0 W cm-2.

ACS Paragon Plus Environment

13

ACS Applied Materials & Interfaces 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 32

3.3. Rheological properties of NC hydrogel. By dispersing CA4 powder into PB@gellan pre-gel at 50 oC under vortex mixing, CA4 is loaded into the hydrogel. The concentration of CA4 in the hydrogel is set at 6.0 mg mL-1 in order to meet a dose of 30 mg kg-1 for in vivo studies.14 The obtained NC hydrogel shows similar optical and photothermal property as PB@gellan hydrogel (Figure 2A and Figure S8, Supporting Information). The SEM image of NC hydrogel also shows a porous structure (Figure S3, Supporting Information). Gellan-based hydrogel is well known for its thermo-responsive gelling and shearthinning properties, as described in the Figure S9, Supporting Information and by previous studies.11,

57

The injectability of NC hydrogel was confirmed by a shear-

dependent viscosity test, and the result reveals an excellent shear-thinning property (Figure 3A). And the NC hydrogel has a broad linear viscoelastic region (0.01% ~ 1%) (Figure 3B). At the strain of 6%, the storage modulus (G’) is lower than the loss modulus (G’’), indicating the damage of hydrogel under large deformation, in accordance with the shear-thinning property. Furthermore, a good self-recovery was observed in the process of alternative strain sweep between 0.1% and 300% (Figure 3C). Temperature sweep shows an intersection of G’ and G’’ around 52 oC during the cooling of NC pre-gel (Figure 3D), which means a sol-to-gel transition above the physiological temperature. Furthermore, G’ and G’’ values are decreasing upon heating, implying that the noncovalent crosslinking of the hydrogel is reduced with temperature increase. The macromolecular disentanglement is dynamically slow as G’ is always higher than G’’ during the temperature increment. Hence, the NC hydrogel can resist the transient hyperthermia such as NIR-triggered photothermal heating. All the rheological tests

ACS Paragon Plus Environment

14

Page 15 of 32 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

ACS Applied Materials & Interfaces

manifest tremendous advantages of the NC hydrogel to serve as an injectable drug delivery depot.

Figure 3. Rheological properties of NC hydrogel. A) Shear rate-dependent viscosity. B) Straindependent moduli. C) Continuous step strain measurements (0.1% strain for 3 min, 300% strain for 30 s). D) Temperature-dependent moduli. 3.4. In vitro release of CA4. In the combinational therapy of PTT and VDA, a prolonged release behavior is preferable to maintain a relatively high concentration of CA4 in local site while minimizing the systematic toxicity. As expected, the cumulative release of CA4 is very low in PBS (less than 10% after 12 days, Figure 4A). The release kinetics is mainly ascribed to the hydrophobicity of CA4. If some surfactant (0.5% SDS) is added to the releasing medium to improve the solubility, the release of CA4 is accelerated (53.4%

ACS Paragon Plus Environment

15

ACS Applied Materials & Interfaces 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 16 of 32

for 24 h, Figure S10, Supporting Information), while the remaining hydrogel exhibits insignificant weight loss either in free PBS or with SDS added medium (~10% for 12 days, Figure S11, Supporting Information).

Figure 4. A) Release behavior of CA4 from NC hydrogel in different media at 37 oC (n = 3). B) Fluorescence imaging of mice after treated with free Cy5 in PBS, Cy5 in PB@gellan, and Cy5 in PB@gellan with 3 min 808 NIR irradiation (1.0 W cm-2). 3.5. In vivo retention of NC hydrogel. Cy5, a hydrophobic fluorescent dye, was selected as a drug model to track the in vivo distribution of hydrophobic CA4.54 Free Cy5 itself suffers fast diffusion and clearance after intratumoral injection (a rapid decay of fluorescence at one day post injection), while Cy5-loaded in the PB@gellan shows long retention in the tumor site (strong retention of fluorescence intensity at 12 day post-injection) (Figure 4B and Figure S12, Supporting information). Notably, the fluorescent intensity in the hydrogel treated group with NIR irradiation is close to that without NIR irradiation. The results further confirmed that the hydrogel shows good resistance against transient heating (below 60 oC in minutes, Figure 3D). Therefore, the PB@gellan hydrogel can restrict the movement and diffusion of cargos under NIR irradiation. As a result, a long-term anti-tumor effect of CA4 is feasible.

ACS Paragon Plus Environment

16

Page 17 of 32 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

ACS Applied Materials & Interfaces

Figure 5. A) Fluorescence images of calcein AM/PI co-stained 4T1 cells after photothermal treatment induced by PB@gellan (Control: gellan hydrogel without PB nanoparticles). B) Tumor-site temperature increment and C) infrared thermal images of mice before and after irradiation. The experiments were carried out under 808 nm laser irradiation for 3 min. The power density was set at 1.0 W cm-2. 3.5. In vitro and in vivo photothermal property. The localized photothermal effect of PB@gellan in vitro was evaluated on mouse breast cancer cells (4T1 cells). After exposure to 808 nm laser irradiation, the cells treated with PB@gellan show a boundary line after stained by the live/dead cell staining assay, indicating a localized photothermal effect of hydrogel (Figure 5A). In addition, the in vitro localized photothermal effect of NC hydrogel is quite similar as that of PB@gellan (Figure S13, Supporting Information). The photothermal effect and retention of PB@gellan in vivo was evaluated by recording

ACS Paragon Plus Environment

17

ACS Applied Materials & Interfaces 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 18 of 32

the in vivo temperature evolution of tumor-site. PB@gellan treated group shows only slightly decreased photothermal effect. After irradiation, the final temperature in successive 3 days is 54.1, 52.4 and 50.5 oC, respectively (Figure 5B and C). The temperature for effective hyperthermia therapy should be higher than 48 oC.58 In case of repeated photothermal therapy, a single injection of the PB@gellan hydrogel is available for multiple irradiations. In contrast, free PB treated group shows significantly reduced photothermal effect. The results indicate that free PB nanoparticles have a rapid clearance while the PB@gellan hydrogel is highly stable against metabolism. 3.6. In vivo anti-tumor synergistic therapy. In vivo anti-tumor efficacy of NC hydrogel was evaluated using the 4T1 tumor bearing mouse models. After the tumor volume reached ~150 mm3, the mice were divided into six groups randomly and received single intratumoral injection. The in vivo photothermal therapy was performed with an 808 nm NIR laser at 1.0 W cm-2 for 3 min. All of the tumor temperatures in the three groups (G4, G5, G6) reached about 52 oC. As compared with the groups of single therapy (G2 and G4), the group treated with NC hydrogel (G6) exhibits significant tumor regression during the treatment (Figure 6A and C), and only three in the overall five mice were found tiny tumors after the PTT-VDA therapy (TSR of 99.6%). The results show the synergistic effect of photothermal therapy of PB and vascular disrupting of CA4. Single PTT either with PB alone (G5) or PB@gellan (G4) partially suppresses the tumor growth at the very beginning, but their long-term anti-tumor effects are not satisfied because of a rapid secondary tumor growth (Figure 6A). In addition, the group treated with CA4loaded gellan hydrogel (G2) has stronger tumor inhibition effect than that of CA4 alone (G3), and the TSR values are 65.5% and 57.5%, respectively (Figure S14, Supporting

ACS Paragon Plus Environment

18

Page 19 of 32 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

ACS Applied Materials & Interfaces

Information). The enhanced efficacy in G2 is probably attributed to the sustained release of CA4 in the hydrogel.

Figure 6. A) Tumor volume and B) body weight of mice during treatment after single intratumoral injection. C) Photos and D) weight of tumor tissues obtained after treatment. The data were shown as mean ± standard deviation (n = 5; *p < 0.05, **p < 0.01, ***p < 0.001, G1: PBS, G2: CA4@gellan, G3: CA4, G4: PB@gellan with NIR, G5: PB with NIR, G6: NC hydrogel with NIR). H&E staining of tumor sections manifests an extensive necrosis area after the treatment of G6, and the residual tumors in G6 still stay in the atrophy state (Figure 7A and B). The tumor vessels were further studied through fluorescent immunohistochemical staining by CD31 antibody.13-14 After the whole treatment (lasting for 22 days), the vessel area in the treatment of G6 is much smaller than other groups (Figure 7C and Figure S15, Supporting

ACS Paragon Plus Environment

19

ACS Applied Materials & Interfaces 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 32

Information), which indicates that CA4 in NC hydrogel could greatly induce the disruption of tumor vessels. Furthermore, tumors treated in G2 (CA4@gellan) also exhibit less vessel signal than those treated in G3 (free CA4).

Figure 7. A) H&E staining of tumor tissues obtained after treatment. B) Quantitative analysis of H&E staining of tumor tissues. C) Quantitative analysis of immunohistochemical staining of tumor tissues. The data were shown as mean ± standard deviation (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). G1: PBS, G2: CA4@gellan, G3: CA4, G4: PB@gellan with NIR, G5: PB with NIR, G6: NC hydrogel with NIR. The antiangiogenic effect of CA4 was also confirmed on human umbilical vein endothelial cells (HUVECs). In accordance with the in vivo anti-tumor effects, the cell viability of HUVECs cells after treatment with the NC hydrogel and free CA4 decreases to 75.9% and 61.7%, respectively, whereas the cell viability of 4T1 tumor cells after

ACS Paragon Plus Environment

20

Page 21 of 32 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

ACS Applied Materials & Interfaces

treatment with the NC hydrogel and free CA4 shows almost negligible decrease (Figure S16, Supporting Information). The results suggest that CA4 selectively acts on HUVECs cells. In addition, the relatively lower cytotoxicity of NC hydrogel than free CA4 is possibly due to the sustained release of CA4. The outstanding synergistic anti-tumor efficacy is probably ascribed to the following aspects: (i) the photothermal effect of PB in gellan produces an intense anti-tumor effect in short periods, and (ii) the disruption of tumor vessels by CA4 consolidates the therapeutic effect in a prolonged release manner. The biocompatibility of NC hydrogel is confirmed with no obvious pathological changes (Figure S17, Supporting information) in H&E staining, and no obvious loss in body weight of mice as well (Figure 6B). The results indicate the safety of local administration of NC hydrogel. 4. CONCLUSIONS. In summary, an injectable hydrogel depot for co-delivery of CA4 and PB nanoparticles is designed for programmed anti-tumor combinational therapy. The NC hydrogel has excellent biocompatibility, high photothermal conversion efficiency and photothermal stability, injectability at body temperature and sustained release of CA4. More importantly, locally synergistic therapy of PTT and vascular disruption achieved remarkable tumor regression. Noteworthy, gellan is generally recognized as safe (GRAS) materials, and PB is approved by FDA. These features pave the way towards the clinical application of the NC hydrogel with facile preparation. ASSOCIATED CONTENT Supporting Information.

ACS Paragon Plus Environment

21

ACS Applied Materials & Interfaces 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 22 of 32

Additional experimental details; photothermal properties and stability characterization of the materials; the release behavior of CA4 and weight loss of the hydrogel during the release of CA4; additional characterization of the anti-tumor effect and biosafety of the hydrogels. AUTHOR INFORMATION Corresponding Author * E-mail: [email protected] (J. Li) Notes The authors declare no competing financial interest. ACKNOWLEDGMENT This work was financially supported by the National Natural Science Foundation of China (Nos. 21574147 and 21374133), Hunan Provincial Natural Science Foundation of China (No. 2018JJ2483), and Innovation-Driven Project of Central South University (No. 2017CX020).

REFERENCES (1) Chen, Q.; Xu, L.; Liang, C.; Wang, C.; Peng, R.; Liu, Z. Photothermal Therapy with Immune-Adjuvant Nanoparticles Together with Checkpoint Blockade for Effective Cancer Immunotherapy. Nat. Commun. 2016, 7, 13193. (2) Conde, J.; Oliva, N.; Zhang, Y.; Artzi, N. Local Triple-Combination Therapy Results in Tumour Regression and Prevents Recurrence in a Colon Cancer Model. Nat. Mater. 2016, 15, 1128-1140.

ACS Paragon Plus Environment

22

Page 23 of 32 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

ACS Applied Materials & Interfaces

(3) Xu, S.; Zhu, X.; Zhang, C.; Huang, W.; Zhou, Y.; Yan, D. Oxygen and Pt(ll) SelfGenerating Conjugate for Synergistic Photo-Chemo Therapy of Hypoxic Tumor. Nat. Commun. 2018, 9, 2053. (4) Jia, X.; Cai, X.; Chen, Y.; Wang, S.; Xu, H.; Zhang, K.; Ma, M.; Wu, H.; Shi, J.; Chen, H. Perfluoropentane-Encapsulated Hollow Mesoporous Prussian Blue Nanocubes for Activated Ultrasound Imaging and Photothermal Therapy of Cancer. ACS Appl. Mater. Interfaces 2015, 7, 4579-4588. (5) Liu, Y.; Shu, G.; Li, X.; Chen, H.; Zhang, B.; Pan, H.; Li, T.; Gong, X.; Wang, H.; Wu, X.; Dou, Y.; Chang, J. Human HSP70 Promoter-Based Prussian Blue Nanotheranostics for ThermoControlled Gene Therapy and Synergistic Photothermal Ablation. Adv. Funct. Mater. 2018, 28, 1802026. (6) Dou, Y.; Yang, X.; Yang, W.; Guo, Y.; Wu, M.; Liu, Y.; Li, X.; Zhang, X.; Chang, J. PB@Au Core-Satellite Multifunctional Nanotheranostics for Magnetic Resonance and Computed Tomography Imaging in Vivo and Synergetic Photothermal and Radiosensitive Therapy. ACS Appl. Mater. Interfaces 2017, 9, 1263-1272. (7) Wang, L.; Liang, K.; Jiang, X.; Yang, M.; Liu, Y.-N. Dynamic Protein–Metal Ion Networks: A Unique Approach to Injectable and Self-Healable Metal Sulfide/Protein Hybrid Hydrogels with High Photothermal Efficiency. Chem. Eur. J. 2018, 24, 6557-6563. (8) Vankayala, R.; Hwang, K. C. Near-Infrared-Light-Activatable Nanomaterial-Mediated Phototheranostic Nanomedicines: An Emerging Paradigm for Cancer Treatment. Adv. Mater. 2018, 30, 1706320.

ACS Paragon Plus Environment

23

ACS Applied Materials & Interfaces 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 24 of 32

(9) Li, H. A.; Li, H.; Liu, H.; Nie, T.; Chen, Y.; Wang, Z.; Huang, H.; Liu, L.; Chen, Y. Molecular Bottlebrush as a Unimolecular Vehicle with Tunable Shape for Photothermal Cancer Therapy. Biomaterials 2018, 178, 620-629. (10) Sheng, J.; Wang, L.; Han, Y.; Chen, W.; Liu, H.; Zhang, M.; Deng, L.; Liu, Y.-N. Dual Roles of Protein as a Template and a Sulfur Provider: A General Approach to Metal Sulfides for Efficient Photothermal Therapy of Cancer. Small 2018, 14, 1702529. (11) Zheng, Y.; Liang, Y.; Zhang, D.; Zhou, Z.; Li, J.; Sun, X.; Liu, Y.-N. Fabrication of Injectable CuS Nanocomposite Hydrogels Based on UCST-Type Polysaccharides for NIRTriggered Chemo-Photothermal Therapy. Chem. Commun. 2018, 54, 13805-13808. (12) Song, W.; Tang, Z.; Zhang, D.; Yu, H.; Chen, X. Coadministration of Vascular Disrupting Agents and Nanomedicines to Eradicate Tumors from Peripheral and Central Regions. Small 2015, 11, 3755-3761. (13) Zheng, Y.; Cheng, Y.; Chen, J.; Ding, J.; Li, M.; Li, C.; Wang, J.-c.; Chen, X. Injectable Hydrogel-Microsphere Construct with Sequential Degradation for Locally Synergistic Chemotherapy. ACS Appl. Mater. Interfaces 2017, 9, 3487-3496. (14) Wei, L.; Chen, J.; Zhao, S.; Ding, J.; Chen, X. Thermo-Sensitive Polypeptide Hydrogel for Locally Sequential Delivery of Two-Pronged Antitumor Drugs. Acta Biomater. 2017, 58, 4453. (15) Song, W.; Tang, Z.; Zhang, D.; Li, M.; Gu, J.; Chen, X. A Cooperative Polymeric Platform for Tumor-Targeted Drug Delivery. Chem. Sci. 2016, 7, 728-736. (16) Ouyang, J.; Wang, L.; Chen, W.; Zeng, K.; Han, Y.; Xu, Y.; Xu, Q.; Deng, L.; Liu, Y.-N. Biomimetic Nanothylakoids for Efficient Imaging-Guided Photodynamic Therapy for Cancer. Chem. Commun. 2018, 54, 3468-3471.

ACS Paragon Plus Environment

24

Page 25 of 32 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

ACS Applied Materials & Interfaces

(17) Yu, S.; Wang, C.; Yu, J.; Wang, J.; Lu, Y.; Zhang, Y.; Zhang, X.; Hu, Q.; Sun, W.; He, C.; Chen, X.; Gu, Z. Injectable Bioresponsive Gel Depot for Enhanced Immune Checkpoint Blockade. Adv. Mater. 2018, 30, 1801527. (18) Chen, Q.; Wang, C.; Zhang, X.; Chen, G.; Hu, Q.; Li, H.; Wang, J.; Wen, D.; Zhang, Y.; Lu, Y.; Yang, G.; Jiang, C.; Wang, J.; Dotti, G.; Gu, Z. In Situ Sprayed Bioresponsive Immunotherapeutic Gel for Post-Surgical Cancer Treatment. Nat. Nanotechnol. 2018, 14, 89-97. (19) Riley, R. S.; June, C. H.; Langer, R.; Mitchell, M. J. Delivery Technologies for Cancer Immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175-196. (20) Qin, S.-Y.; Cheng, Y.-J.; Lei, Q.; Zhang, A.-Q.; Zhang, X.-Z. Combinational Strategy for High-Performance Cancer Chemotherapy. Biomaterials 2018, 171, 178-197. (21) Cooney, M. M.; van Heeckeren, W.; Bhakta, S.; Ortiz, J.; Remick, S. C. Drug Insight: Vascular Disrupting Agents and Angiogenesis—Novel Approaches for Drug Delivery. Nat. Clin. Pract. Oncol. 2006, 3, 682-92. (22) Melamed, J. R.; Edelstein, R. S.; Day, E. S. Elucidating the Fundamental Mechanisms of Cell Death Triggered by Photothermal Therapy. ACS Nano 2015, 9 (1), 6-11. (23) Fan, W.; Yung, B.; Huang, P.; Chen, X. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem. Rev. 2017, 117, 13566-13638. (24) Cano-Mejia, J.; Burga, R. A.; Sweeney, E. E.; Fisher, J. P.; Bollard, C. M.; Sandler, A. D.; Cruz, C. R. Y.; Fernandes, R. Prussian Blue Nanoparticle-Based Photothermal Therapy Combined with Checkpoint Inhibition for Photothermal Immunotherapy of Neuroblastoma. Nanomedicine: NBM 2017, 13, 771-781.

ACS Paragon Plus Environment

25

ACS Applied Materials & Interfaces 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 26 of 32

(25) Sengupta, S.; Eavarone, D.; Capila, I.; Zhao, G.; Watson, N.; Kiziltepe, T.; Sasisekharan, R. Temporal Targeting of Tumour Cells and Neovasculature with a Nanoscale Delivery System. Nature 2005, 436, 568-72. (26) Shaked, Y.; Ciarrocchi, A.; Franco, M.; Lee, C. R.; Man, S.; Cheung, A. M.; Hicklin, D. J.; Chaplin, D.; Foster, F. S.; Benezra, R.; Kerbel, R. S. Therapy-Induced Acute Recruitment of Circulating Endothelial Progenitor Cells to Tumors. Science 2006, 313, 1785-1787. (27) Tozer, G.; Kanthou, C.; Baguley, B. Disrupting Tumour Blood Vessels. Nat. Rev. Cancer 2005, 5, 423-435. (28) Yang, W. J.; Zhou, P.; Liang, L.; Cao, Y.; Qiao, J.; Li, X.; Teng, Z.; Wang, L. NanogelIncorporated Injectable Hydrogel for Synergistic Therapy Based on Sequential Local Delivery of Combretastatin-A4 Phosphate (CA4P) and Doxorubicin (DOX). ACS Appl. Mater. Interfaces 2018, 10, 18560-18573. (29) Li, T.; Zhang, M.; Wang, J.; Wang, T.; Yao, Y.; Zhang, X.; Zhang, C.; Zhang, N. Thermosensitive Hydrogel Co-loaded with Gold Nanoparticles and Doxorubicin for Effective Chemoradiotherapy. AAPS J. 2016, 18 (1), 146-155. (30) Segovia, N.; Pont, M.; Oliva, N.; Ramos, V.; Borros, S.; Artzi, N. Hydrogel Doped with Nanoparticles for Local Sustained Release of siRNA in Breast Cancer. Adv. Healthcare Mater. 2015, 4 (2), 271-280. (31) Zhang, Z.-Q.; Song, S.-C. Thermosensitive/Superparamagnetic Iron Oxide NanoparticleLoaded Nanocapsule Hydrogels for Multiple Cancer Hyperthermia. Biomaterials 2016, 106, 1323. (32) Zhang, X.; Xia, L.-Y.; Chen, X.; Chen, Z.; Wu, F.-G. Hydrogel-Based Phototherapy for Fighting Cancer and Bacterial Infection. Sci. China-Mater. 2017, 60, 487-503.

ACS Paragon Plus Environment

26

Page 27 of 32 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

ACS Applied Materials & Interfaces

(33) Qi, Y.; Min, H.; Mujeeb, A.; Zhang, Y.; Han, X.; Zhao, X.; Anderson, G. J.; Zhao, Y.; Nie, G. Injectable Hexapeptide Hydrogel for Localized Chemotherapy Prevents Breast Cancer Recurrence. ACS Appl. Mater. Interfaces 2018, 10, 6972-6981. (34) Hosoya, H.; Dobroff, A. S.; Driessen, W. H. P.; Cristini, V.; Brinker, L. M.; Staquicini, F. I.; Cardo-Vila, M.; D'Angelo, S.; Ferrara, F.; Proneth, B.; Lin, Y.-S.; Dunphy, D. R.; Dogra, P.; Melancon, M. P.; Stafford, R. J.; Miyazono, K.; Gelovani, J. G.; Kataoka, K.; Brinker, C. J.; Sidman, R. L.; Arap, W.; Pasqualini, R. Integrated Nanotechnology Platform for TumorTargeted Multimodal Imaging and Therapeutic Cargo Release. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, 1877-1882. (35) Zhang, W.; Ning, C.; Xu, W.; Hu, H.; Li, M.; Zhao, G.; Ding, J.; Chen, X. PrecisionGuided Long-Acting Analgesia by Gel-Immobilized Bupivacaine-Loaded Microsphere. Theranostics 2018, 8 (12), 3331-3347. (36) Zhang, W.; Xu, W.; Ning, C.; Li, M.; Zhao, G.; Jiang, W.; Ding, J.; Chen, X. Long-Acting Hydrogel/Microsphere Composite Sequentially Releases Dexmedetomidine and Bupivacaine for Prolonged Synergistic Analgesia. Biomaterials 2018, 181, 378-391. (37) Zhang, Y.; Zhang, J.; Xu, W.; Xiao, G.; Ding, J.; Chen, X. Tumor MicroenvironmentLabile Polymer-Doxorubicin Conjugate Thermogel Combined with Docetaxel for In Situ Synergistic Chemotherapy of Hepatoma. Acta Biomater. 2018, 77, 63-73. (38) Xing, R.; Liu, K.; Jiao, T.; Zhang, N.; Ma, K.; Zhang, R.; Zou, Q.; Ma, G.; Yan, X. An Injectable Self-Assembling Collagen-Gold Hybrid Hydrogel for Combinatorial Antitumor Photothermal/Photodynamic Therapy. Adv. Mater. 2016, 28 (19), 3669-3676.

ACS Paragon Plus Environment

27

ACS Applied Materials & Interfaces 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 28 of 32

(39) Melancon, M. P.; Elliott, A. M.; Shetty, A.; Huang, Q.; Stafford, R. J.; Li, C. NearInfrared Light Modulated Photothermal Effect Increases Vascular Perfusion and Enhances Polymeric Drug Delivery. J. Controlled Release 2011, 156 (2), 265-272. (40) Diagaradjane, P.; Shetty, A.; Wang, J. C.; Elliott, A. M.; Schwartz, J.; Shentu, S.; Park, H. C.; Deorukhkar, A.; Stafford, R. J.; Cho, S. H.; Tunnell, J. W.; Hazle, J. D.; Krishnan, S. Modulation of in Vivo Tumor Radiation Response via Gold Nanoshell-Mediated VascularFocused Hyperthermia: Characterizing an Integrated Antihypoxic and Localized Vascular Disrupting Targeting Strategy. Nano Lett. 2008, 8 (5), 1492-1500. (41) Green, H. N.; Crockett, S. D.; Martyshkin, D. V.; Singh, K. P.; Grizzle, W. E.; Rosenthal, E. L.; Mirov, S. B. A Histological Evaluation and In Vivo Assessment of Intratumoral Near Infrared Photothermal Nanotherapy-Induced Tumor Regression. Int. J. Nanomed. 2014, I9, 5093-5102. (42) Seshadri, M.; Bellnier, D. A. The Vascular Disrupting Agent 5,6-Dimethylxanthenone-4Acetic Acid Improves the Antitumor Efficacy and Shortens Treatment Time Associated with Photochlor-sensitized Photodynamic Therapy In Vivo. Photochem. Photobiol. 2009, 85, 50-56. (43) Ng, Q.-S.; Mandeville, H.; Goh, V.; Alonzi, R.; Milner, J.; Carnell, D.; Meer, K.; Padhani, A. R.; Saunders, M. I.; Hoskin, P. J. Phase Ib Trial of Radiotherapy in Combination with Combretastatin-A4-Phosphate in Patients with Non-Small-Cell Lung Cancer, Prostate Adenocarcinoma, and Squamous Cell Carcinoma of the Head and Neck. Ann. Oncol. 2012, 23, 231-237. (44) Zhou, J.; Li, M.; Hou, Y.; Luo, Z.; Chen, Q.; Cao, H.; Huo, R.; Xue, C.; Sutrisno, L.; Hao, L.; Cao, Y.; Ran, H.; Lu, L.; Li, K.; Cai, K. Engineering of a Nanosized Biocatalyst for

ACS Paragon Plus Environment

28

Page 29 of 32 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

ACS Applied Materials & Interfaces

Combined Tumor Starvation and Low-Temperature Photothermal Therapy. ACS Nano 2018, 12, 2858-2872. (45) Cheng, L.; Gong, H.; Zhu, W.; Liu, J.; Wang, X.; Liu, G.; Liu, Z. PEGylated Prussian Blue Nanocubes as a Theranostic Agent for Simultaneous Cancer Imaging and Photothermal Therapy. Biomaterials 2014, 35, 9844-9852. (46) Chen, W.; Zeng, K.; Liu, H.; Ouyang, J.; Wang, L.; Liu, Y.; Wang, H.; Deng, L.; Liu, Y.N. Cell Membrane Camouflaged Hollow Prussian Blue Nanoparticles for Synergistic Photothermal-/Chemotherapy of Cancer. Adv. Funct. Mater. 2017, 27, 1605795. (47) Shokouhimehr, M.; Soehnlen, E. S.; Hao, J.; Griswold, M.; Flask, C.; Fan, X.; Basilion, J. P.; Basu, S.; Huang, S. D. Dual Purpose Prussian Blue Nanoparticles for Cellular Imaging and Drug Delivery: A New Generation of T1-Weighted MRI Contrast and Small Molecule Delivery Agents. J. Mater. Chem. 2010, 20, 5251-5259. (48) Cai, X.; Gao, W.; Zhang, L.; Ma, M.; Liu, T.; Du, W.; Zheng, Y.; Chen, H.; Shi, J. Enabling Prussian Blue with Tunable Localized Surface Plasmon Resonances: Simultaneously Enhanced Dual-Mode Imaging and Tumor Photothermal Therapy. ACS Nano 2016, 10, 1111511126. (49) Liu, Y.; Guo, Q.; Zhu, X.; Feng, W.; Wang, L.; Ma, L.; Zhang, G.; Zhou, J.; Li, F. Optimization of Prussian Blue Coated NaDyF4:x%Lu Nanocomposites for Multifunctional Imaging-Guided Photothermal Therapy. Adv. Funct. Mater. 2016, 26, 5120-5130. (50) Hu, M.; Furukawa, S.; Ohtani, R.; Sukegawa, H.; Nemoto, Y.; Reboul, J.; Kitagawa, S.; Yamauchi, Y. Synthesis of Prussian Blue Nanoparticles with a Hollow Interior by Controlled Chemical Etching. Angew. Chem., Int. Ed. 2012, 51, 984-988.

ACS Paragon Plus Environment

29

ACS Applied Materials & Interfaces 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 30 of 32

(51) Sun, C.; Wen, L.; Zeng, J.; Wang, Y.; Sun, Q.; Deng, L.; Zhao, C.; Li, Z. One-Pot Solventless Preparation of PEGylated Black Phosphorus Nanoparticles for Photoacoustic Imaging and Photothermal Therapy of Cancer. Biomaterials 2016, 91, 81-89. (52) Chen, J.; Tao, N.; Fang, S.; Chen, Z.; Liang, L.; Sun, X.; Li, J.; Liu, Y.-N. Incorporation of Fmoc-Y Nanofibers into Ca-Alginate Hydrogels for Improving their Mechanical Properties and the Controlled Release of Small Molecules. New J. Chem. 2018, 42, 9651-9657. (53) Liu, T.; Zhang, D.; Song, W.; Tang, Z.; Zhu, J.; Ma, Z.; Wang, X.; Chen, X.; Tong, T. A Poly(L-glutamic acid)-Combretastatin A4 Conjugate for Solid Tumor Therapy: Markedly Improved Therapeutic Efficiency through its Low Tissue Penetration in Solid Tumor. Acta Biomater. 2017, 53, 179-189. (54) Lu, Z.; Li, Y.; Shi, Y.; Li, Y.; Xiao, Z.; Zhang, X. Traceable Nanoparticles with Spatiotemporally Controlled Release Ability for Synergistic Glioblastoma Multiforme Treatment. Adv. Funct. Mater. 2017, 27, 1703967. (55) Nossol, E.; Zarbin, A. J. G. A Simple and Innovative Route to Prepare a Novel Carbon Nanotube/Prussian Blue Electrode and its Utilization as a Highly Sensitive H2O2 Amperometric Sensor. Adv. Funct. Mater. 2009, 19 (24), 3980-3986. (56) Fu, G.; Liu, W.; Li, Y.; Jin, Y.; Jiang, L.; Liang, X.; Feng, S.; Dai, Z. Magnetic Prussian Blue Nanoparticles for Targeted Photothermal Therapy under Magnetic Resonance Imaging Guidance. Bioconjugate Chem. 2014, 25, 1655-1663. (57) Zheng, Y.; Liang, Y.; Zhang, D.; Sun, X.; Liang, L.; Li, J.; Liu, Y.-N. Gelatin-Based Hydrogels Blended with Gellan as an Injectable Wound Dressing. ACS Omega 2018, 3 (5), 4766-4775.

ACS Paragon Plus Environment

30

Page 31 of 32 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

ACS Applied Materials & Interfaces

(58) Jaque, D.; Martinez Maestro, L.; del Rosal, B.; Haro-Gonzalez, P.; Benayas, A.; Plaza, J. L.; Martin Rodriguez, E.; Garcia Sole, J. Nanoparticles for Photothermal Therapies. Nanoscale 2014, 6, 9494-9530.

ACS Paragon Plus Environment

31

ACS Applied Materials & Interfaces 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 32 of 32

Table of Contents

ACS Paragon Plus Environment

32