Polydopamine Nanoparticles Enhances Drug Release for Combined

5 hours ago - Support. Get Help · For Advertisers · Institutional Sales; Live Chat. Partners. Atypon · CHORUS · COPE · COUNTER · CrossRef · CrossCheck...
0 downloads 0 Views 3MB Size
Subscriber access provided by Kaohsiung Medical University

Biological and Medical Applications of Materials and Interfaces

Polydopamine Nanoparticles Enhances Drug Release for Combined Photodynamic and Photothermal Therapy Barbara Poinard, Samuel Zhan Yuan Neo, Eugenia Li Ling Yeo, Howard Peng Sin Heng, Koon-Gee Neoh, and James Chen Yong Kah ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.8b04799 • Publication Date (Web): 06 Jun 2018 Downloaded from http://pubs.acs.org on June 6, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a 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 49 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

Polydopamine Nanoparticles Enhances Drug Release for Combined Photodynamic and Photothermal Therapy Barbara Poinard1, Samuel Zhan Yuan Neo2, Eugenia Li Ling Yeo3, Howard Peng Sin Heng3, Koon Gee Neoh1,4, James Chen Yong Kah1,3* 1

NUS Graduate School of Integrative Sciences and Engineering, National University of

Singapore, Singapore 117456 2

School of Life Sciences & Chemical Technology, Ngee Ann Polytechnic, Singapore 599489

3

Department of Biomedical Engineering, National University of Singapore, Singapore 117583

4

Department of Chemical and Biomolecular Engineering, National University of Singapore,

Singapore, 117585

CORRESPONDING AUTHOR *[email protected]

KEYWORDS Polydopamine, photosensitizer, drug delivery, photodynamic therapy, photothermal therapy

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 49

ABSTRACT

Our study shows a facile two-step method which does not require the use of core templates to load a hydrophobic photosensitizer drug Chlorin e6 (Ce6) within polydopamine (PDA) nanoparticles (NPs) while maintaining the intrinsic surface properties of PDA NPs. This structure is significantly different from hollow nanocapsules which are less stiff as they do not possess a core. To our knowledge there exist no similar studies in the literature on drug loading within the polymer matrix of PDA NPs. We characterized the drug loading and release behavior of a photosensitizer Chlorin e6 (Ce6) and demonstrated therapeutic efficacy of combined photodynamic (PDT) and photothermal therapy (PTT) from Ce6 and PDA respectively, under a single wavelength 665 nm irradiation on bladder cancer cells. We obtained saturated loading amount of 14.2 ± 0.85 µM Ce6 in 1 nM PDA NPs by incubating 1 mg/mL dopamine solution with 140 µM of Ce6 for 20 h. The PDA NPs maintained colloidal stability in biological media while the pi-pi (π-π) interaction between PDA and Ce6 enabled a release profile of the photosensitizer until Day 5. Interestingly, loading of Ce6 in polymer matrix of PDA NPs significantly enhanced the cell uptake due to endocytosis. Increased cell kill was observed with combined PDT+PTT from 1 nM PDA-Ce6 compared to PTT alone with 1 nM PDA and PDT alone with 15 µM equivalent concentration of free Ce6. PDA-Ce6 NPs could be a promising PDT/PTT therapeutic agent for cancer therapy.

ACS Paragon Plus Environment

2

Page 3 of 49 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

INTRODUCTION Multitudes of nanomaterials have been developed as nanocarriers for drug delivery such as liposomes, polymers, organic and inorganic nanoparticles (NPs). While many of these nanocarriers have demonstrated the loading of photosensitizer drugs

1-3

, polydopamine (PDA)

maintained its uniqueness over other types of nanocarriers in its ability not just to encapsulate and load drugs, but also in possessing photothermal properties, while being a nature-inspired coating. PDA is the oxidative self-polymerized form of the biomolecule dopamine in alkaline conditions 4, and dopamine was derived from early observations of the adhesiveness of invertebrate mussels to surfaces in wet marine conditions

5-6

, due to their catechol-containing

compound 3,4-dihydroxyphenyl-L-alanine (DOPA) and lysine located in their foot threads 7-8.

PDA was therefore originally developed as a surface modification that relied on similar catecholamine functional groups 5 to provide the adhesiveness to form organic nanometer-scale thin films with low toxicity surface

12

9-10

, and high chemical and thermal stability

11

on practically any

. Surface coating with PDA confers hydrophilicity and zwitterionic property with its

isoelectric pH of 4.5

11, 13

that could potentially facilitate mucopenetration of PDA-coated

surfaces at physiological pH for mucosal delivery of hydrophobic payloads, which would otherwise interact with hydrophobic and negatively charged moieties in mucus to limit their delivery. Furthermore, it is well established that the surface hydrophobicity of nanoparticles dictates immune recognition by macrophages, hence the importance of designing hydrophilic PDA-coating to reduce undesirable immunostimulation 14.

It was only recently in 2009 that PDA was developed in nanoparticulate form chemical and structural similarity to naturally occurring black melanin particles

17

15-16

, with

found in

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 49

human skin. PDA NPs thus possess excellent biocompatibility 18-19, aqueous solubility and strong optical absorption to enable photothermal property

11

, particularly in the near infrared (NIR)

region for photothermal therapy (PTT) in vivo 20-21.

The combination of photothermal properties and versatile surface chemistry have therefore motivated the conjugation of photosensitizers on the surface of PDA NPs to enable combined PTT and photodynamic therapy (PDT) under dual laser irradiation 22, which has been demonstrated to be more efficacious than individual therapy 23-25. Other have made use of coreshell structures with gold 26 or carbon core 27 and a PDA shell modified to bind photosensitizers via electrostatic and π-π stacking on its surface. Advantages of such surface loading approaches lie in facile control of exact drug amount loaded on the NP surface. However, not only were the capacities for surface loading limited, the surface properties of PDA were also lost with the bound drugs. To retain the unique surface properties of PDA without surface blocking by drugs, others have developed PDA nanocapsules for loading small molecule drugs inside the capsular structure 28-29

. Such complex preparation involved hazardous etching reagents to create the hollow interior

and the resulting PDA capsules were fragile and prone to folding, distortion or collapse, after removal of the templates or during subsequent treatment processes due to their reduced mechanical rigidity

30

. Furthermore, such weak stiffness impacts cellular uptake by hindering

endocytosis31. Here, we developed a facile two-step method to prepare PDA NPs loaded with a hydrophobic photosensitizer Chlorin e6 (Ce6) in its polymer matrix instead of its surface or in capsular form, while maintaining the hydrophilicity and negative surface charge of PDA. Our protocol generated PDA NPs with a PDA core stiffer than other reported nanocapsules. We

ACS Paragon Plus Environment

4

Page 5 of 49 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

characterized the drug loading and release behavior of Ce6 and subsequently demonstrated improved therapeutic efficacy of PDT and PTT from Ce6 and PDA respectively, under a single wavelength 665 nm irradiation in vitro. There are presently no studies that have loaded a hydrophobic photosensitizer in the polymer matrix of PDA NPs to our knowledge.

MATERIALS AND METHODS Synthesis and characterization of PDA and PDA-Ce6 NPs PDA NPs were synthesized using a previously established protocol 32. Briefly, 1 mg/mL of dopamine hydrochloride (Sigma-Aldrich, Missouri, USA) was dissolved in 10 mM Tris (Sigma-Aldrich, Missouri, USA) at pH 10.5 and allowed to polymerize at room temperature (25 °C) over time. The synthesized PDA NPs were then washed with Tris buffer (pH 10.5) by repeated centrifugation at 16,100 g for 4 min and stored for use. The concentration of PDA NPs was determined from their absorbance at 808 nm (εPDA = 7.3 x 108 M-1 cm-1)21.

Ce6 was dissolved in Tris buffer (pH 10.5) at 0.5 mg/mL to form a 837 µM Ce6 stock solution before being added to as-synthesized PDA colloid formed after 5 h of polymerization at a final Ce6 concentration of 140 µM (Figure 1). The mixture was allowed to incubate for 20 h at 25 °C for the Ce6 to be loaded into the PDA NPs. The Ce6-loaded PDA NPs (PDA-Ce6) were washed thrice after incubation by repeated centrifugation at 16,100 g for 4 min to remove residual unloaded Ce6 molecules.

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 49

Figure 1. Schematic of the preparation of PDA NPs and loading of hydrophobic photosensitizer Ce6 into the polymer matrix of PDA NPs.

The morphology of PDA and PDA-Ce6 NPs was examined under transmission electron microscopy (TEM) (JEM-1220, JEOL ltd., Japan) and their hydrodynamic diameter (DH) and zeta potential (ζ) were measured using a Zetasizer (Nano ZS, Malvern, UK) at 25 °C in different media including Mili-Q water, RPMI, RPMI+10%FBS, human serum and urine to examine their colloidal stability in different commonly used biological media. The size of the particles was quantified with Image J and its macro plugin NS-Size analyzer. The scale of the images pixel to nm ratio was entered and the different particles were delimited and the diameter measured. The hydrophilicity of PDA and PDA-Ce6 NPs was determined from both Rose Bengal (RB) assay for surface hydrophobicity 33-34 and partition coefficient measurements of their LogP

ACS Paragon Plus Environment

6

Page 7 of 49 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

value based on their partition into water and 1-octanol. The concentration of PDA NPs in both water and octanol phase was determined from their absorbance at 808 nm (εPDA = 7.3 x 108 M-1 cm-1) 21 and Log P value was calculated from the equation: Log  =

     

.

Quantification of Ce6 loading and passive release We determined the amount of Ce6 loaded in the PDA NPs by subtracting the amount of unloaded Ce6 remaining in the supernatant from the initial 140 µM Ce6 added to the synthesized PDA NPs, as measured from the fluorescence of Ce6 against its concentration calibration curve. We also confirmed the amount of loaded Ce6 by thermally releasing Ce6 from PDA-Ce6 NPs in a water bath at 100°C over 5 to 60 min

35

. A saturation in released Ce6 was observed after 10

minutes. The heated sample was centrifuged at 16,100 g for 4 min and fluorescence of Ce6 (λEx/λEm = 400/665 nm) in the supernatant was measured using a microplate reader (Tecan, Switzerland) to determine its concentration being released. The passive release of Ce6 from PDA-Ce6 in different pH: 5, 7, and 10.5), and different commonly used biological media (RPMI with 10%FBS, human serum and urine) was also examined over time from 3 h to Day 17 after preparation. At each time point, the sample was centrifuged at 16,100 g for 4 min and fluorescence of the Ce6 in supernatant was measured to determine the concentration of passively released Ce6 over time against a calibration curve specific to the pH or media.

Photothermal heating and ROS production by PDA-Ce6 in buffer We irradiated 200 µL of 1 nM PDA and PDA-Ce6 in 10 mM of Tris buffer in a 96-well plate with a 665 nm continuous wave (CW) laser (Photonitech, Singapore) for 15 min at 250

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 49

mW/cm2. The light dose was delivered directly above the plate via an optical fiber, with a laser spot size of 12.57 mm2, giving a total fluence of 28.3 J. The temperature of the colloid was monitored by a thermocouple microprobe (Fluke, USA). Tris buffer alone served as a control. The same PDA-Ce6 was prepared by adding varying initial Ce6 concentration (0, 28, 140, 280 µM) to synthesized PDA NPs, and irradiated under the same conditions as described above. The ROS generated from the loaded Ce6 in PDA-Ce6 NPs was measured based on the fluorescence of a ROS indicator dye 30-(p-aminophenyl) fluorescein (APF) (λex/λem = 490/515 nm) (Invitrogen, USA). Briefly, 0.8 µL of 5 mM APF was added to 200 µL of 1 nM PDA-Ce6 before irradiation, and the supernatant of PDA-Ce6 after irradiation was then measured for its APF fluorescence (Safire2, Tecan, Switzerland). To compare if the ROS generation from Ce6 was affected by their loading in PDA NPs, the PDA-Ce6 NPs were heated at 100°C for 10 min to thermally release the loaded Ce6, which was subsequently irradiated to determine its ROS generation. The amount of ROS generation from free Ce6 at the same concentration as that released from thermal treatment was used as our control. Similarly, the effect of pH in ROS generation was examined by irradiating 1 nM of PDA-Ce6 in three different pH conditions (pH 5, 7, 10.5) and measuring the amount of ROS generation as described above.

Cellular uptake of PDA-Ce6 Human urothelial carcinoma T24 cells (ATCC, Manassas, U.S.A.) were cultured in RPMI-1640, supplemented with 10% fetal bovine serum (FBS), 1 mM L-glutamine and 1% penicillin. The culture was maintained at 37 °C in a humidified 5% CO2 atmosphere. The T24 cells were seeded at 5 x 105 cells per well in 6-well culture plates for 24 h before 1 mL of PDA-Ce6 in culture

ACS Paragon Plus Environment

8

Page 9 of 49 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

media was then added to each well at varying final concentrations of 0.10, 0.25, 0.50, 0.75 or 1 nM with 1mL of 15 µM free Ce6 concentration equivalent to that loaded in 1 nM of PDA as a control, and incubated over time from 2 to 24 h at 37 °C. The cellular uptake of PDA-Ce6 in T24 was examined by flow cytometry and absorbance measurement of remaining PDA NPs recovered from cell culture supernatant after incubation. The supernatant in each well was removed after incubation and the concentration of PDA-Ce6 and free Ce6 were quantified by absorbance measurements at 808 nm (εPDA = 7.3x108 M-1 cm-1) and fluorescence measurements (λex/λem = 404/665 nm), before being subtracted from the initial concentration of PDA-Ce6 or Ce6 added. The concentration of PDA-Ce6 was determined from its absorbance measurements (calculated with εPDA = 7.3x108 M-1 cm-1) measured against its concentration calibration curve. The concentration of free Ce6 uptaken in cells was determined from its fluorescence measured against its concentration calibration curve. For flow cytometry, the cells were rinsed three times in PBS (pH 7.4) after incubation to remove the excess PDA-Ce6, trypsinized and fixed with 4% formaldehyde for 15 min. The Ce6 fluorescence in cells was acquired with a FACSCanto flow cytometer using FACSDiva software (Becton, Dickinson, USA). The data were then analyzed based on at least 1.5x104 cells using FlowJo version 7.2.2 (FlowJo, USA). Qualitative analysis of cell uptake was also performed with fluorescence imaging by seeding 105 cells on a glass cover slip in 24 well plates for 24 h. The T24 cells were dosed with 1 nM of PDA-Ce6 in culture media or 15 µM of free Ce6 and incubated over time from 2 to 24 h at 37 °C. The cells were then fixed with 4% formaldehyde for 15 min after incubation, washed thrice in PBS and stained with DAPI (4',6-diamidino-2-phenylindole) for 5 min before being imaged by a camera (SCMOS PcoEdge, Einst Technology Pte. Ltd., Singapore) mounted on a

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 49

fluorescence microscope (Nikon, Eclipse, Ci) equipped with a 60x oil-immersion objective (N.A., 1.25) and appropriate filters. The Ce6 fluorescence intensities of the fluorescence images showing Ce6 uptake in cells were quantified with image J by converting the images to gray scale and calculating their mean gray intensity level.

Therapeutic efficacy of PDA-Ce6 on bladder cancer cells The cultured T24 cells were seeded at 3 x 104 cells per well in 96-well culture plates for 24 h before 200 µL of PDA-Ce6 and PDA NPs in culture media were added to each well at varying final concentrations of 0.1, 0.5 and 1 nM, and incubated for 24 h at 37 °C. Equivalent concentrations of free Ce6 as that loaded (15 µM) and released (1.5 µM) were also dosed to cells as controls. These two concentrations were chosen for their relevance to 1 nM of PDA-Ce6 with 15µM of free Ce6 being the amount loaded and 1.5 µM being the free Ce6 that could be released in RPMI with 10% FBS. The excess PDA-Ce6 and free Ce6 were removed and cells rinsed three times with PBS (pH 7.4) after incubation before being irradiated by a 665 nm CW laser for 15 min at 250 mW/cm2 to initiate PDT with free Ce6, PTT with PDA NPs and combined PDT+PTT with PDA-Ce6 NPs. Cell viability was measured 24 h post treatment with PrestoBlue® based on its absorbance at 570 nm normalized to 600 nm, and compared to the dark toxicities of free Ce6, PDA and PDA-Ce6 NPs without laser irradiation.

RESULTS AND DISCUSSION Synthesis of PDA NPs and loading of Ce6.

ACS Paragon Plus Environment

10

Page 11 of 49 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

PDA NPs were synthesized from polymerizing its monomer dopamine at high alkaline pH with Tris buffer. We synthesized a range of PDA NPs sizes by varying both the dopamine concentration and the pH of Tris buffer, where the increase in dopamine concentration and lowering of basic pH led to larger sizes (Figure S1, Supporting Information). In all cases, the growth of PDA NPs slowed after 20 h. Della Vecchia et al. attributed the mechanism of polymerization and growth to sequential chain elongation and aggregation steps of dopamine monomers oxidized to quinone monomers which acted as electrophilic acceptors for the existing oligomers

36

. Growth slowed as this process was subsequently hindered as Tris residues

covalently linked on quinone moieties to result in steric hindrance or increased nucleophilicity. A linear increase in PDA NPs concentration was observed over time as they stopped growing (Figure S2, Supporting Information), due to continuous synthesis of new PDA NPs from quinones unhindered by the Tris residues. Successful synthesis of PDA NPs was determined by Fourier Transform Infra-red spectroscopy (FTIR) which showed presence of characteristic primary and secondary amines peaks at 1650 cm-1 and 1500 cm-1 respectively (Figure S3, Supporting Information). PDA NPs were synthesized from 1 mg/mL dopamine in alkaline Tris buffer (pH 10.5) and used for loading Ce6 after 5 h of polymerization. We observed a maximum 14.2 ± 0.85 µM of Ce6 loaded on 1 nM PDA NPs after 20 h incubation when Ce6 was added at an initial concentration of 140 µM (~10% loading of Ce6) (Figure 2H). The loaded amount reached a saturation and did not increase as the initial Ce6 concentration added to 1 nM PDA NPs was increased further beyond 140 µM. Here, Ce6 possesses aromatic rings similar to PDA, which allowed them to interact and be stably bound in the PDA polymer matrix via π-π stacking

ACS Paragon Plus Environment

11

ACS Applied Materials & Interfaces

interactions. The loading saturation occurred when the limited number of π-π interacting sites in the PDA NPs was reached.

B

6 4 2 0

PDA-Ce6

1000

10

100

PDA

Hydrodynamic Diameter (DH) in nm

pH

F

Rose bengal

1.0

PDA

0.5

PDA-Ce6

0.15 0.10

0.0 -0.5 -1.0

125000 100000

Ce6 (15 uM) PDA-Ce6 PDA

75000

Tris

50000 25000 0 620

640

660

680

700

6

0 Fr

H

Initial [Ce6] in µM Loaded concentration (µM)

G

ee

Wavelength (nm)

Ce

14

28

14

2.

0

450 475 500 525 550 575 600

8

-1.5

0.05

4

0.20

1.

Absorbance (a.u)

0.25

Log P values

E

.5

PDA-Ce6

10 8

40 30 20 10 0 -10 -20 -30 -40 -50

3

Frequency

PDA

7

D

16 14 12

Surface charge (mV)

C

5

A

Fluorescence intensity (a.u)

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 49

15

10

5

0 1

10

100

1000

Initial [Ce6] (µM)

Wavelength (nm)

ACS Paragon Plus Environment

12

Page 13 of 49 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. Characterization of PDA and PDA-Ce6 NPs with TEM images showing (A) PDA NPs without Ce6 and (B) PDA-Ce6 NPs. (C) Hydrodynamic diameter (DH) histogram distribution profile of PDA NPs (black) and PDA-Ce6 (red). (D) Zeta Potential of PDA NPs with (red) and without (black) Ce6 loaded under different pH conditions. (E) Characterization of the hydrophilicity and hydrophobicity using Rose Bengal. PDA NPs with (red) and without (black) Ce6 were re-suspended in rose Bengal solution for 2 h. The colloidal suspension was then centrifuged and the absorbance of the supernatant measured. The absorbance of the supernatant without any PDA NPs is shown as a reference (blue). (F) The partition coefficient of the PDA NPs incubated with varying concentrations of Ce6 to form PDA-Ce6 NPs. (G) Fluorescence emission spectra after excitation at 404 nm of 1 nM PDA NPs (black); 1 nM PDA NPs loaded with Ce6 (Blue); and 15 µM free Ce6 (red) at the same concentration as the amount of Ce6 loaded onto the PDA-Ce6. (H) Loading capacity of 1 nM of PDA in presence of varying initial Ce6 concentrations after 20 h incubation.

The prepared PDA and PDA-Ce6 NPs were monodispersed with an average size of 141.3 ± 4.3 nm and 142.4 ± 4.1 nm respectively. No significant change in size of NPs was observed with Ce6 loading (Figure 2A and 2B, respectively). However, the hydrodynamic diameter (DH) of unloaded PDA NPs (244.2 ± 10.1 nm) was slightly larger than that of PDA-Ce6 NPs (200.1 ± 8.5 nm) (Figure 2C), while the polydispersity index (PDI) of both PDA NPs and PDA-Ce6 NPs was in the range of 0.2 which is generally accepted as a proof of polymers homogeneity and monodispersity. The larger value obtained from DLS measurements over TEM was likely attributed to the swelling of PDA NPs as they hydrated in aqueous solution due to their hydrophilicity, whereas the hydrophobic Ce6 in PDA-Ce6 NPs minimized the hydration to result

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 49

in less swelling and smaller increase in DH. Nonetheless, the DH of PDA and PDA-Ce6 generally agreed well with the size from TEM images with differences attributed to the swelling of hydrophilic PDA in solution during DLS measurement and shrinkage when the same samples were dried for TEM. Such an observation was common amongst polymers 37-38. Both PDA and PDA-Ce6 NPs were negatively charged in Tris buffer at pH 10.5 with a zeta potential of -48.7 ± 1.3 mV and -45.45 ±1.4 mV respectively due to the deprotonation of phenolic and amino groups on the PDA (Figure 2D). The similarity in their surface potential also supported the loading of Ce6 within the polymer matrix via π-π stacking instead of surface loading. In both cases, the zeta potential became less negative as the pH decreased towards the zwitterionic isoelectric pH of 4.5 for PDA, before flipping to a positive surface charge of 35.3 ± 2.4 mV and 31.2 ± 2.0 mV for PDA and PDA-Ce6 NPs respectively as the pH crossed the isoelectric pH to pH 3. This confirmed the zwitterionic surface property of PDA NPs that was not lost with Ce6 loading in the polymer matrix. Apart from surface charge, the hydrophilicity of PDA was also critical to their biological response. We determined this using the RB assay for surface hydrophobicity where the negatively charged hydrophobic RB dye molecule would interact with a hydrophobic NP and be pelleted to result in reduced absorbance of the dye in the supernatant. Here, electrostatic interaction between the negatively charged RB dye and PDA NPs was ruled out. The RB assay showed similar peak absorbance at 542 nm between the supernatant of PDA, PDA-Ce6 and equivalent concentration of RB solution (Figure 2E), suggesting minimal hydrophobic interaction and hence hydrophilicity of both PDA and PDA-Ce6 NPs. The hydrophilicity of PDA-Ce6 NPs was further confirmed from their negative LogP value, which became slightly less negative from -1.40 ± 0.16 for PDA NPs to -1.12 ±0.04 when

ACS Paragon Plus Environment

14

Page 15 of 49 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

increasing Ce6 concentration was added to PDA NPs (Figure 2F). The hydrophilicity of PDACe6 also confirmed that Ce6 was loaded within the polymer matrix instead of NP surface since Ce6 was hydrophobic with a LogP of 0.940 ± 0.005. The hydrophilic PDA-Ce6 NPs would decrease the immune recognition by macrophages since hydrophobic NPs are preferentially uptaken by macrophages

14

. The negative surface charge and hydrophilicity of PDA NPs could

also potentially confer them good muco-penetration properties with minimal interactions with the hydrophobic and negatively charged regions of mucins39-42. The PDA NPs became fluorescent after Ce6 loading, although the fluorescence of PDACe6 was significantly lower compared to free Ce6 at the same loaded concentration (Figure 2G). PDA NPs showed negligible fluorescence on their own due to their very low quantum yield (0.2%). They also possessed strong fluorescence quenching from the π-π stacking interaction between the monomeric units in PDA NPs4, 11. As Ce6 was loaded in PDA NPs, the π-π stacking of their aromatic groups with those of PDA could have resulted in the fluorescence quenching of 5.5 times over free Ce6 as measured by the change in fluorescence intensity in Figure 2G.

Colloidal stability of PDA NPs and passive release characteristics of Ce6 The colloidal stability of NPs in biological media is crucial to their biological utility. As the PDA NPs were reconstituted in RPMI alone, they aggregated after 3 h with their DH increased from 259.2 ± 1.8 nm to 885.82± 32.1nm (Figure 3A). Their zeta potential was undetermined due to the severe aggregation. The aggregation of PDA NPs in RPMI alone was not unexpected since RPMI media typically presented a high ionic environment that screened the electrostatic repulsive charges between the NPs. However, the PDA NPs became colloidally

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 49

stable even up to 12 h with a DH of 292.0 ± 6.0 nm in cell culture media when serum proteins were incorporated into RPMI. Such colloidal stability in RMPI with 10% FBS was more significant since the presence of serum proteins in biological media was more physiologically relevant in biological systems. These serum proteins from 10% FBS formed a protein corona 25, 43-44

on the PDA NPs to confer steric stabilization.

While PDA NPs could potentially be used as a drug delivery vector in a wide range of context, urine is one biological media of interest, especially in the context of PDA NPs as a drug delivery vector in intravesical instillation, which is the preferred delivery mode for the treatment of bladder malignancies. In bladder cancer where the tumor is usually localized on the luminal surface of the bladder urothelium, intravesical delivery is preferred over systemic delivery to minimize systemic side effects. Here, drugs could be instilled directly into the bladder via the ureter, and the bladder urothelium with a mucus layer serves as a physical barrier to limit the systemic absorption of drugs, thereby reducing the host toxicities. Since the PDA NPs would be in contact with urine before being uptaken by the bladder cancer cells, we examined their colloidal stability in urine to ensure minimum aggregation. The PDA NPs were also colloidally stable in urine (DH = 296.8 ± 6.7 nm), demonstrating their biological applicability in biological media. The DH of PDA NPs in different media after 12 hours was summarized in Figure 3B.

ACS Paragon Plus Environment

16

Page 17 of 49

A

B 1000

Hydrodynamic diameter (DH in nm)

PDA in different media

3 hours

Hydrodynamic diameter (DH in nm)

Surface Charge (mV)

259.2 ± 1.8

-29.8± 2.1

6 hours

800

12 hours

600

water

24 hours

TRIS buffer

275.4 ± 2.9

-39.9 ± 1.9

400

RPMI

885.8 ± 32.1

N.A

200

RPMI +10% FBS

292.0 ± 6.0

-14.2 ± 1.2

urine

296.8 ± 6.7

-15.9 ± 3.7

e in

FB 0%

R

PM

I+ 1

ur

S

I PM R

Bu ffe r

at

er

0

w

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) Colloidal stability of PDA NPs in different media over time as measured by their hydrodynamic diameter, DH in the different media. (B) Table summarizing the average DH and surface charge of PDA NPs in different media after 12 hours.

With 14.2 µM of Ce6 loaded on 1 nM PDA NPs, we examined their passive release over time in different pH and biological media. At high basic pH of 10.5 in Tris buffer, Ce6 showed a first-order burst release profile in the first 5 days, before the release rate slowed and stopped after 7 days (Figure 4A). The burst release became less pronounced at physiological pH 7, and eventually switched towards a zeroth-order linear release profile as the pH was further reduced to a mild acidic pH 5. The final amount of Ce6 passively released after 10 days also reduced with pH from 3.0 ± 0.13 µM (pH 10.5) to 1.46 ± 0.09 µM (pH 7) and 0.51 ± 0.09 µM (pH 5). The pHdependent release profile of Ce6 could be explained by the zwitterionic property of PDA, which became less negative at lower pH, thus reducing the electrostatic repulsion with the negatively charged Ce6 molecule due to its deprotonation of pendant carboxyl groups. This suggested that pH could be a handle to modulate the release behavior of loaded therapeutic compounds.

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 49

While a good drug release was typically preferred in lower pH for cancer treatment since the tumor microenvironment was known to possess a slightly acidic pH of ~6, the same low release at slightly acidic pH of ~6 could be beneficial in developing PDA NPs as a drug delivery vector in intravesical instillation for treatment of bladder malignancies. In this context, the release of Ce6 needs to be minimized not just in the slightly acidic urine during instillation, but also during mucopenetration through the acidic bladder mucus layer, before reaching the acidic tumor microenvironment, and entering cancer cells through the acidic endosomes. Until here, minimum release of Ce6 would be preferred until the PDA-Ce6 escapes the endosomes and enters the cytoplasm with physiological pH which triggers greater release of Ce6 for PDT in the cytoplasm. Previous studies have demonstrated that PDA-coated NPs could enable the escape from endosomes to the cytosol 21. Regardless of the pH, the amount of Ce6 that could be released from 1 nM PDA NPs passively over time still represented a low percentage (2.67% to 20%) of the 14.2 ± 0.85 µM Ce6 loaded (Figure 2H). It appeared that PDA NPs did not release all their Ce6 payload passively overtime with a significant Ce6 concentration which remained trapped in the PDA polymer matrix possibly due to the strong π-π stacking loading mechanism as discussed earlier.

ACS Paragon Plus Environment

18

Page 19 of 49

pH 5 pH 7 pH 10.5

3

25 20 15

2

10 1

5

0

0 2

4

6

8

RPMI + 10 % FBS Human Serum

2.0 1.5

10

1.0 5 0.5

0

2

4

6

0.8

4

2 0.2 1 0.0

Ce6 released (%)

3

0.4

8

0 10 12 14 16 18

Time (days)

D

0.6

15

urine

0.0

Time (days)

C

2.5

10

[Ce6] released in (µM)

0

[Ce6] released in urine (µM)

[Ce6] released (µM)

B

4

Room T°

37°

45°

0.6

0.4

0.2

0 0

4

8

12

16

20

24

0

10

20

30

40

50

60

Time (min)

Time (hours)

Figure 4. Passive release of Ce6 from 1 nM of PDA-Ce6. (A) Release of Ce6 from PDA-Ce6 over 10 days in Tris buffer of different pH. (B) Release of Ce6 from PDA-Ce6 NPs in RPMI with 10%FBS, human serum, and urine over a period of 2 weeks. In all cases, PDA-Ce6 was formed by incubating PDA NPs with 140 µM of Ce6 for 20 h. (C) Burst release profile in urine over a span of 1 day of PDA NPs incubated with 140 µM of Ce6. (D) Amount of Ce6 thermally released after heating 1 nM of PDA-Ce6 at 37 and 45 °C for different duration.

Similar first-order burst release profile of Ce6 was also observed in various biological media for the first 5 days. Here, RPMI with 10% FBS at pH 7.4 resulted in the highest release of Ce6 (1.59 ± 0.12 µM) after 17 days, followed by human serum (1.03 ± 0.06 µM) and urine (0.96 ± 0.04 µM) (Figure 4B). It seemed that the formation of a protein corona around PDA-Ce6 NPs

ACS Paragon Plus Environment

19

Ce6 released (%)

[Ce6] released (µM)

A

Ce6 released (%)

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

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 49

in RPMI with 10% FBS and human serum did not affect the Ce6 release profile or release amount as the amount of Ce6 released was comparable or even higher than that in Tris buffer at pH 7 (Figure 4A). Urine induced the lowest amount of Ce6 released possibly due to its slightly acidic pH of ~6, and a shorter burst release which took place in the first 10 hours (Figure 4C). The photothermal properties of PDA could induce an increase in temperature of the microenvironment surrounding the PDA NPs to >37 °C. Here, we investigated if this heating could be exploited to trigger the release of Ce6 from PDA NPs. We observed significant increase in concentration of Ce6 released over time at higher temperatures of 37 °C (body temperature) and 45 °C (after photothermal heating by laser irradiation, as discussed in next section) compared to 25 °C (room temperature) (Figure 4D). Further increase in temperature to 100 ºC by heating the solution led to an even more significant Ce6 released from PDA-Ce6 (Figure S4, Supporting Information). It appeared that the release of Ce6 from the PDA-Ce6 was temperature dependent, where higher temperature enabled more disruption of the pi-pi stacking interactions between Ce6 and PDA, leading to a more significant increase in Ce6 release. This demonstrated that the photothermal heating of PDA NPs upon laser irradiation could modulate the release of Ce6 by disrupting the π-π stacking bonds. In general, the burst release behavior of Ce6 that we observed in PDA NPs was similar to other drug delivery systems such as CaCO3 and lactoferrin which when loaded with Ce6 exhibited a release profile reaching a plateau at 5 hours45 and 6 hours 46 respectively. This faster saturation compared to PDA NPs (5 days) presents limitations as the nanocarriers would have released most of their payload before reaching the target cells.

Photothermal behavior of PDA and PDA-Ce6 NPs

ACS Paragon Plus Environment

20

Page 21 of 49 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

Being chemically similar to naturally occurring black melanin particles found in human skin, Zhu et al. showed strong optical absorption and photothermal conversion in PDA NPs when irradiated with a 808 nm laser 20, 47. We observed that a different wavelength source of 665 nm used to excite Ce6 at 250 mW/cm2 could also induced a significant increase in the temperature of 1 nM PDA NPs colloid over buffer solution in a pH independent manner after irradiation for 15 min. Here, a significant increase in temperature from 23.9 ± 1.12 °C to 42.1 ± 1.5 °C was observed (Figure 5A). We further demonstrated that the photothermal property of PDA NPs was concentration dependent with an observed saturation at >3 nM (Figure S5A, Supporting Information). There was also no significant difference in the temperature rise between 1 nM of PDA-Ce6 (43.33 ± 0.83 °C) and PDA NPs (42.10 ± 0.47 °C) when irradiated with the same 665 nm laser 15 min (Figure S5B, Supporting Information). This significant temperature rise was largely attributed to the strong optical absorption in the near infra-red (NIR) region, particularly at the 665 nm wavelength of irradiation (Figure 5B), leading to rapid absorption of photons and heat conversion by PDA and PDA-Ce6

48-49

. This enabled a rapid

temperature rise that was otherwise not possible with buffer alone. We also determined the photothermal efficiency of 1 nM PDA and PDA-Ce6 under this irradiation condition to be ~60.4 % (Input energy of 28.3 J and output energy of 17.1 J, see Supporting Information for detailed calculations). Comparatively, gold nanoparticles present a photothermal efficiency which is inversely proportional to their size 50, with smaller sizes of 15 nm range having ~100 % of photothermal efficiency, which decreased gradually to 55 % when the size increased to 100 nm 51. The use of 665 nm laser irradiation to effect the temperature rise not only matched the excitation of Ce6 and potentially allowed simultaneous PDT and PTT, but was also biologically

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 49

advantageous since the wavelength also provided for deeper optical penetration in tissue to reach sub-epithelial tumor tissue

52

. The use of a single 665 nm laser demonstrated significant

therapeutic efficacy with a near complete tumor eradication in in vivo experimental models in our previous study by Yeo et al.53. While the typical absorbance peak of Ce6 at ~665 nm was not clearly observed, this was not due to its low loading capacity of Ce6 since we have earlier determined that 15 µM of Ce6 was loaded on 1 nM PDA, giving a ratio of ~15,000 Ce6 molecules loaded on each PDA NP. The absence of a strong Ce6 absorbance peak at 665 nm in the absorption spectrum of PDA-Ce6 was likely a combination of Ce6 molecules loaded inside the PDA polymer matrix, coupled with the PDA NPs by themselves having a strong absorbance spectrum across the entire visible wavelengths (Figure 5B), which masked the absorbance of Ce6 in the PDA. This strong absorbance of PDA in itself was not surprising, hence its use as an effective photothermal agent.

ACS Paragon Plus Environment

22

Page 23 of 49

A

50

Dark

B

Irradiated

Absorbance (a.u)

T° (celsius)

40 30 20 10

1.5

0.6 0.4 0.2

500

600

10

2.25

2000 1000 0.50

60000 ROS generation Fluorescence level (a.u)

Free Ce6 (Dark) Free Ce6 (Irr) Dark/ no heat Irr/ no heat Dark /heated Irr/Heat

0.25

0.75

700

800

900

D 3

3000

0 0.00

0.8

pH 0.75

Tris buffer 15 µM Ce6

PDA PDA-Ce6

Wavelength (nm)

Free Ce6 (µM)

C 50000 40000 30000 20000 10000

1.0

0.0 400

.5

7 pH

5 pH

3 pH

Tr is

bu ffe r

0

ROS generation Fluorescence level (a.u)

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

PDA Dark PDA Irr

PDA-Ce6 Dark/No Heat PDA-Ce6 Irr/No Heat

40000

PDA-Ce6 Dark/Heat PDA-Ce6 Irr/Heat

20000 2000 1500 1000 500 0

1.00

[PDA-Ce6] (nM)

4

5

6

7

8

9

10

pH

Figure 5. Photothermal and photodynamic properties of PDA and PDA-Ce6 NPs. (A) Absolute temperature obtained after irradiating 1 nM PDA NPs in different pH conditions and 10mM Tris buffer without any NPs as a control with a 665 nm CW laser at 250 mW/cm² for 15 min. (B) Absorbance spectra of PDA, PDA-Ce6, 15 µM Free Ce6 and Tris buffer without any NPs. ROS generation with (C) increasing PDA-Ce6 concentrations at physiological pH and (D) increasing pH at fixed concentration of 1nM PDA-Ce6. The PDA-Ce6 NPs were either subjected to heat (100 °C for 10 min) (red, dotted) to release the maximal amount of Ce6 for irradiation or unheated to leave the Ce6 loaded within the PDA NPs during irradiation (blue, dotted). Free Ce6 at equivalent concentration as that released from the heating was included as a comparison

ACS Paragon Plus Environment

23

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 24 of 49

(green, dotted). The samples were either kept in the dark (solid lines) or irradiated with a 665 nm CW laser 250 mW/cm² for 15 min (dotted lines).

The results also suggested that the photothermal heating of PDA-Ce6 was not affected by the loaded Ce6 since there was no significant difference in the temperature measured between PDA and PDA-Ce6. Hence PDA-Ce6 remained effective as a PTT agent even after Ce6 loading. The temperature reached (42.4 ± 0.5°C) was within the range of 41 to 50 °C known to cause hyperthermia and cell kill by thermolysis in cancer cells, which were known to be more susceptible to heat than the more heat-tolerant healthy cells 54-55.

Reactive oxygen species (ROS) production by PDT with PDA-Ce6 Most photosensitizers such as Ce6 were hydrophobic, which posed not just aqueous solubility issues, but also poor delivery in biological barriers such as the mucus. Here, PDA NPs provided a mean to load and deliver hydrophobic Ce6 in a highly aqueous and hydrophilic environment to target tissue for PDT. When irradiated by a laser, Ce6 generated ROS which were capable of eliciting cancer cell kill by oxidative stress. We treated PDA-Ce6 thermally by heating them at in a water bath at 100 °C to release a maximum of 2.8 ± 0.06 µM Ce6 from 1 nM PDA-Ce6 after heating for 10 min (Figure S4, Supporting Information) as π-π stacking between aromatic cycles of Ce6 and PDA molecules was non-covalent and could be disrupted by heating 35

. This amount of thermally released Ce6 from 1 nM PDA-Ce6 NPs was comparable to the

maximum amount that was passively released over time (Figure 4A). The thermally released Ce6 was irradiated and the level of ROS generated was comparable to that from the same

ACS Paragon Plus Environment

24

Page 25 of 49 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

concentration of free Ce6 (Figure 5C, green and red dotted lines), suggesting that the Ce6 molecules that were originally loaded did not lose their photodynamic properties with loading. In the absence of thermal treatment to leave the Ce6 loaded within the PDA NPs, we also observed an increased level of ROS generation from 1 nM PDA-Ce6 after irradiation by a 665 nm CW laser at 250 mW/cm² for 15 min compared to non-irradiated PDA-Ce6 in the dark (Figure 5C, blue lines). This was due to the excitation of Ce6 as the level of ROS generated increased with higher concentration of Ce6 loaded on PDA NPs that could be effectively released from the PDA NPs. However, the amount of ROS generated was much lower when Ce6 was loaded in the PDA NPs (fluorescence intensity = 2684.3 ±136.9, blue dotted) than when the same equivalent concentration of Ce6 was thermally released (fluorescence intensity = 39236 ±954.9, red dotted). The Ce6 loaded in the PDA polymer matrix generated a much lower level of ROS compared to the amount of Ce6 that was thermally released, even though the amount of Ce6 loaded (14.2 ± 0.85 µM) was ~5 times higher than that released thermally (2.8 ± 0.06 µM). This was likely due to a combination of strong optical absorption of irradiated light by PDA that reduced the excitation of Ce6 loaded in the polymer matrix and the reduced oxygenation in the polymer matrix needed for ROS generation. Nonetheless, our observations not only further proved that Ce6 was loaded within the PDA and not on its surface, but also suggested that the passive release of Ce6 over time could support fractionated PDT at chosen time points with a single PDA-Ce6 dosing. We also characterized the ROS generation from PDA and PDA-Ce6 NPs in different pH, with either prior thermal heating or no thermal heating to release the Ce6. PDA did not generate any ROS regardless of irradiation (Figure 5D, black lines). As observed earlier, the absence of

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 49

Ce6 released from PDA NPs and non-irradiation of the samples resulted in low level of ROS generation (Figure 5D, solid lines), while irradiated samples with Ce6 thermally released from PDA-Ce6 (Figure 5D, red dotted) resulted in much higher ROS generation compared to PDACe6 with no Ce6 released (Figure 5D, blue dotted). More importantly, the ROS generation from PDA-Ce6 increased with pH increased. This increase in ROS generation with pH correlated well with the increased in Ce6 released with pH as previously shown in figure 4A.

Cell uptake and therapeutic efficacy The uptake of Ce6 in T24 bladder cancer cells with 1 nM PDA-Ce6 carrying 15 µM of Ce6 was significantly higher than the uptake of an equivalent 15 µM free Ce6 (Figure 6C). By using two different quantitative techniques of flow cytometry and subtraction of absorbance measurement of non-uptaken PDA-Ce6 NPs recovered from cell culture supernatant from the initial concentration of PDA-Ce6 added, we showed that the uptake of PDA-Ce6 into T24 cells were not just dose-dependent, but also increased over time up to a maximum after 24 h of dosing (Figure 6A and 6B). Here, the fluorescence of Ce6 was used to determine its concentration uptaken by the cells instead of its absorbance as the Ce6 absorbance would be masked by the absorbance of PDA. This was observed in Figure 5B with the disappearance of Ce6 characteristic peak at 665 nm on loading in the PDA-Ce6. Such dose- and time-dependent uptake was not observed with free Ce6 at the same equivalent concentration, thus demonstrating the merit of exploiting PDA NPs for Ce6 delivery into cells (Figure 6C). For the same Ce6 concentration of 15 µM, 35.7% were uptaken by T24 bladder cancer cells when loaded in PDA NPs compared to 4.3% dosed in its free form.

ACS Paragon Plus Environment

26

Page 27 of 49

B [PDA-Ce6] uptaken (nM)

0.4

Fluorescence intensity (a.u)

A 2h 4h 6h

0.3

24 h

0.2 0.1 0.0 0.00

0.25

0.50

0.75

1.00

1800 1600 1400 1200 1000 800 600 400 200 0 0.00

2h 4h 6h 24 h

0.25

0.50

0.75

1.00

[PDA-Ce6] added (nM)

[PDA-Ce6] added (nM)

C [Ce6] uptaken (µM)

6.0

40

Free Ce6 PDA-Ce6

4.5

30

3.0

20

1.5

10

0.0 0

5

10

15

20

% Ce6 uptake

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

0 25

Incubation time (hours)

Figure 6. Cellular uptake of PDA-Ce6 NPs over a range of PDA-Ce6 concentration dosed to T24 cells and incubation time as quantified by (A) subtracting the absorbance measurement of nonuptaken PDA-Ce6 NPs recovered from cell culture supernatant from the initial absorbance of PDA-Ce6 added to cells, and (B) by measuring the fluorescence intensity of Ce6 within cells using flow cytometry. (C) Comparison of cellular uptake of 15 µM of free Ce6 and 15 µM of Ce6 loaded in 1 nM of PDA-Ce6 in terms of Ce6 concentration uptaken and uptake percentage in T24 cells over the concentration dosed for varying incubation times as quantified via the supernatant loss absorbance measurement technique in (A).

We confirmed the improved Ce6 uptake in cells with PDA-NPs from fluorescence microscopy images of Ce6 uptake over time. With increasing incubation time of 15 µM free Ce6

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 49

(equivalent amount loaded in 1nM PDA-Ce6), we observed a peak in uptake of Ce6 after 6 hours with localization mainly confined to the cytoplasm (Figure 7A and 7C). The peak uptake agreed well with the supernatant loss absorbance measurement technique (Figure 6C). In a similar manner, the Ce6 fluorescence in cells dosed with 1 nM of PDA-Ce6 agreed with our absorbance measurement and flow cytometry results, where a peak uptake was observed after 24 h with localization mainly confined to the cytoplasm (Figure 7B and 7C). In comparing the uptake between Ce6 loaded in PDA-NPs and free Ce6 at the same concentration, a much higher Ce6 fluorescence in cells were observed when delivered with 1 nM PDA-Ce6 NPs as compared to delivery of the same Ce6 concentration in its free form (Figure 7) under the same excitation and acquisition parameters. After 24 h incubation, the measured intensity from the quantitative image analysis showed free Ce6 uptake with a mean gray level intensity of 15.7 ±1.2 compared to PDA-Ce6 uptake with a mean gray level intensity of 39.9 ±2.0. Here, a significantly higher uptake of Ce6 in cells (as indicated by higher fluorescence intensity in cells) using PDA-Ce6 compared to the equivalent amount of free Ce6 was already observed despite the quenching of Ce6 by PDA. Hence, the enhancement in Ce6 uptake with PDA-Ce6 would be even more significant if the quenching was corrected for. We determined the quenching effect of PDA on Ce6 fluorescent to be ~5.5 times (Figure 2G). Hence at 24 h, the corrected fluorescence grey intensity level for PDA-Ce6 would be ~220 compared to 15.7 for free Ce6 alone.

ACS Paragon Plus Environment

28

Page 29 of 49 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 7. Fluorescence imaging of the cellular uptake and localization of (A) 15 µM free Ce6 and (B) 1 nM PDA-Ce6 loaded with the same 15 µM Ce6 over a range of incubation time. Ce6 fluorescence was shown in the red channel, while the cell nucleus (DAPI-stained) was shown in the blue channel. Scale bar = 20 µm. (C) Quantification of Ce6 fluorescence intensity of the different fluorescent images using ImageJ.

The 8.3-fold higher Ce6 uptake with PDA NPs as determined from the supernatant loss absorbance measurement technique (Figure 6C) could be explained by the endocytosis internalization pathway. It was demonstrated that stiffer nanoparticles are predominantly uptaken by endocytosis31, whereas small molecules have limited receptor-mediated endocytosis 56-57.

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 49

T24 bladder cancer cells maintained a high cell viability of > 80% when dosed with up to 1 nM PDA and PDA-Ce6 NPs for 24 h (Figure 8, solid lines). Upon irradiation with a 665 nm CW laser at 250 mW/cm2 for 15 min after dosing for 24 h, we observed dose-dependent cell kill with PDA NPs, PDA-Ce6 NPs and free Ce6 at the equivalent concentration loaded (Figure 8, dotted lines). Between the three therapeutic agents, the highest cell kill was observed with combined PTT+PDT from 1 nM PDA-Ce6 (76.0 ± 3.4% cell kill), followed by PDT with 15 µM Ce6 (57.8 ±7.1%) and PTT with 1 nM PDA NPs (47.1 ± 7.4%). This was true not just for the highest concentration of these therapeutic agents, but also across all the concentrations studied (Figure 8, dotted lines). However, at physiological pH, the amount of Ce6 which was effectively released from the 1 nM of PDA-Ce6 particles (i.e. ~1.5 µM) resulted in ~32.6% of cell death. We have shown earlier that ROS generation correlated to the amount of Ce6 released from PDA-Ce6 (Figure 5C), thus Ce6 that remained unreleased in the PDA polymer matrix were unable to generate ROS for PDT. This level of cell death from PDT of released Ce6 (i.e. 32.6%), if added to cell death from PTT by PDA NPs alone (i.e. 47.1%), would result in an additive total of 79.7% cell kill. This similarity in cell kill level observed in combined PTT+PDT with PDA-Ce6 (i.e. 76.0%) suggested an additive effect between PTT alone and PDT alone, which when combined together resulted in an enhanced therapeutic effect over a single treatment.

ACS Paragon Plus Environment

30

Page 31 of 49

Equivalent Free Ce6 (µM) 0

5

10

15

100 Cell viability (%)

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

80 60 40 20 0 0.00

PDA (dark)

PDA (irr)

PDA-Ce6 (dark)

PDA-Ce6 (irr)

Ce6 (dark)

Ce6 (irr)

0.25

0.50

0.75

1.00

[PDA] and [PDA-Ce6] (nM) Figure 8. Cell viability after dosing T24 bladder cancer cells with varying concentrations of PDA NPs, PDA-Ce6 NPs and the equivalent free Ce6 in medium supplemented with serum for 24 h and irradiating them with a 665 nm CW laser at 250 mW/cm2 for 15 min. Using PrestoBlue cell viability assay, the dark toxicity of PDA NPs, PDA-Ce6 NPs and free Ce6 were minimal (high cell viability > 80%). The viability decreased drastically with combined PDT+PTT with 1 nM PDA-Ce6 (dotted red) compared to PTT alone with 1 nM PDA (dotted black) and PDT alone with 15 µM free Ce6 (dotted green).

As discussed above, the enhanced cell kill by PDA-Ce6 was likely attributed to the combination of photothermal therapy by PDA, coupled with ROS generation from loaded Ce6

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 49

for PDT and enhanced uptake of Ce6 by PDA-Ce6, which collectively resulted in enhanced killing of T24 bladder cells. PDT by Ce6 is oxygen-dependent for generating ROS. As the oxygen level in cells and tissues was gradually depleted from PDT over time, its efficacy decreased and the T24 cells became more sensitive to heat

58

. Under such circumstances, the

oxygen-independent PTT by PDA played a complementary role in maintaining a high therapeutic efficacy

59-60

. Therefore, the combination of PTT by PDA and PDT by Ce6 played

significant roles in creating a strong enhancement in therapeutic efficacy by PDA-Ce6 over a single treatment. Such cell kill levels were not observed in the absence of irradiation where T24 cells maintained a high viability of > 80% when dosed with up to 1 nM of PDA and PDA-Ce6 containing an equivalent Ce6 concentration of 15 µM (Figure 8, solid lines). This showed that PDA NPs and PDA-Ce6 NPs possessed low levels of dark toxicity, similar to free Ce6, and was expected since PDA was known to have good biocompatibility with a LD50 of 400–585 mg/kg in vivo in a murine model as determined by Liu et al. 21, 61. While effective cell kill was also demonstrated by Zhang et al., the studies were performed on a different cell line (Human liver cancer HepG2 ) and subjected to a dual laser irradiation by 665 nm and 808 nm lasers operating at much higher laser power intensity (50 mW/cm2 for 665 nm laser and 2 W/cm2 for 808 nm laser) was used in their therapy 22 compared to our single laser operating at a lower power intensity (250 mW/cm2). Furthermore, the Ce6 was covalently conjugated on the surface of PDA NPs, thus altering the desirable hydrophilic and zwitterionic properties of PDA NPs, making them more hydrophobic. The release characteristics of these covalently bound Ce6 were also not characterized. Here, the loading of Ce6 in the PDA polymer matrix preserved the hydrophilicity and zwitterionic properties of PDA NPs and

ACS Paragon Plus Environment

32

Page 33 of 49 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

allowed for a significantly higher payload and gradual release of Ce6 over a few days. Such a drug loading and release behavior afforded by PDA NPs would enable fractionated PDT at different time points, which was not possible with surface loading.

CONCLUSION We have developed a facile two-step method to load a hydrophobic photosensitizer Ce6 within the polymer matrix of a biopolymer PDA NPs which has not been demonstrated to date. Such a drug loading approach removed the need for complex preparation involving hazardous etching reagents to create the hollow interior of nanocapsules, and maintained the mechanical and structural integrity of PDA NPs as well as their intrinsic surface hydrophilicity and zwitterionic properties, which was not possible with surface conjugation of hydrophobic drugs. We also extensively characterized the colloidal stability, drug loading and release behavior of Ce6 over prolonged period of a few days and in different biological media. Such data provided new insights on the characteristics of PDA NPs as a nanocarrier which have been lacking in the literature to date. It appears that the π-π stacking interaction between PDA and Ce6 could be exploited for prolonged drug release. With our approach to loading Ce6 in PDA NPs, we also demonstrated enhanced cell uptake by endocytosis and therapeutic efficacy of combined PDT and PTT from PDA-Ce6 under a single laser irradiation. By maintaining the surface charge and hydrophilicity of PDA NPs, these NPs could avoid uptake by macrophages and potentially be muco-penetrative to enable more effective cancer phototherapy in organs requiring mucosal drug delivery such as bladder, intestinal and nasopharyngeal cavity, which is currently limited by the poor muco-penetration of hydrophobic photosensitizers. While recent advances have suggested PDA as a viable nanomaterial in

ACS Paragon Plus Environment

33

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 34 of 49

nanomedicine applications, many aspects of PDA NPs remained as scientific gaps, including their muco-penetrative properties, immunogenic response and biodegradation behavior, which needed to be addressed before eventual clinical utility. This formed our motivation for subsequent further studies.

ASSOCIATED CONTENT Supporting Information. Additional data on the synthesis and characterization of PDA and PDA-Ce6 as well as the active release of Ce6 triggered by heating PDA-Ce6 NPs are available as Supporting Information.

AUTHOR INFORMATION Corresponding Author *Email: [email protected] Author Contributions The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript. Funding Sources The funding used to support the research of the manuscript was from the Ministry of Education (MOE) AcRF Tier 2 Grant (MOE2014-T2-2-147). B. Poinard would like to acknowledge the scholarship support from the NGS Graduate scholarship.

ACS Paragon Plus Environment

34

Page 35 of 49 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

ACKNOWLEDGMENT The funding used to support the research of the manuscript was from the Ministry of Education (MOE) AcRF Tier 2. B.P. acknowledges scholarship support from NUS Graduate School of Integrative Sciences and Engineering. We also acknowledge Laurent Bekale, Lanry Yung and Giorgia Pastorin for helpful discussions.

ABBREVIATIONS APF, 3’-( p-Aminophenyl) fluorescein; Ce6, Chlorin e6; CW, Continuous wave; DH, hydrodynamic diameter; DLS, Dynamic light Scattering; EPR, Enhanced permeability and retention; FBS, Fetal bovine serum; FTIR, Fourier Transform Infrared Spectroscopy; HS, Human serum; NIR, Near infrared; NP, Nanoparticle; PDA, Polydopamine; PDT, Photodynamic therapy; PBS, Phosphate buffer Saline; PTT, Photothermal therapy; ROS, Reactive oxygen species; TEM, Transition Electron Microscopy

REFERENCES (1) Hu, D.; Chen, L.; Qu, Y.; Peng, J.; Chu, B.; Shi, K.; Hao, Y.; Zhong, L.; Wang, M.; Qian, Z. Oxygen-Generating Hybrid Polymeric Nanoparticles with Encapsulated Doxorubicin and Chlorin e6 for Trimodal Imaging-Guided Combined Chemo-Photodynamic Therapy. Theranostics 2018, 8, 1558-1574. (2) Liu, J.; Yang, G.; Zhu, W.; Dong, Z.; Yang, Y.; Chao, Y.; Liu, Z. Light-Controlled Drug Release From Singlet-Oxygen Sensitive Nanoscale Coordination Polymers Enabling Cancer Combination Therapy. Biomaterials 2017, 146, 40-48. (3) Xia, F.; Hou, W.; Zhang, C.; Zhi, X.; Cheng, J.; de la Fuente, J. M.; Song, J.; Cui, D. pHResponsive Gold Nanoclusters-Based Nanoprobes for Lung Cancer Targeted Near-Infrared Fluorescence Imaging and Chemo-Photodynamic Therapy. Acta Biomater 2018, 68, 308319.

ACS Paragon Plus Environment

35

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 36 of 49

(4) Dreyer, D. R.; Miller, D. J.; Freeman, B. D.; Paul, D. R.; Bielawski, C. W. Elucidating the Structure of Poly(dopamine). Langmuir 2012, 28, 6428-6435. (5) Lee, H.; Dellatore, S. M.; Miller, W. M.; Messersmith, P. B. Mussel-Inspired Surface Chemistry for Multifunctional Coatings. Science 2007, 318, 426-30. (6) Waite, J. H.; Qin, X. Polyphosphoprotein from the Adhesive Pads of Mytilus Edulis. Biochemistry 2001, 40, 2887-2893. (7) Sun, K.; Xie, Y.; Ye, D.; Zhao, Y.; Cui, Y.; Long, F.; Zhang, W.; Jiang, X. Mussel-Inspired Anchoring for Patterning Cells Using Polydopamine. Langmuir 2012, 28, 2131-6. (8) Faure, E.; Falentin-Daudré, C.; Jérôme, C.; Lyskawa, J.; Fournier, D.; Woisel, P.; Detrembleur, C. Catechols as Versatile Platforms in Polymer Chemistry. Progress in Polymer Science 2013, 38, 236-270. (9) Liu, X.; Cao, J.; Li, H.; Li, J.; Jin, Q.; Ren, K.; Ji, J. Mussel-Inspired Polydopamine: a Biocompatible and Ultrastable Coating for Nanoparticles in vivo. ACS Nano 2013, 7, 9384-95. (10) Jeong KJ, W. L., Stefanescu CF, Lawlor MW, Polat J, Dohlman CH, Langer RS, Kohane DS. Polydopamine Coatings Enhance Biointegration of a Model Polymeric Implant. Soft Matter 2011, 7, 8305-12. (11) Liu, Y.; Ai, K.; Lu, L. Polydopamine and its derivative materials: synthesis and promising applications in energy, environmental, and biomedical fields. Chem Rev 2014, 114 (9), 5057-115, DOI: 10.1021/cr400407a. (12) Kord Forooshani, P.; Lee, B. P. Recent Approaches in Designing Bioadhesive Materials Inspired by Mussel Adhesive Protein. Journal of Polymer Science Part A: Polymer Chemistry 2017, 55, 9-33. (13) Liu, Q.; Yu, B.; Ye, W.; Zhou, F. Highly Selective Uptake and Release of Charged Molecules by pH-Responsive Polydopamine Microcapsules. Macromol Biosci 2011, 11, 1227-34. (14) Moyano, D. F.; Goldsmith, M.; Solfiell, D. J.; Landesman-Milo, D.; Miranda, O. R.; Peer, D.; Rotello, V. M. Nanoparticle Hydrophobicity Dictates Immune Response. Journal of the American Chemical Society 2012, 134, 3965-3967. (15) Postma, A.; Yan, Y.; Wang, Y.; Zelikin, A. N.; Tjipto, E.; Caruso, F. Self-Polymerization of Dopamine as a Versatile and Robust Technique to Prepare Polymer Capsules. Chemistry of Materials 2009, 21, 3042-3044. (16) Ochs, C. J.; Hong, T.; Such, G. K.; Cui, J.; Postma, A.; Caruso, F. Dopamine-Mediated Continuous Assembly of Biodegradable Capsules. Chemistry of Materials 2011, 23, 31413143. (17) d'Ischia, M.; Napolitano, A.; Ball, V.; Chen, C. T.; Buehler, M. J. Polydopamine and Eumelanin: from Structure-Property Relationships to a Unified Tailoring Strategy. Acc Chem Res 2014, 47, 3541-50. (18) Penghua Yan, J. W., Lin Wang, Bin Liu, Ziqiang Lei, Shengrong Yang. The in vitro Biomineralization and Cytocompatibility of Polydopamine Coated Carbon Nanotubes. Applied Surface Science 2010, 257, 4849-4855. (19) Ku, S. H.; Ryu, J.; Hong, S. K.; Lee, H.; Park, C. B. General Functionalization Route for Cell Adhesion on Non-Wetting Surfaces. Biomaterials 2010, 31, 2535-41. (20) Black, K. C.; Yi, J.; Rivera, J. G.; Zelasko-Leon, D. C.; Messersmith, P. B. PolydopamineEnabled Surface Functionalization of Gold Nanorods for Cancer Cell-Targeted Imaging and Photothermal Therapy. Nanomedicine (Lond) 2013, 8, 17-28.

ACS Paragon Plus Environment

36

Page 37 of 49 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

(21) 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. Advanced Materials 2013, 25, 1353-1359. (22) Zhang, D.; Wu, M.; Zeng, Y.; Wu, L.; Wang, Q.; Han, X.; Liu, X.; Liu, J. Chlorin e6 Conjugated Poly(dopamine) Nanospheres as PDT/PTT Dual-Modal Therapeutic Agents for Enhanced Cancer Therapy. ACS Applied Materials & Interfaces 2015, 7, 8176-8187. (23) Chen, R.; Wang, X.; Yao, X.; Zheng, X.; Wang, J.; Jiang, X. Near-IR-Triggered Photothermal/Photodynamic Dual-Modality Therapy System via Chitosan Hybrid Nanospheres. Biomaterials 2013, 34, 8314-8322. (24) Guo, M.; Mao, H.; Li, Y.; Zhu, A.; He, H.; Yang, H.; Wang, Y.; Tian, X.; Ge, C.; Peng, Q.; Wang, X.; Yang, X.; Chen, X.; Liu, G.; Chen, H. Dual imaging-Guided Photothermal/Photodynamic Therapy Using Micelles. Biomaterials 2014, 35, 4656-66. (25) Yeo, E. L. L.; Cheah, J. U. J.; Lim, B. Y.; Thong, P. S. P.; Soo, K. C.; Kah, J. C. Y. Protein Corona around Gold Nanorods as a Drug Carrier for Multimodal Cancer Therapy. ACS Biomaterials Science & Engineering 2017, 3, 1039-1050. (26) Du, B.; Gu, X.; Zhao, W.; Liu, Z.; Li, D.; Wang, E.; Wang, J. Hybrid of Gold Nanostar and Indocyanine Green for Targeted Imaging-Guided Diagnosis and Phototherapy using LowDensity Laser Irradiation. Journal of Materials Chemistry B 2016, 4, 5842-5849. (27) Li, N.; Li, T.; Hu, C.; Lei, X.; Zuo, Y.; Han, H. Targeted Near-Infrared Fluorescent Turn-on Nanoprobe for Activatable Imaging and Effective Phototherapy of Cancer Cells. ACS Applied Materials & Interfaces 2016, 8, 15013-15023. (28) Cui, J.; Wang, Y.; Postma, A.; Hao, J.; Hosta-Rigau, L.; Caruso, F. Monodisperse Polymer Capsules: Tailoring Size, Shell Thickness, and Hydrophobic Cargo Loading via Emulsion Templating. Advanced Functional Materials 2010, 20, 1625-1631. (29) Cui, J.; Yan, Y.; Such, G. K.; Liang, K.; Ochs, C. J.; Postma, A.; Caruso, F. Immobilization and Intracellular Delivery of an Anticancer Drug using Mussel-Inspired Polydopamine Capsules. Biomacromolecules 2012, 13, 2225-8. (30) Mrowczynski, R. Polydopamine-Based Multifunctional (Nano)materials for Cancer Therapy. ACS Appl Mater Interfaces 2018, 10, 7541-7561. (31) Guo, P.; Liu, D.; Subramanyam, K.; Wang, B.; Yang, J.; Huang, J.; Auguste, D. T.; Moses, M. A. Nanoparticle Elasticity Directs Tumor Uptake. Nat Commun 2018, 9, 130. (32) Ho, C. C.; Ding, S. J. The pH-controlled Nanoparticles Size of Polydopamine for AntiCancer Drug Delivery. J Mater Sci Mater Med 2013, 24, 2381-90. (33) Doktorovova, S.; Shegokar, R.; Martins-Lopes, P.; Silva, A. M.; Lopes, C. M.; Muller, R. H.; Souto, E. B. Modified Rose Bengal Assay for Surface Hydrophobicity Evaluation of Cationic Solid Lipid Nanoparticles (cSLN). Eur J Pharm Sci 2012, 45, 606-12. (34) Xiao, Y.; Wiesner, M. R. Characterization of Surface Hydrophobicity of Engineered Nanoparticles. J Hazard Mater 2012, 215-216, 146-51. (35) Hunter, C. A.; Sanders, J. K. M. The Nature of .Pi.-.Pi. Interactions. Journal of the American Chemical Society 1990, 112, 5525-5534. (36) Della Vecchia, N. F.; Luchini, A.; Napolitano, A.; D’Errico, G.; Vitiello, G.; Szekely, N.; d’Ischia, M.; Paduano, L. Tris Buffer Modulates Polydopamine Growth, Aggregation, and Paramagnetic Properties. Langmuir 2014, 30, 9811-9818. (37) Fissan, H.; Ristig, S.; Kaminski, H.; Asbach, C.; Epple, M. Comparison of Different Characterization Methods for Nanoparticle Dispersions Before and After Aerosolization. Analytical Methods 2014, 6, 7324-7334.

ACS Paragon Plus Environment

37

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 38 of 49

(38) Eaton, P.; Quaresma, P.; Soares, C.; Neves, C.; de Almeida, M. P.; Pereira, E.; West, P. A Direct Comparison of Experimental Methods to Measure Dimensions of Synthetic Nanoparticles. Ultramicroscopy 2017, 182, 179-190. (39) Cone, R. A. Barrier Properties of Mucus. Advanced Drug Delivery Reviews 2009, 61, 75-85. (40) Lai, S. K.; Wang, Y.-Y.; Hanes, J. Mucus-Penetrating Nanoparticles for Drug and Gene Delivery to Mucosal Tissues. Advanced drug delivery reviews 2009, 61, 158-171. (41) Liu, M.; Zhang, J.; Shan, W.; Huang, Y. Developments of Mucus Penetrating Nanoparticles. Asian Journal of Pharmaceutical Sciences 2015, 10, 275-282. (42) Boegh, M.; Nielsen, H. M. Mucus as a Barrier to Drug Delivery – Understanding and Mimicking the Barrier Properties. Basic & Clinical Pharmacology & Toxicology 2015, 116, 179-186. (43) Kah, J. C. Y.; Chen, J.; Zubieta, A.; Hamad-Schifferli, K. Exploiting the Protein Corona around Gold Nanorods for Loading and Triggered Release. ACS Nano 2012, 6, 6730-6740. (44) Ritz, S.; Schöttler, S.; Kotman, N.; Baier, G.; Musyanovych, A.; Kuharev, J.; Landfester, K.; Schild, H.; Jahn, O.; Tenzer, S.; Mailänder, V. Protein Corona of Nanoparticles: Distinct Proteins Regulate the Cellular Uptake. Biomacromolecules 2015, 16, 1311-1321. (45) Park, D. J.; Min, K. H.; Lee, H. J.; Kim, K.; Kwon, I. C.; Jeong, S. Y.; Lee, S. C. Photosensitizer-Loaded Bubble-Generating Mineralized Nanoparticles for Ultrasound Imaging and Photodynamic Therapy. Journal of Materials Chemistry B 2016, 4, 12191227. (46) Adimoolam, M. G.; A, V.; Nalam, M. R.; Sunkara, M. V. Chlorin e6 Loaded Lactoferrin Nanoparticles for Enhanced Photodynamic Therapy . Journal of Materials Chemistry B 2017, 5, 9189-9196. (47) Zhu, Z.; Su, M. Polydopamine Nanoparticles for Combined Chemo- and Photothermal Cancer Therapy. Nanomaterials 2017, 7, 160. (48) Huang, X.; Jain, P. K.; El-Sayed, I. H.; El-Sayed, M. A. Plasmonic Photothermal Therapy (PPTT) Using Gold Nanoparticles. Lasers in Medical Science 2007, 23, 217. (49) Pattani, V. P.; Tunnell, J. W. Nanoparticle-Mediated Photothermal Therapy: A Comparative Study of Heating for Different Particle Types. Lasers in Surgery and Medicine 2012, 44, 675-684. (50) Jiang, K.; Smith, D. A.; Pinchuk, A. Size-Dependent Photothermal Conversion Efficiencies of Plasmonically Heated Gold Nanoparticles. The Journal of Physical Chemistry C 2013, 117, 27073-27080. (51) Qin, Z.; Wang, Y.; Randrianalisoa, J.; Raeesi, V.; C.W. Chan, W.; Lipinski, W.; Bischof, J. Quantitative Comparison of Photothermal Heat Generation between Gold Nanospheres and Nanorods, Scientific Reports, 2016, 6. (52) Weissleder, R. A Clearer Vision for in vivo Imaging. Nat Biotech 2001, 19, 316-317. (53) Yeo, E. L. L.; Thong, P. S. P.; Soo, K. C.; Kah, J. C. Y. Protein Corona in Drug Delivery for Multimodal Cancer Therapy in vivo. Nanoscale 2018, 10, 2461-2472. (54) Wust, P.; Hildebrandt, B.; Sreenivasa, G.; Rau, B.; Gellermann, J.; Riess, H.; Felix, R.; Schlag, P. M. Hyperthermia in Combined Treatment of Cancer. The Lancet Oncology 2002, 3, 487-497. (55) Field, S. B.; Bleehen, N. M. Hyperthermia in the Treatment of Cancer. Cancer Treatment Reviews 1979, 6, 63-94.

ACS Paragon Plus Environment

38

Page 39 of 49 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

(56) Oh, J.-M.; Choi, S.-J.; Kim, S.-T.; Choy, J.-H. Cellular Uptake Mechanism of an Inorganic Nanovehicle and Its Drug Conjugates:  Enhanced Efficacy Due To Clathrin-Mediated Endocytosis. Bioconjugate Chemistry 2006, 17, 1411-1417. (57) Cheng, Y.; C. Samia, A.; Meyers, J. D.; Panagopoulos, I.; Fei, B.; Burda, C. Highly Efficient Drug Delivery with Gold Nanoparticle Vectors for in Vivo Photodynamic Therapy of Cancer. Journal of the American Chemical Society 2008, 130, 10643-10647. (58) Ochsner, M. Photophysical and Photobiological Processes in the Photodynamic Therapy of Tumours. Journal of Photochemistry and Photobiology B: Biology 1997, 39, 1-18. (59) Kah, J. C. Y.; Yeo, E. L. L.; Koh, W. L.; Poinard, B. E. A.; Neo, D. J. H. Nanoparticle Interface to Biology: Applications in Probing and Modulating Biological Processes. 2013, 41, 323-341. (60) Camerin, M.; Rodgers, M. A. J.; Kenney, M. E.; Jori, G. Photothermal Sensitisation: Evidence for the Lack of Oxygen Effect on the Photosensitising Activity. Photochemical & Photobiological Sciences 2005, 4, 251-253. (61) Park, J.; Brust, T. F.; Lee, H. J.; Lee, S. C.; Watts, V. J.; Yeo, Y. Polydopamine-Based Simple and Versatile Surface Modification of Polymeric Nano Drug Carriers. ACS nano 2014, 8, 3347-3356.

ACS Paragon Plus Environment

39

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 40 of 49

TABLE OF CONTENTS GRAPHIC

Polydopamine (PDA) nanoparticles was loaded with a hydrophobic photosensitizer Chlorin e6 (Ce6) by pi-pi interactions between the aromatic cycles in the polymer matrix for enhanced drug release. This also enabled enhanced Ce6 cell uptake and therapeutic anti-cancer efficacy with dual photodynamic and photothermal therapy compared to free Ce6.

ACS Paragon Plus Environment

40

Page 41 of 49 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

Polydopamine (PDA) nanoparticles was loaded with a hydrophobic photosensitizer Chlorin e6 (Ce6) by pi-pi interactions between the aromatic cycles in the polymer matrix for enhanced drug release. This also enabled enhanced Ce6 cell uptake and therapeutic anti-cancer efficacy with dual photodynamic and photothermal therapy compared to free Ce6. 309x188mm (72 x 72 DPI)

ACS Paragon Plus Environment

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

Figure 1. Schematic of the preparation of PDA NPs and loading of hydrophobic photosensitizer Ce6 into the polymer matrix of PDA NPs. 251x139mm (72 x 72 DPI)

ACS Paragon Plus Environment

Page 42 of 49

Page 43 of 49 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. Characterization of PDA and PDA-Ce6 NPs with TEM images showing (A) PDA NPs without Ce6 and (B) PDA-Ce6 NPs. (C) Hydrodynamic diameter (DH) histogram distribution profile of PDA NPs (black) and PDA-Ce6 (red). (D) Zeta Potential of PDA NPs with (red) and without (black) Ce6 loaded under different pH conditions. (E) Characterization of the hydrophilicity and hydrophobicity using Rose Bengal. PDA NPs with (red) and without (black) Ce6 were re-suspended in rose Bengal solution for 2 h. The colloidal suspension was then centrifuged and the absorbance of the supernatant measured. The absorbance of the supernatant without any PDA NPs is shown as a reference (blue). (F) The partition coefficient of the PDA NPs incubated with varying concentrations of Ce6 to form PDA-Ce6 NPs. (G) Fluorescence emission spectra after excitation at 404 nm of 1 nM PDA NPs (black); 1 nM PDA NPs loaded with Ce6 (Blue); and 15 µM free Ce6 (red) at the same concentration as the amount of Ce6 loaded onto the PDA-Ce6. (H) Loading capacity of 1 nM of PDA in presence of varying initial Ce6 concentrations after 20 h incubation. 165x204mm (72 x 72 DPI)

ACS Paragon Plus Environment

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

ACS Paragon Plus Environment

Page 44 of 49

Page 45 of 49 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) Colloidal stability of PDA NPs in different media over time as measured by their hydrodynamic diameter, DH in the different media. (B) Table summarizing the average DH and surface charge of PDA NPs in different media after 12 hours. 176x73mm (300 x 300 DPI)

ACS Paragon Plus Environment

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

Figure 4. Passive release of Ce6 from 1 nM of PDA-Ce6. (A) Release of Ce6 from PDA-Ce6 over 10 days in Tris buffer of different pH. (B) Release of Ce6 from PDA-Ce6 NPs in RPMI with 10%FBS, human serum, and urine over a period of 2 weeks. In all cases, PDA-Ce6 was formed by incubating PDA NPs with 140 µM of Ce6 for 20 h. (C) Burst release profile in urine over a span of 1 day of PDA NPs incubated with 140 µM of Ce6. (D) Amount of Ce6 thermally released after heating 1 nM of PDA-Ce6 at 37 and 45 °C for different duration. 173x115mm (72 x 72 DPI)

ACS Paragon Plus Environment

Page 46 of 49

Page 47 of 49 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. Photothermal and photodynamic properties of PDA and PDA-Ce6 NPs. (A) Absolute temperature obtained after irradiating 1 nM PDA NPs in different pH conditions and 10 mM Tris buffer without any NPs as a control with a 665 nm CW laser at 250 mW/cm² for 15 min. (B) Absorbance spectra of PDA, PDA-Ce6, 15 µM Free Ce6 and Tris buffer without any NPs. ROS generation with (C) increasing PDA-Ce6 concentrations at physiological pH and (D) increasing pH at fixed concentration of 1 nM PDA-Ce6. The PDA-Ce6 NPs were either subjected to heat (100 °C for 10 min) (red, dotted) to release the maximal amount of Ce6 for irradiation or unheated to leave the Ce6 loaded within the PDA NPs during irradiation (blue, dotted). Free Ce6 at equivalent concentration as that released from the heating was included as a comparison (green, dotted). The samples were either kept in the dark (solid lines) or irradiated with a 665 nm CW laser 250 mW/cm² for 15 min (dotted lines). 179x121mm (72 x 72 DPI)

ACS Paragon Plus Environment

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

Figure 6. Cellular uptake of PDA-Ce6 NPs over a range of PDA-Ce6 concentration dosed to T24 cells and incubation time as quantified by (A) subtracting the absorbance measurement of non-uptaken PDA-Ce6 NPs recovered from cell culture supernatant from the initial absorbance of PDA-Ce6 added to cells, and (B) by measuring the fluorescence intensity of Ce6 within cells using flow cytometry. (C) Comparison of cellular uptake of 15 µM of free Ce6 and 15 µM of Ce6 loaded in 1 nM of PDA-Ce6 in terms of Ce6 concentration uptaken and uptake percentage in T24 cells over the concentration dosed for varying incubation times as quantified via the supernatant loss absorbance measurement technique in (A). 171x117mm (72 x 72 DPI)

ACS Paragon Plus Environment

Page 48 of 49

Page 49 of 49 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 7. Fluorescence imaging of the cellular uptake and localization of (A) 15 µM free Ce6 and (B) 1 nM PDA-Ce6 loaded with the same 15 µM Ce6 over a range of incubation time. Ce6 fluorescence was shown in the red channel, while the cell nucleus (DAPI-stained) was shown in the blue channel. Scale bar = 20 µm. (C) Quantification of Ce6 fluorescence intensity of the different fluorescent images using ImageJ. 176x121mm (72 x 72 DPI)

ACS Paragon Plus Environment

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

Figure 8. Cell viability after dosing T24 bladder cancer cells with varying concentrations of PDA NPs, PDACe6 NPs and the equivalent free Ce6 in medium supplemented with serum for 24 h and irradiating them with a 665 nm CW laser at 250 mW/cm2 for 15 min. Using PrestoBlue cell viability assay, the dark toxicity of PDA NPs, PDA-Ce6 NPs and free Ce6 were minimal (high cell viability > 80%). The viability decreased drastically with combined PDT+PTT with 1 nM PDA-Ce6 (dotted red) compared to PTT alone with 1 nM PDA (dotted black) and PDT alone with 15 µM free Ce6 (dotted green). 169x129mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 50 of 49