Near-Infrared Polymeric Nanoparticles with High Content of Cyanine

Aug 9, 2016 - The formed nanoparticles could be used for bimodal bioimaging and ... The bimodal bioimaging provided complementary message about ...
0 downloads 0 Views 2MB Size
Subscriber access provided by CORNELL UNIVERSITY LIBRARY

Article

Near-Infrared Polymeric Nanoparticles with High Content of Cyanine for Bimodal Imaging and Photothermal Therapy Wenhai Lin, Yang Li, Wei Zhang, Shi Liu, Zhigang Xie, and Xiabin Jing ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.6b07103 • Publication Date (Web): 09 Aug 2016 Downloaded from http://pubs.acs.org on August 31, 2016

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

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

Page 1 of 25

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

Near-Infrared Polymeric Nanoparticles with High Content of Cyanine for Bimodal Imaging and Photothermal Therapy Wenhai Lin,†, ‡ Yang Li, § Wei Zhang,†, ‡ Shi Liu,† Zhigang Xie*,† and Xiabin Jing† †

State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied

Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, Jilin 130022, P. R. China ‡

University of Chinese Academy of Sciences, Beijing 100049, P. R. China

§

College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao

060004, P.R. China

ABSTRACT: The discovery and synthesis of theranostic nanomedicines with high loading of imaging and therapeutic agents is challenging. In this work, a polymer assembling strategy was used to make nanoparticles with exceptionally high loading of theranostic agent. As an example, poly(heptamethine) was synthesized via multi-component Passerini reaction, and then assembled into nanoparticles in the presence of poly(ethylene glycol)2k-block-poly(D,L-lactide)2k (PEGPLA) with high heptamethine loading (> 50 %). The formed nanoparticles could be used for bimodal

bioimaging

and

photothermal

therapy.

The

bimodal

bioimaging

provided

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 25

complementary message about biodistribution, and photothermal treatment inhibited the growth of cervical carcinoma upon laser irradiation. This assembly of polymers formed by imaging and therapeutic agents opens new possibilities for the construction of multifunctional nanomedicines.

Keywords: near-infrared, fluorescence imaging, photoacoustic imaging, photothermal therapy. INTRODUCTION In order to track nanomedicines systemically and facilitate clinical translation, there has been an increasing focus on imaging techniques, such as near-infrared fluorescence imaging (NIRF),1,2 photoacoustic imaging (PA),3,4 magnetic resonance imaging,5,6 computed tomography7 and positron emission tomography8 for monitoring drug delivery and tumor treatment.9 However, a single imaging technique is not enough for the course of cancer diagnosis, prognosis and therapy due to the limitations in specificity, sensitivity, resolution and imaging depths. An ideal strategy is to develop multimodal imaging system, which allows for the integration of complementary information for improved analysis.10-12 A good example is incorporation of NIR and PA, which would afford a real-time, comprehensive, intact way for monitoring drug biodistribution, mastering drug delivery process and tumor diagnosis.13 However, direct incorporating two imaging agents and drugs into one nanoplatform is complicated and not easily controlled with desired formula.14,15 Polymeric nanoparticles (PNPs) are one of the most widely used formulations for medicines.16 Various drugs and imaging agents can be incorporated into PNPs through the self-assembling process.17 However, physical package of the functional molecules into PNPs possesses several challenges, including low loading content and loading efficiency, poor stability and uncontrolled drug release.18,19 For a theranostic nanomedicine, it will be very complicated for loading imaging

ACS Paragon Plus Environment

2

Page 3 of 25

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

and therapeutic agents synchronously in one formulation. Cyanine (Cy) is a versatile organic dye and has many advantages, such as a NIR imaging window, high extinction coefficients and narrow absorption bands, which are promising for cancer imaging.20,21 It is well known that indocyanine green (ICG) could be used in medical diagnostics.22-24 Cy also could act as medicaments for photothermal therapy (PTT).25,26 Nanoparticles containing Cy dye could be obtained via encapsulation or chemical modification with enhanced physiochemical properties, which can overcome the limitations of organic dyes, including poor solubility, short circulation time and low tumor accumulation. Cai et al. used human serum albumin-ICG nanoscale formulations for cancer theranostics.27 We prepared ICG-loaded nanocapsules for enhanced photothermal therapy.28 However, the content of Cy in these formulations usually is lower than 20%, and large amount of polymeric carrier is undesirable due to their unknown toxicity.18,28 Recently, Cheng et al. reported the dimeric drug-encapsulating polymeric NPs with high drug loading (> 50%).29 We hypothesize that using polymers of therapeutic agents will be a better way to fabricate nanomedicines with high content of theranostic agents.

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 25

Scheme 1. Synthetic route of CyP and preparing CyP@PEG-PLA NPs.

ACS Paragon Plus Environment

4

Page 5 of 25

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

In this work, theranostic polymers were used to assemble into nanoparticles with super high content of imaging and therapeutic agents. Firstly, we made a heptamethine Cy-containing polymer (CyP) via a one-pot multi-component Passerini reaction (Scheme 1). Then CyP was coprecipitated with PEG-PLA to form the CyP@PEG-PLA nanoparticles (CyP@PEG-PLA NPs). CyP@PEG-PLA NPs showed not only outstanding imaging capability for NIRF and PA but also effective tumor inhibition through local hyperthermia in vivo. RESULTS AND DISCUSSION At first, the Cy containing two carboxylic acids (CyCOOH) was made.26 CyCOOH was mixed with o-nitrobenzaldehyde and 1,6-diisocyanohexane in methylene chloride, and stirring for 96 hours. CyP was obtained after purification by silica gel column chromatography, and characterized by proton nuclear magnetic resonance (1H NMR) and size exclusion chromatography (SEC). Intensity ratios for protons signals corresponding to CyP confirmed the successful synthesis of CyP (Figure S1, supporting information). Content of CyCOOH in CyP was 63.5 wt % according to the molecular structure. Passerini reaction was widely used in synthesis of various polymers.30-33 Moreover, one peak was found in the SEC trace of CyP (Figure S1B). The calculated number average molecular weight is 9100 and the polydispersity is 1.17. The optical properties of CyCOOH and CyP were similar in dimethylformamide (DMF) and both of absorption and emission spectra located in the NIR region (700-850 nm) as seen in Figure S1C and S1D. And photothermal conversion ability kept unchanged before and after the conjugation (Figure S2).

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 25

Figure 1. A) Photos of CyCOOH@PEG-PLA NPs and CyP@PEG-PLA NPs. B) The TEM photo of CyP@PEG-PLA NPs (77.4 %). Scale bar, 500 nm. C) Absorption spectra of CyP@PEG-PLA NPs in H2O and H2O+DMF (v/v, 1:1). D) and E) PA signal of CyP@PEG-PLA in water+DMF and water. The numbers are the concentrations of CyP. Unit, µg mL-1. The hydrophobic CyP cannot self-assemble into nanoparticles directly, so PEG-PLA was chosen to facilitate the formation of nanoparticles.34 CyP and PEG-PLA could assemble into CyP@PEG-PLA NPs in water. Briefly, we added an acetone solution of CyP and PEG-PLA into

ACS Paragon Plus Environment

6

Page 7 of 25

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

water, and stirred for several hours. The mixture was dialyzed for 1 day. The content of CyP in CyP@PEG-PLA NPs was determined by calibration curve of UV-Vis. When CyP and PEG-PLA were co-precipitated at a ratio of 1:1 (w/w), the loading content and loading efficiency of CyP was 44.2 wt % and 79.3 %, respectively. CyCOOH content in CyP@PEG-PLA NPs was about 28.1 wt %. However, the loading content was 2.7 wt % and loading efficiency was 2.7 % when CyCOOH and PEG-PLA were co-precipitated at the same weight ratio. A ten times increasing of CyCOOH content in polymeric nanoparticles was realized by using the polymer of CyCOOH. The green color of CyP@PEG-PLA NPs (right photo, Figure 1A) was much stronger than that of CyCOOH@PEG-PLA NPs (left photo, Figure 1A). With the weight ratio of CyP and PEG-PLA increasing from 1:1 to 10:1, CyCOOH loading content increased from 28.1 to 53.1 wt % (Table S1). The maximum contents of CyP and CyCOOH in NPs were 83.6 and 53.1 wt %, respectively. The morphology and size distribution of CyP@PEG-PLA NPs with different loading contents were compared. Transmission electron microscopy (TEM) images (Figure 1B and Figure S3) showed the typical spherical nanoparticles with average diameter of 50 nm. The diameter between 90 nm and 100 nm obtained dynamic light scattering (DLS) were listed in Table S1. Distinct stability for CyP@PEG-PLA NPs in physiological condition was validated by the unchanged size for half a year (Table S2). And CyP and CyP@PEG-PLA NPs were a little more stable than CyCOOH according to absorption changes at 790 nm upon irradiation (2 W cm-2) for 150 seconds (Figure S4). The optical properties of CyP@PEG-PLA NPs were recorded in water. The mixture of DMF and water could dissolve and destroy the nanoparticles. As shown in Figure 1C, the maximum absorption of CyP@PEG-PLA NPs in water was red-shifted because CyP was successfully encapsulated by PEG-PLA and possibly formed aggregates in NPs.28 The maximum fluorescence

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 25

wavelength of CyP@PEG-PLA in DMF+water was centered at 831 nm, but no fluorescence was observed for CyP@PEG-PLA NPs in water because of aggregation-caused quenching (ACQ) (Figure S3D).34 CyP@PEG-PLA NPs could disassemble in the presence of DMF, and released CyP to emit fluorescence. Photos of CyP@PEG-PLA in water and DMF+water were taken to visually observe the phenomenon of ACQ. With increasing concentration of CyP, the fluorescence of CyP@PEG-PLA in DMF+water gradually became strong (Figure S3E). We then detected the photoacoustic signal of CyP@PEG-PLA through phantom test by multispectral photoacoustic tomography (MSOT).36 Both of CyP@PEG-PLA NPs in water and DMF+water exhibited enhanced photoacoustic signal with the increasing concentration of CyP as shown in Figure 1D and 1E. These results demonstrated that CyP@PEG-PLA NPs possess the ability for bimodal NIRF and PA imaging.

ACS Paragon Plus Environment

8

Page 9 of 25

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 2. Cytotoxicity of A) CyP and B) CyP@PEG-PLA NPs toward HeLa cells. C) Heating curves of CyP@PEG-PLA NPs and PEG-PLA NPs irradiated by NIR laser. D) Cytotoxicity of CyP@PEG-PLA NPs upon irradiation of 2 W cm-2. The cellular uptake and cytocompatibility were investigated by using the sample of CyP@PEG-PLA NPs with CyP content of 44.2 wt %. Firstly, the cellular uptake in human cervical carcinoma (HeLa) cells was evaluated by using confocal laser scanning microscopy (CLSM). The intensive red fluorescence from 0.5 to 2 h (Figure S5) indicated that CyP@PEGPLA NPs could be endocytosed and CyP could be released in cells. The cytotoxicities of CyP and CyP@PEG-PLA NPs toward HeLa cells were calculated. CyP did not exhibit obvious cell inhibition effects in 48 h even with concentrations up to 40 µg mL-1 (Figure 2A). About 60 % of

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 25

cells became dead in the presence of 40 µg mL-1 of CyP because CyP were cytotoxic to cancer cells at high concentrations with incubation for a long time.20 However, CyP@PEG-PLA NPs showed better biocompatibility than CyP because of nanoscale formulation (Figure 2B). In order to effectively inhibit cancer cells proliferation, CyP@PEG-PLA NPs could be used as photothermal agents. The temperature of CyP@PEG-PLA NPs in water with CyP content of 44.4 µg mL-1 increased from 24.4 oC to 44.5 oC while PEG-PLA solution exhibited few increase of temperature after irradiation (Figure 2C). The efficiency of photothermal conversion is 12 % on the basis of Figure S6. Then photothermal therapy experiment towards HeLa cell was carried out. After irradiated for 5 min, more than 60 % of HeLa cells were killed in 24 h (Figure 2D), indicating that the formed CyP@PEG-PLA NPs still kept the photothermal activity for cancer therapy.

ACS Paragon Plus Environment

10

Page 11 of 25

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 3. A) NIRF and B) PA imaging of HeLa-tumor-containing mouse administered with CyP@PEG-PLA NPs (100µg mL-1, 100µL) intratumorally. The area of white circle is the tumor.

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 25

Cancer imaging and therapy were further studied in living animal. The mouse with HeLa tumor was injected intratumorally with CyP@PEG-PLA NPs (100µg mL-1, 100µL). As shown in Figure 3A, NIRF become strong at first, and then weak in 48 h because nanoparticles were gradually destroyed and CyP was slowly released in tumor microenvironment to emit fluorescence. MOST images showed that CyP@PEG-PLA NPs had the ability of migration inside tumor, as the photoacoustic signal was seen in the depths of the tumor (Figure 3B) and in more tumor areas (Figure S7). Furthermore, mice (n=3) were administered intravenously with CyP@PEG-PLA NPs, and the imaging of NIRF and PA imaging was recorded. As time went by, NIRF enhanced in tumor until 32 h and then weakened slowly in 69 h (Figure 4A). Meanwhile, NIRF was observed in organs such as liver, intestines, spleen and so on. There was little NIRF signal in tumor and organs in 2 h because CyP@PEG-PLA NPs still kept the nanoscale structure. NIRF in liver and intestines became strong due to the accumulation and dissociation of nanoparticles in 20 h, which also demonstrated that CyP@PEG-PLA NPs were biodegradable and could be eliminated from the body. After 32 h, the intensity of NIRF in tumor was similar to that in liver. NIRF was strong in tumor site even in 69 h. This long time retention of NIRF was ascribed to the nanoscale formulation, which enhanced the accumulation and cellular uptake.37 However, PA imaging offered additional information. The PA signal in liver enhanced in 20 h and weakened in 69 h because of metabolism to be excreted from the body. Comparing with PA signal in liver, the PA signal in tumor remained weak all the time in 69 h (Figure 4B and Figure S8), indicating the low tumor accumulation of CyP@PEG-PLA NPs. We quantified the intensities and calculated the intensity ratios of NIRF and PA in tumor and liver at 32 h (Figure 4 and Figure S9). To our surprise, the ratio for NIRF was 0.87 ± 0.06 while the ratio for PA was

ACS Paragon Plus Environment

12

Page 13 of 25

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

only 0.10 ± 0.04. This result was ascribed to the different penetration depths and detectable signals of NIRF and PA. The depth of NIRF imaging is less than 8 mm,38 while the PA enables penetration depths of several centimetres.3 In addition, NIRF could be detected only when the CyP@PEG-PLA NPs were destroyed, while PA imaging could not distinguish whether CyP were packed in NPs. NIRF and PA imaging as noninvasive imaging techniques are widely used for monitoring drug delivery and release. We could clearly observe NIRF and PA signal in a living mouse for different times in Figure 3 and Figure 4, which partly reflected the biodistribution in tissue of mice. No fluorescence was observed for CyP@PEG-PLA NPs in water because of ACQ as revealed in Figure S2D, which meant that NIRF imaging could not detect CyP@PEG-PLA NPs. When CyP@PEG-PLA NPs were destroyed to release CyP, the NIRF was observed. So the fluorescence intensity could only stand for a part of CyP released from CyP@PEG-PLA NPs. What’s more, the depth of NIRF imaging was less than 8 mm, which demonstrated that NIRF of CyP could not be detected in deep tissues and the NIRF showed in Figure 3A and 4A came from CyP released for NPs in the superficial tissues. So NIRF imaging was not good for studying CyP@PEG-PLA NPs but might be suitable for monitoring CyP released from CyP@PEG-PLA. As for PA imaging, both of CyP@PEG-PLA NPs in water and CyP in DMF exhibited strong PA signal (Figure 1D and 1E). Furthermore, the PA possesses penetration depths of several centimeters. So the total CyP was detected in PA imaging in vivo. We could not observe the tumor clearly according to PA imaging of the total body because the large collection of NPs in liver for elimination led to strong PA signal in liver. The PA signal in tumor was too weak to be observed comparing with signal in liver. So it was useful for PA imaging to monitor metabolism of total CyP but PA imaging could not detect the release of CyP from NPs. In a short, both NIRF and PA were indispensable for monitoring drug delivery.

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 25

Figure 4. A) NIRF and B) PA imaging of tumor-bearing mouse. The area of black or white circle is the tumor.

ACS Paragon Plus Environment

14

Page 15 of 25

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

Mice with HeLa tumor was used for cancer treatment. Temperature of the tumor was 39.2 oC upon 808 nm light irradiation for 0.5 h with the mice injected with saline (Figure 5A). The temperature reached to 44 oC for the mouse injected intratumorally with CyP@PEG-PLA NPs (200 µg mL-1,150 µL) upon irradiation. The temperature was kept between 42 oC to 45 oC under light irradiation for half an hour because this temperature could cause irreversible tumor cellular damage.39-41 Tumors treated by saline with or without laser irradiation grew rapidly in 12 days, indicating laser irradiation did not affect the tumor growth (Figure 5B). The growth of tumors was slightly inhibited by CyP@PEG-PLA NPs without irradiation because CyP was toxic to HeLa cells at the high concentration. The experimental group was treated by CyP@PEG-PLA NPs under irradiation. The growth of tumors was successfully inhibited because of photothermal treatment. The photo of the represent tumors was shown in Figure 5C, showing the tumor in the experimental group was the smallest in these tumors. The potential adverse effect of photothermal agents is evaluated by the variation of body weight. All animals showed very little body weight reduction (< 10 %) after treatments (Figure 5D). No significant difference between these groups was observed, suggesting that these treatments were well tolerated and no acute toxicity at the current dose. Tumor was stained with hematoxylin and eosin (H&E) at the 12th day (Figure S10). Cells of tumor tissue were killed after treatment by CyP@PEG-PLA NPs and irradiation. More importantly, H&E staining showed no apparent abnormality or lesion for major organs after treatment of CyP@PEG-PLA NPs, indicating that CyP@PEG-PLA NPs possess a good biocompatibility (Figure S11).

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 25

Figure 5. A) Infrared thermograms of mice. B) The tumor volume ratio of mice in 12 days. C) The representative tumors of mice. D) The body weight of mice in 12 days.

CONCLUSION In summary, a theranostic nanomedicine (CyP@PEG-PLA NPs) was constructed via polymer co-precipitation method. The as-prepared CyP@PEG-PLA NPs could be used for NIRF and PA imaging and PTT simultaneously. Unlike direct encapsulation of drug molecules, this polymer assembling approach show great potential in improving drug content, which is possibly applied

ACS Paragon Plus Environment

16

Page 17 of 25

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

to miscellaneous drug delivery systems. This work highlights the potential of using polymers of therapeutics to develop the multifunctional nanoparticle for cancer treatment. METHODS Materials: Heptamethine cyanine containing a dicarbocycbic acid (CyCOOH) was synthesized as described in the literature.26 PEG-PLA was synthesized in our previous work.34 All reagents were available commercially and used without further treatment. The instruments used in this work have been provided in our previous work.32 SEC on a PL GPC50PLUS instrument purchased from Varian (column: PLgel-Mixed-D×2) was used at 50 oC (eluent: DMF, rate: 0.6 mL min-1). CyP was synthesized as described in reference by using CyCOOH as diacid.32 Preparation of CyP@PEG-PLA NPs: CyP (10 mg) and PEG-PLA (10 mg) were dissolved in acetone (5 mL). We added the mixture into water, and then stirred for several hours. The mixture was dialyzed for 1 day. The solution was filtered using disposable 450 nm Millipore filters prior to analysis. CyP@PEG-PLA NPs (44.2%) were obtained. CyP@PEG-PLA NPs (77.4% and 83.6%) were obtained through changing the weight of CyP. CyCOOH@ PEG-PLA NPs was made with similar method as CyP@PEG-PLA NPs. Cell experiments: The experimental procedure were given in our previous work.28, 32, 34 Photothermal Experiments: CyP@PEG-PLA NPs with different concentrations were treated under an 808 nm laser with a temperature detector used to monitor their temperature. Conversion efficiency was obtained according to previous work.42 The detail for cell ablation were provided in our previous work.[34] Briefly, Cells were incubated CyP@PEG-PLA NPs for 6 h, then

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 25

irradiated by the NIR laser (wavelength: 808 nm, 2W min-1) for 5 min. The cells were incubated for another 18 h after irradiation. Animal experiments: mice were treated according to the regulation of Research Animals established by Jilin University Studies Committee. HeLa-tumor mice with naked-eye-visible tumor nodules in the armpit of the left anterior limb were applied. Tumor volume was calculated according to published procedure.42 For PTT, the mice were treated by different formulations: 1) CyP@PEG-PLA NPs + laser; 2) CyP@PEG-PLA NPs; 3) saline + laser; 4) saline. The mice were intratumorally injected with saline (150 µL) or CyP@PEG-PLA NPs (150 µL). 2 h after injuection, therapy was conducted. The tumors of group 1 were treated by NIR laser (maximal power: 0.9 W/ cm2; the diameter of spot: 8 mm) to adjust the temperature ranging 42 oC to 45 oC for 0.5 h. The tumors of group 3 were irradiated for 0.5 h (0.9 W cm-2). ASSOCIATED CONTENT Supporting Information. The supporting information is available free of charge via the Internet at http://pubs.acs.org.

AUTHOR INFORMATION Corresponding Author *Zhigang Xie E-mail: [email protected] ACKNOWLEDGMENT

ACS Paragon Plus Environment

18

Page 19 of 25

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

We thank the financial support from the National Natural Science Foundation of China (Project No. 51522307). REFERENCES 1.

Luo, S.; Zhang, E.; Su, Y.; Cheng, T.; Shi, C. A Review of NIR Dyes in Cancer Targeting and Imaging. Biomaterials 2011, 32, 7127-7138.

2.

Liu, L.; Fu, L.; Jing, T.; Ruan, Z.; Yan, L. pH-Triggered Polypeptides Nanoparticles for Efficient BODIPY Imaging-Guided Near Infrared Photodynamic Therapy. ACS Appl. Mater. Interfaces 2016, 8, 8980-8990.

3.

Taruttis, A.; Ntziachristos, V. Advances in Real-Time Multispectral Optoacoustic Imaging and Its Applications. Nat. Photonics 2015, 9, 219-227.

4.

Wang, G.; Zhang, F.; Tian, R.; Zhang, L.; Fu, G.; Yang, L.; Zhu, L. Nanotubes-Embedded Indocyanine Green–Hyaluronic Acid Nanoparticles for Photoacoustic-Imaging-Guided Phototherapy. ACS Appl. Mater. Interfaces 2016, 8, 5608-5617.

5.

Zhang, B.; Li, Q.; Yin, P.; Rui, Y.; Qiu, Y.; Wang, Y.; Shi, D. Ultrasound-Triggered BSA/SPION Hybrid Nanoclusters for Liver-Specific Magnetic Resonance Imaging. ACS Appl. Mater. Interfaces 2012, 4, 6479-6486.

6.

Miao, Z.-H.; Wang, H.; Yang, H.; Li, Z.-L.; Zhen, L.; Xu, C.-Y. Intrinsically Mn2+Chelated Polydopamine Nanoparticles for Simultaneous Magnetic Resonance Imaging and Photothermal Ablation of Cancer Cells. ACS Appl. Mater. Interfaces 2015, 7, 16946-16952.

7.

Liu, H.; Wang, H.; Xu, Y.; Guo, R.; Wen, S.; Huang, Y.; Liu, W.; Shen, M.; Zhao, J.; Zhang, G.; Shi, X. Lactobionic Acid-Modified Dendrimer-Entrapped Gold Nanoparticles for Targeted Computed Tomography Imaging of Human Hepatocellular Carcinoma. ACS Appl. Mater. Interfaces 2014, 6, 6944-6953.

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

8.

Page 20 of 25

Guo, J.; Hong, H.; Chen, G.; Shi, S.; Nayak, T. R.; Theuer, C. P.; Barnhart, T. E.; Cai, W.; Gong, S. Theranostic Unimolecular Micelles Based on Brush-Shaped Amphiphilic Block Copolymers for Tumor-Targeted Drug Delivery and Positron Emission Tomography Imaging. ACS Appl. Mater. Interfaces 2014, 6, 21769-21779.

9.

Kunjachan, S.; Ehling, J.; Storm, G.; Kiessling, F.; Lammers, T. Noninvasive Imaging of Nanomedicines and Nanotheranostics: Principles, Progress, and Prospects. Chem. Rev. 2015, 115, 10907-10937.

10. Bao, G.; Mitragotri, S.; Tong, S. Multifunctional Nanoparticles for Drug Delivery and Molecular Imaging. Annu. Rev. Biomed. Eng. 2013, 15, 253-282. 11. de la Zerda, A.; Kircher, M. F.; Jokerst, J. V.; Zavaleta, C. L.; Kempen, P. J.; Mittra, E.; Pitter, K.; Huang, R. M.; Campos, C.; Habte, F.; Sinclair, R.; Brennan, C. W.; Mellinghoff, I. K.; Holland, E. C.; Gambhir, S. S. A Brain Tumor Molecular Imaging Strategy using a New Triple-Modality MRI-Photoacoustic-Raman Nanoparticle. Nat. Med. 2012, 18, 829834. 12. Lv, R.; Yang, P.; He, F.; Gai, S.; Li, C.; Dai, Y.; Yang, G.; Lin, J. A Yolk-like Multifunctional Platform for Multimodal Imaging and Synergistic Therapy Triggered by a Single Near-Infrared Light. ACS Nano 2015, 9, 1630-1647. 13. Sreejith, S.; Joseph, J.; Lin, M.; Menon, N. V.; Borah, P.; Ng, H. J.; Loong, Y. X.; Kang, Y.; Yu, S. W.-K.; Zhao, Y. Near-Infrared Squaraine Dye Encapsulated Micelles for in Vivo Fluorescence and Photoacoustic Bimodal Imaging. ACS Nano 2015, 9, 5695-5704. 14. Cheng, L.; Yang, K.; Li, Y.; Chen, J.; Wang, C.; Shao, M.; Lee, S.-T.; Liu, Z. Facile Preparation of Multifunctional Upconversion Nanoprobes for Multimodal Imaging and Dual-Targeted Photothermal Therapy. Angew. Chem. Int. Ed. 2011, 50, 7385-7390.

ACS Paragon Plus Environment

20

Page 21 of 25

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

15. Wang, Y.; Song, S.; Liu, J.; Liu, D.; Zhang, H. ZnO-Functionalized Upconverting Nanotheranostic Agent: Multi-Modality Imaging-Guided Chemotherapy with On-Demand Drug Release Triggered by pH. Angew. Chem. Int. Ed. 2015, 54, 536-540. 16. Elsabahy, M.; Heo, G. S.; Lim, S.-M.; Sun, G.; Wooley, K. L. Polymeric Nanostructures for Imaging and Therapy. Chem. Rev. 2015, 115, 10967-11011. 17. Li, K.; Liu, B. Polymer-Encapsulated Organic Nanoparticles for Fluorescence and Photoacoustic Imaging. Chem. Soc. Rev. 2014, 43, 6570-6597. 18. Cheng, L.; He, W. W.; Gong, H.; Wang, C.; Chen, Q.; Cheng, Z. P.; Liu, Z. PEGylated Micelle Nanoparticles Encapsulating a Non-Fluorescent Near-Infrared Organic Dye as a Safe and Highly-Effective Photothermal Agent for In Vivo Cancer Therapy. Adv. Funct. Mater. 2013, 23, 5893-5902. 19. Yan, L.; Wu, W.; Zhao, W.; Qi, R.; Cui, D.; Xie, Z.; Huang, Y.; Tong, T.; Jing, X. Reduction-Sensitive

Core-Cross-Linked

mPEG–Poly(ester-carbonate)

Micelles

for

Glutathione-Triggered Intracellular Drug Release. Polym. Chem. 2012, 3, 2403-2412. 20. Luo, S.; Tan, X.; Qi, Q.; Guo, Q.; Ran, X.; Zhang, L.; Zhang, E.; Liang, Y.; Weng, L.; Zheng, H.; Cheng, T.; Su, Y.; Shi, C. A Multifunctional Heptamethine Near-Infrared Dye for Cancer Theranosis. Biomaterials 2013, 34, 2244-2251. 21. Chen, Q.; Liang, C.; Wang, X.; He, J.; Li, Y.; Liu, Z. An Albumin-Based Theranostic Nano-Agent for Dual-Modal Imaging Guided Photothermal Therapy to Inhibit Lymphatic Metastasis of Cancer Post Surgery. Biomaterials 2014, 35, 9355-9362. 22. Schaafsma, B. E.; Mieog, J. S.; Hutteman, M.; van der Vorst, J. R.; Kuppen, P. J.; Lowik, C. W.; Frangioni, J. V.; van de Velde, C. J.; Vahrmeijer, A. L. The Clinical Use of

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 25

Indocyanine Green as a Near-Infrared Fluorescent Contrast Agent for Image-Guided Oncologic Surgery. J. Surg. Oncol. 2011, 104, 323-332. 23. Namikawa, T.; Sato, T.; Hanazaki, K. Recent Advances in Near-Infrared FluorescenceGuided Imaging Surgery Using Indocyanine Green. Surg. Today 2015, 45, 1467-1474. 24. Schols, R. M.; Connell, N. J.; Stassen, L. P. Near-Infrared Fluorescence Imaging For RealTime Intraoperative Anatomical Guidance in Minimally Invasive Surgery: a Systematic Review of The Literature. World J. Surg. 2015, 39, 1069-1079. 25. Chen, Q.; Wang, C.; Zhan, Z.; He, W.; Cheng, Z.; Li, Y.; Liu, Z. Near-Infrared Dye Bound Albumin with Separated Imaging and Therapy Wavelength Channels for Imaging-Guided Photothermal Therapy. Biomaterials 2014, 35, 8206-8214. 26. Tan, X.; Luo, S.; Wang, D.; Su, Y.; Cheng, T.; Shi, C. A NIR Heptamethine Dye with Intrinsic Cancer Targeting, Tmaging and Photosensitizing Properties. Biomaterials 2012, 33, 2230-2239. 27. Sheng, Z.; Hu, D.; Zheng, M.; Zhao, P.; Liu, H.; Gao, D.; Gong, P.; Gao, G.; Zhang, P.; Ma, Y.; Cai, L. Smart Human Serum Albumin-Indocyanine Green Nanoparticles Generated by Programmed Assembly for Dual-Modal Imaging-Guided Cancer Synergistic Phototherapy. ACS Nano 2014, 8, 12310-12322. 28. Lin, W.; Sun, T.; Xie, Z.; Gu, J.; Jing, X. A Dual-Responsive Nanocapsule via DisulfideInduced Self-Assembly for Therapeutic Agent Delivery. Chem. Sci. 2016, 7, 1846-1852. 29. Cai, K.; He, X.; Song, Z.; Yin, Q.; Zhang, Y.; Uckun, F. M.; Jiang, C.; Cheng, J. Dimeric Drug Polymeric Nanoparticles with Exceptionally High Drug Loading and Quantitative Loading Efficiency. J. Am. Chem. Soc. 2015, 137, 3458-3461.

ACS Paragon Plus Environment

22

Page 23 of 25

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

30. Kreye, O.; Toth, T.; Meier, M. A. Introducing Multicomponent Reactions to Polymer Science: Passerini Reactions of Renewable Monomers. J. Am. Chem. Soc. 2011, 133, 17901792. 31. Li, L.; Lv, A.; Deng, X. X.; Du, F. S.; Li, Z. C. Facile Synthesis of Photo-Cleavable Polymers via Passerini Reaction. Chem. Commun. 2013, 49, 8549-8551. 32. Lin, W.; Guan, X.; Sun, T.; Huang, Y.; Jing, X.; Xie, Z. Reduction-Sensitive Amphiphilic Copolymers Made via Multi-Component Passerini Reaction for Drug Delivery. Colloids Surf., B 2015, 126, 217-223. 33. Lin, W.; Sun, T.; Zheng, M.; Xie, Z.; Huang, Y.; Jing, X. Synthesis of Cross-Linked Polymers via Multi-Component Passerini Reaction and Their Application as Efficient Photocatalysts. RSC Adv. 2014, 4, 25114-25117. 34. Sun, T.; Qi, J.; Zheng, M.; Xie, Z.; Wang, Z.; Jing, X. Thiadiazole Molecules and Poly(ethylene glycol)-Block-Polylactide Self-Assembled Nanoparticles as Effective Photothermal Agents. Colloids Surf., B 2015, 136, 201-206. 35. Ren, C.; Wang, H.; Mao, D.; Zhang, X.; Fengzhao, Q.; Shi, Y.; Ding, D.; Kong, D.; Wang, L.; Yang, Z. When Molecular Probes Meet Self-Assembly: An Enhanced Quenching Effect. Angew. Chem. Int. Ed. 2015, 54, 4823-4827. 36. Song, W.; Tang, Z.; Zhang, D.; Burton, N.; Driessen, W.; Chen, X. Comprehensive Studies of Pharmacokinetics and Biodistribution of Indocyanine Green and Liposomal Lndocyanine Green by Multispectral Optoacoustic Tomography. RSC Adv. 2015, 5, 3807-3813. 37. Fang, J.; Nakamura, H.; Maeda, H. The EPR effect: Unique Features of Tumor Blood Vessels for Drug Delivery, Factors Involved, and Limitations and Augmentation of the Effect. Adv. Drug Delivery Rev. 2011, 63, 136-151.

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 25

38. Vahrmeijer, A. L.; Hutteman, M.; van der Vorst, J. R.; van de Velde, C. J.; Frangioni, J. V. Image-Guided Cancer Surgery Using Near-Infrared Fluorescence. Nat. Rev. Clin. Oncol. 2013, 10, 507-518. 39. Chu, K. F.; Dupuy, D. E. Thermal Ablation of Tumours: Biological Mechanisms and Advances in Therapy. Nat. Rev. Cancer 2014, 14, 199-208. 40. Overgaard, J.; Suit, H. D. Time-Temperature Relationship in Hyperthermic Treatment of Malignant and Normal Tissue in Vivo. Cancer Res. 1979, 39, 3248-3253. 41. Thomsen, S. Pathologic Analysis of Photohermal and Photomechanical Effects of Laser– Tissue Interactions. Photochem. Photobiol. 1991, 53, 825-835. 42. Liu, Y.; Ai, K.; Liu, J.; Deng, M.; He, Y.; Lu, L. Dopamine-Melanin Colloidal Nanospheres: An Efficient Near-Infrared Photothermal Therapeutic Agent for In Vivo Cancer Therapy. Adv. Mater. 2013, 25, 1353-1359.

ACS Paragon Plus Environment

24

Page 25 of 25

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

TOC

ACS Paragon Plus Environment

25