Accepted Manuscript Title: Targeting delivery of tocopherol and doxorubicin grafted-chitosan polymeric micelles for cancer therapy: in vitro and in vivo evaluation Author: Joung-Pyo Nam Kyeong-Jae Lee Joung-Woo Choi Chae-OK Yun Jae-Woon Nah PII: DOI: Reference:

S0927-7765(15)00391-4 http://dx.doi.org/doi:10.1016/j.colsurfb.2015.06.018 COLSUB 7148

To appear in:

Colloids and Surfaces B: Biointerfaces

Received date: Revised date: Accepted date:

31-3-2015 14-5-2015 8-6-2015

Please cite this article as: J.-P. Nam, K.-J. Lee, J.-W. Choi, C.-O.K. Yun, J.-W. Nah, Targeting delivery of tocopherol and doxorubicin grafted-chitosan polymeric micelles for cancer therapy: in vitro and in vivo evaluation, Colloids and Surfaces B: Biointerfaces (2015), http://dx.doi.org/10.1016/j.colsurfb.2015.06.018 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Highlights We synthesized targeting ligand-modified multidrug grafted polymeric micelle (HPTOC-DOX).  HPTOC-DOX formed micelles of approximately 151.9 – 311.2 nm in size in aqueous solution.  Doxorubicin was released from HPTOC-DOX micelles at acidic pH 5.2 or 6.2.  In vitro and in vivo anti-tumor assays showed higher anti-cancer activity than target peptide un-modified polymeric micelles.

d

M

an

us

cr

ip t



te

2 3 4 5 6 7 8 9 10

Ac ce p

1

1

Page 1 of 36

1

Targeting delivery of tocopherol and doxorubicin grafted-chitosan

2

polymeric micelles for cancer therapy: in vitro and in vivo evaluation

3

Joung-Pyo Nam1, Kyeong-Jae Lee1, Joung-Woo Choi2, Chae-OK Yun2, Jae-Woon Nah1,*

ip t

4

cr

5 6

1

7

University, 255 Jungang-ro, Suncheon, Jeollanam-do, Republic of Korea

8

2

9

Republic of Korea

us

Department of Polymer Science and Engineering, College of Engineering, Sunchon National

an

Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791,

10

M

11

* Corresponding authors: Department of Polymer Science and Engineering, Sunchon National

13

University, Suncheon, Republic of Korea. Tel: 82 61 750 3566, Fax: 82 61 750 5423, Email:

14

[email protected]

te

Ac ce p

15

d

12

3

Page 2 of 36

1

Abstract In this study, we report the development of a novel, redox-sensitive chitosan-based targeted

3

drug delivery system, containing two drugs. We determined whether the synthesized

4

polymeric micelles (HPTOC-DOX) were suitable as a drug carrier. The formation of

5

HPTOC-DOX micelles was confirmed by 1H-NMR. HPTOC-DOX formed micelles of

6

approximately 151.9–311.2 nm in size in aqueous solution. Analysis of the drug release

7

profile of HPTOC-DOX in different pH conditions (pH 5.2, 6.2, and 7.4) indicated that DOX

8

was released from HPTOC-DOX micelles at acidic pH (5.2 or 6.2), while almost no DOX

9

was released at pH 7.4. In vitro cell cytotoxicity and hemolysis assays indicated that HPTOC-

10

DOX micelles safely deliver anti-cancer drugs and decrease the cytotoxicity of DOX. In vitro

11

anti-cancer activity assays, confocal laser scanning microscopy analysis of SK-BR-3 cells,

12

and in vivo anti-tumor activity in SK-BR-3-derived tumor-bearing mice were used to evaluate

13

synergistic drug effects and the effect of the targeting peptide (anti-human epidermal growth

14

factor receptor 2 [HER2] target peptide, epitope form; LTVSPWY) on receptor-mediated

15

endocytosis.

17 18

cr

us

an

M

d

te

Ac ce p

16

ip t

2

Keywords: chitosan, doxorubicin, pH-sensitive, targeted delivery

4

Page 3 of 36

1

1. Introduction In the past few decades, drug delivery systems (DDS) have been developed to effectively

3

deliver drugs for cancer therapy. Several recent studies have described delivery carriers with

4

redox-sensitive and site-specific targeting functions [1-4]. These carriers have advantages

5

such as controlled release, decreased side effects, site-specific drug targeting, improved drug

6

utilization, lower dose administration, and drug loss prevention [5]. Polymeric micelles that

7

form a core-shell structure based on amphiphilic copolymers are ideal drug carriers because

8

of their numerous advantages such as improved solubility and bioavailability of poorly

9

soluble hydrophobic drugs in aqueous solutions [6, 7]. In addition, polymeric micelles

10

effectively protect the encapsulated drug from fast degradation after intravenous injection.

11

However, polymeric micelles have low cellular uptake because of the reticuloendothelial

12

system [7, 8]. To overcome this problem, various targeting ligands such as transferrin [9],

13

folic acid [10, 11], galactose [12], mannose [13], and an anti-human epidermal growth factor

14

receptor 2 (HER2) antibody (trastuzumab) [14] have been used for drug or gene delivery.

15

These targeting ligands enhance not only cellular uptake through receptor-mediated

16

endocytosis, but also therapeutic efficacy.

cr

us

an

M

d

te

Ac ce p

17

ip t

2

Chitosan is a linear polysaccharide with randomly distributed β-(1-4)-linked

D-

18

glucosamine (deacetylated unit) and N-acetyl-D-glucosamine (acetylated unit). Chitosan is

19

one of the major cationic polymers and has a structure similar to cellulose [15, 16]. Chitosan

20

is obtained from chitin by alkaline deacetylation and inter- or intra-molecular hydrogen

21

bonding between hydroxyl and amine groups [17]. Chitosan is widely used in DDS

22

development as a backbone for polymeric micelles owing to its biocompatibility,

23

biodegradability, high stability, and lack of toxicity. O-Carboxymethyl chitosan (OCMCh) is

24

a derivative of chitosan with a CH2COOH modification at the C6 position of chitosan, which

25

enhances its solubility in aqueous solutions. OCMCh has various advantages such as good 5

Page 4 of 36

1

bioactivity, solubility in aqueous solutions, antibacterial activity, and stability [18-20]. Doxorubicin (DOX), the model drug used in this study, is an anti-cancer drug also known

3

as hydroxydaunorubicin, which is used in chemotherapy to treat cancers such as bladder,

4

breast, stomach, ovarian, thyroid, and lung cancers, multiple myeloma, and soft tissue

5

sarcoma [21]. The high antitumor activity of DOX is associated with multidrug resistance

6

and acute cardiotoxicity [22, 23]. To solve these problems, several methods of DOX delivery

7

were developed mainly using hydrophobic interactions with the polymeric micelles [24-26].

8

Polymeric micelles with drugs loaded by hydrophobic interaction have some advantages,

9

such as easy preparation and applicability for all hydrophobic drugs, but there are also some

10

disadvantages associated with their use, such as an inconsistent drug release rate and

11

modification However, polymeric micelles, with a drug covalently conjugated to the micelle

12

backbone, have the potential to overcome these problems.

M

an

us

cr

ip t

2

α-Tocopherol (TP), also known as vitamin E, is a hydrophobic compound consisting of

14

various methylated phenols. TP is not toxic to normal cells, but has anti-cancer activity

15

against different cancer cell lines [27-30]. It can induce apoptosis and inhibit cell

16

differentiation by blocking the cell cycle [27]. In addition, it can self-assemble into polymeric

17

micelles.

Ac ce p

te

d

13

18

The aim of this study was to synthesize novel polymeric micelles consisting of TP, DOX,

19

and OCMCh, to achieve a synergistic anti-cancer effect. We designed anti-cancer drug-

20

micelle conjugates to overcome the limitations of micelles, in which drugs are loaded through

21

hydrophobic interactions. DOX was conjugated to redox-sensitive dithiobis-succinimidyl

22

propionate (DTSP) that is cleavable in the cytoplasm by an intracellular reducing agent such

23

as glutathione (GSH, 0.5 – 10 mM) [1, 2, 31, 32]. Furthermore, a targeting ligand (anti-

24

HER2/neu peptide-polyethylene glycol [PEG]; HP) was added to TP and DOX-conjugated

25

polymeric micelles (TOC-DOX) to promote site-specific targeting. The synthesized targeting 6

Page 5 of 36

ligand-containing novel polymeric micelles (HPTOC-DOX) were expected to have the

2

following effects: (1) synergistic effect of the conjugated TP and DOX, (2) redox-sensitive

3

drug release in solid tumors owing to a disulfide bond, and (3) site-specific delivery of anti-

4

cancer drugs such as TP and DOX. In this study, we performed in vitro and in vivo

5

experiments to determine whether the novel polymeric HPTOC-DOX micelles can be used

6

for cancer therapy.

cr

ip t

1

Ac ce p

te

d

M

an

us

7

7

Page 6 of 36

1

2. Materials and methods 2.1. Materials

3

O-Carboxymethyl chitosan (OCMCh, molecular weight (MW) = 12,000 Da, deacetylation

4

degree = 95%, and carboxymethylation degree = 83%) was prepared as previously described

5

[33]. TP and DOX were purchased from Sigma-Aldrich Chemical Co. USA. The crosslinkers

6

DTSP, 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC), and N-

7

hydroxysuccinimide (NHS) were purchased from Sigma-Aldrich Chemical Co. USA.

8

HER2/neu-targeting peptides (epitope form, LTVSPWY-COOH) were synthesized with a

9

Liberty microwave peptide synthesizer (CEM Co. Matthews, NC) using Fmoc solid-phase

10

methods. NH2-PEG-COOH (MW = 3,400) was purchased from SunBio, Korea. Dulbecco’s

11

Modified Eagle’s Medium (DMEM), Roswell Park Memorial Institute (RPMI)-1640 medium,

12

and fetal bovine serum (FBS) were obtained from Gibco BRL (MD, USA). LysoTracker and

13

Hoechst 33258 were purchased from Invitrogen. All other chemicals and solvents were of

14

analytical or reagent grade and were used without further purification.

15

2.2. Preparation of anti-cancer drug conjugated polymeric micelles

16

The detailed method of TP conjugation to OCMCh (TOC), DOX conjugation to TOC (TOC-

17

DOX), and targeting ligand addition to TOC-DOX (HPTOC-DOX) polymeric micelles is

18

described in the Supplementary Information.

Ac ce p

te

d

M

an

us

cr

ip t

2

19

2.3. Characterization of anti-cancer drug conjugated polymeric micelles

20

The synthesized HPTOC-DOX micelles were characterized by 1H-NMR. The 1H-NMR

21

spectra of micelles were measured in deuterium oxide (D2O) by using a 400 MHz NMR

22

spectrometer (AVANCE 400FT-NMR 400 MHz, Bruker). Five milligrams of HPTOC-DOX 8

Page 7 of 36

1

were dissolved in 0.5 mL of D2O and analyzed with an NMR spectrometer. The products of

2

intermediate structures, such as TOC, DTSP-modified DOX (DOX-DTSP), and TOC-DOX

3

were also measured. The detailed method is described in the Supplementary Information. The average particle size and size distribution of TOC, TOC-DOX, and HPTOC-DOX

5

micelles were determined using an ELS-8000 electrophoretic LS spectrophotometer

6

(NICOMP 380 ZLS zeta potential/particle sizer; Otsuka Electronics INC., Japan), equipped

7

with a He-Ne laser, at a wavelength of 632.8 nm at 25 °C (scattering angle of 90°). A micelle

8

solution (10 µg/µL) was used for particle measurement (HE. 013.016-ALU cell adapter)

9

without filtering.

an

us

cr

ip t

4

DOX concentrations and conjugation efficiency of TOC-DOX and HPTOC-DOX polymeric

11

micelles were determined using high-performance liquid chromatography (HPLC) analysis.

12

The polymeric micelles were dissolved in 50% DMSO (1 mg/mL) and then the solution was

13

centrifuged. The supernatant was collected for analysis. The HPLC system consisted of a

14

mobile phase delivery pump (LC-20AD HPLC pump, Shimadzu, Japan) and a UV detector

15

(SPD-20A, UV/Vis detector, Shimadzu, Japan). A ZORBAX 300SB-C18 reverse-phase

16

column (250 mm × 4.6 mm, 5 µm, Agilent Technologies Inc., USA) was used for separation

17

of components. The mobile phase was composed of acetonitrile and water at a ratio of 40:60

18

(v/v). The flow rate and column temperature were set at 1.0 mL/min and 30 °C, respectively.

19

The UV absorbance at 280 nm was determined using an injection volume of 20 µL. DOX

20

concentration and conjugation efficiency were calculated from standard curves. The curve

21

was linear over the tested concentration range. The drug content (DC) and conjugation

22

efficiency (CE) were calculated as follows:

Ac ce p

te

d

M

10

23

Weight of the drug in micelles

24 25

DC% =

—————————————————— × 100% 9

Page 8 of 36

Weight of the feeding polymer and drug

1 2 3

CE% =

————————————————— × 100% Weight of the feeding drug

6

cr

5

ip t

Weight of the drug in micelles

4

us

7

2.4. DOX release profile from micelles against different pH conditions

9

The drug release profile of DOX from TOC-DOX or HPTOC-DOX was investigated by

10

HPLC. One milligram of TOC-DOX or HPTOC-DOX was dissolved in 1 mL of phosphate-

11

buffered saline (PBS) and then filtered using centrifugal filter devices (molecular weight cut-

12

off [MWCO], 1 kDa; AMICON® Ultra-0.5, Millipore). The solution obtained by filtration

13

and centrifugation was analyzed by HPLC. To confirm drug release from micelles in different

14

pH conditions, we used acidic pH (pH 5.2 and 6.2) and neutral pH (7.4) with interval times

15

from 30 min to 30 h. The pH conditions were stable at 7.4 from 30 min to 24 h and then

16

decreased to 5.2.

Ac ce p

te

d

M

an

8

17

2.5. Cells and cell culture conditions

18

Human breast cancer cells (SK-BR-3), human embryonic kidney cells (HEK 293), and

19

human keratinocytes (HaCaT) were purchased from KCLB® (Seoul, Republic of Korea). The

20

cells were cultured in RPMI-1640 or DMEM supplemented with 10% FBS and 1%

21

penicillin/streptomycin in a humidified atmosphere containing 5% CO2 at 37 °C. The cells

22

grown as a monolayer were harvested after incubation with trypsin-EDTA.

10

Page 9 of 36

2.6. Confocal laser scanning microscopy (CLSM) assay

2

The localization of TOC-DOX or HPTOC-DOX in SK-BR-3 cells was determined with

3

CLSM analysis. SK-BR-3 cells were seeded at a density of 1 × 105 cells/well in eight-well

4

culture plates and cultured overnight. After 12 h, free DOX, TOC-DOX, and HPTOC-DOX

5

(0.5 µg/mL equivalent concentration of DOX) were added to the SK-BR-3 cell culture

6

medium. The cells were incubated for 1 h and then washed three times with PBS. The cells

7

were pre-incubated with Hoechst 33258 (Dojindo Molecular Technologies), a nucleic acid

8

stain, to determine the intracellular localization of micelles. In addition, the cells were labeled

9

with the lysosome stain LysoTracker (Molecular Probes, OR, USA). The localization of

10

TOC-DOX or HPTOC-DOX was determined using an inverted LSM510 laser scanning

11

microscope (Carl Zeiss, Gottingen, Germany). The diode laser excitation wavelength of 405

12

nm and helium neon laser excitation wavelengths of 511 and 543 nm were used to detect

13

DOX-conjugated micelles, LysoTracker, and Hoechst 33258. Images were recorded digitally

14

using a 512 × 512 pixel format.

Ac ce p

te

d

M

an

us

cr

ip t

1

15

2.7. Cytotoxicity and anticancer activity assay in vitro

16

The cytotoxicity and anti-cancer activity of OCMCh, TP, DOX, and polymeric micelles

17

(TOC-DOX and HPTOC-DOX) in different cell lines (both normal and cancer cells) were

18

evaluated with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)

19

assay. The experiment was carried out as follows: HEK293, HaCaT, and SK-BR-3 cells were

20

seeded into 96-well microplates at a density of 5 × 103 cells/well in 100 µL of medium. When

21

cell confluence reached 80%, the cells were treated with OCMCh, TP, DOX, and polymeric

22

micelles for 24 h at DOX equivalent concentrations ranging from 0.078125 to 10 µg/mL.

23

After the incubation, 10 µL of MTT solution (5 mg/mL in PBS) was added to each well, and

11

Page 10 of 36

the plate was incubated for an additional 2 h at 37 °C. At the determined time, unreacted

2

MTT was removed by aspiration. After removing the MTT-containing medium, the formazan

3

crystals formed in live cells were dissolved with 100 µL of DMSO. Finally, the absorbance

4

was measured at 570 nm (optical density) and 670 nm (background subtraction) with a

5

VersaMax ELISA Microplate Reader (StakMax®, USA). The relative cell viability (%) was

6

calculated according to the following equation: OD value = (OD560 - OD670) (OD sample - OD blank)

8

us

7

cr

ip t

1

Relative cell viability (%) = ————————————————— × 100% (OD control - OD blank)

an

9

M

10

2.8. Hemolytic activity of TOC-DOX and HPTOC-DOX

12

The hemolytic activity of TOC-DOX and HPTOC-DOX was evaluated in red blood cells

13

(RBCs) from inbred rats (Orientbio Inc., Korea) by using a previously described method [34].

14

Fresh rat blood was centrifuged at 800 × g and the collected RBCs were washed with pH 7.4

15

PBS until the supernatant was clear. Polymeric micelles were dissolved in PBS at

16

concentrations ranging from 0.625 to 10 µg/mL. Polymeric micelle solutions (equal volumes)

17

were aliquoted in a 96-well plate and RBCs were then added to a final concentration of 8%

18

(v/v). The samples were incubated with mild agitation for 1 h at 37 °C and then centrifuged at

19

800 × g for 10 min. The absorbance of the supernatant was measured with a VersaMax

20

ELISA Microplate Reader (StakMax®, USA) at 414 nm. Each measurement was made in

21

triplicate, and the percentage of hemolysis was calculated using followed equation [35]:

Ac ce p

te

d

11

(A414 sample - A414 PBS)

22 23 24

Hemolysis (%) =

× 100% (A414 0.1% Triton X-100 - A414 PBS) 12

Page 11 of 36

1

In addition, 100% hemolysis was defined as the absorbance of RBCs resuspended in 0.1%

3

Triton X-100 and zero hemolysis as the absorbance of RBCs in PBS.

ip t

2

2.9. In vivo anti-tumor effect of TOC-DOX and HPTOC-DOX and histological analysis

5

To assess the anti-tumor effect of TOC-DOX and HPTOC-DOX, xenograft tumors of HER2

6

overexpressing SK-BR-3 cells were established by injecting 1 × 106 cells into the lower

7

abdomen of 6- to 8-week-old female athymic nude mice (Orientbio Inc.). Once tumor volume

8

reached 100–120 mm3, the mice were randomized into three groups and injected

9

intravenously with 200 µL of PBS, TOC-DOX, or HPTOC-DOX two times every other day

10

(n = 5, 20 mg/kg). Anti-tumor efficacy was analyzed by measuring tumor size with a caliper

11

three times a week until the end of study. The length (l) and width (w) of the tumor were

12

measured and the tumor volume was calculated according to the following formula: tumor

13

volume = 0.523 lw2.

te

d

M

an

us

cr

4

For histological analysis, tumor tissue was fixed in 4% formalin, embedded in paraffin, and

15

cut into 5-μm sections by using a microtome (Wax-it, Vancouver, Canada). Representative

16

sections were stained with hematoxylin and eosin (H&E), and then examined by light

17

microscopy. For immunohistochemistry, slides were deparaffinized in xylene and then

18

processed as previously described [36]. Immunohistochemical staining for proliferating cell

19

nuclear antigen (PCNA) was performed on paraffin-embedded tissue sections by using an

20

anti-PCNA antibody (Dako) as described previously [36]. Stained sections were

21

counterstained with Mayer’s hematoxylin. All animal studies were approved by the Animal

22

Care Committee of Sunchon National University (SCNU_IACUC-2013-6), and all animal

23

care was in accordance with the guidelines of Korea Council on Animal Care.

Ac ce p

14

13

Page 12 of 36

2.10. Statistical Analysis

2

All data are expressed as mean ± standard error (SE) or standard deviation (SD).

3

Statistically significant differences were determined using Stat View software (Abacus

4

Concepts, Inc., Berkeley, CA) and the Mann-Whitney test (non-parametric rank sum test). P

5

values less than 0.05 were considered statistically significant (*, P < 0.05; **, P < 0.01; ***,

6

P < 0.001).

cr

ip t

1

us

7

Ac ce p

te

d

M

an

8

14

Page 13 of 36

1

3. Results 3.1. Characterization of TOC-DOX and HPTOC-DOX micelles

3

In this study, we prepared novel polymeric micelles that have a redox-sensitive bond and

4

contain anti-cancer drugs. The reaction scheme of the final product (HPTOC-DOX) and

5

intermediate structures (TOC, DOX-DTSP, and TOC-DOX) is shown in Fig. S1.

cr

ip t

2

The 1H-NMR results of intermediate structures that were synthesized to prepare novel

7

polymeric micelles are shown in Figs. S2–S4 (see Supplementary Information). The final

8

product, which is a targeting ligand-modified TOC-DOX (HPTOC-DOX), was analyzed with 1

an

9

us

6

H-NMR to confirm successful synthesis. The proton peaks of TP, DOX, OCMCh, PEG, and

anti-HER2 in HPTOC-DOX are classified in Fig. 1. Peak assignments are as follow: OCMCh,

11

2.0 to 4.6 ppm; TP, 2.4 to 2.6 ppm; DOX, 1.1 to 1.4 ppm; PEG, 3.6 ppm; anti-HER2, 0.8 to 1

12

ppm. These 1H-NMR results showed that targeting ligand- and anti-cancer drug-conjugated

13

novel polymeric micelles were successfully synthesized.

14

The particle size and distribution of TOC, TOC-DOX, and HPTOC-DOX micelles were

15

measured by dynamic light scattering (DLS; Fig. 2). TOC, TOC-DOX, and HPTOC-DOX

16

micelles had a unimodal particle size distribution (Fig. 2B, presented HPTOC-DOX). When

17

TP increased the in the conjugation ratio of TOC, particle size increased from 1096.8 nm

18

(TOC1) to 1347.1 nm (TOC3). The particle size significantly decreased when the

19

hydrophobic drug was conjugated to TOC. In addition, targeting ligand-modified TOC-DOX

20

had a larger particle size of approximately 151.9 nm (HPTOC1-DOX) and 311.2 nm

21

(HPTOC3-DOX) compared to the unmodified TOC-DOX.

22 23

Ac ce p

te

d

M

10

3.2. In vitro DOX release profile of TOC-DOX and HPTOC-DOX micelles against different pH conditions

15

Page 14 of 36

The DOX release profile of TOC-DOX or HPTOC-DOX in different pH conditions (pH 5.2,

2

6.2, and 7.4) was determined using HPLC to confirm drug release from the novel polymeric

3

micelles. As shown Fig. 3, almost no DOX was released from any polymeric micelles

4

(TOC1-DOX, TOC3-DOX, HPTOC1-DOX, and HPTOC3-DOX) at pH 7.4 (the pH was

5

maintained in a time range from 30 min to 24 h). However, DOX was released when the pH

6

was changed from 7.4 to 5.2 or 6.2. TOC3-DOX and HPTOC3-DOX micelles released less

7

DOX compared to that released from TOC1-DOX and HPTOC1-DOX micelles at pH 6.2

8

(Fig. 3A). More DOX was released from polymeric micelles at pH 5.2 than at pH 6.2. These

9

results suggest that the novel polymeric micelles are maintained at neutral pH and start to

cr

us

an

10

ip t

1

release the drug in acidic pH conditions.

In addition, drug concentration (DC) and drug conjugation efficiency (CE) of polymeric

12

micelles was investigated using HPLC (Table S1). DC did not change, but CE increased

13

when the amount of TP, which was conjugated to OCMCh, increased. However, DC and CE

14

decreased when a targeting ligand was added to TOC-DOX. This phenomenon can be

15

explanined the accrued loss during the dialysis process and the targeting ligand modifying

16

process.

Ac ce p

te

d

M

11

17

3.3. Localization assay of polymeric micelles in SK-Br3 cells using CLSM

18

The polymeric micelles were taken up by cells and localized to the cytosol, endosomes,

19

lysosomes, or nuclei. As shown Fig. 4, polymeric micelles were localized in lysosomes and

20

nuclei, but free DOX localized only in nuclei. These results demonstrated that polymeric

21

micelles were taken up by endocytosis. TOC1 and TOC3 had similar LysoTracker intensity.

22

However, when the amount of conjugated TP increased in TOC1 to TOC3 (molar ratio), the

23

fluorescence intensity of DOX also increased, indicating that more DOX was conjugated to

24

TOC3 than to TOC1. This is consistent with the CE determined by HPLC. The merged 16

Page 15 of 36

images showed that targeting ligand-modified HPTOC-DOX micelles localized in cell nuclei,

2

while unmodified TOC-DOX showed perinuclear localization. These results clearly

3

demonstrated the effect of targeting ligand in HPTOC-DOX micelles. The targeting ligand

4

(HER2/neu-targeting peptide [pepitope form, LTVSPWY]) induced HER2/neu receptor-

5

mediated endocytosis in SK-BR-3 cells [37]. Therefore, the uptake of HPTOC-DOX by SK-

6

BR-3 cells was more efficient than that of unmodified TOC-DOX.

cr

ip t

1

3.4. In vitro stability of polymeric micelles

8

The results of the cell viability and hemolysis assays are shown in Fig. 5. The viability of

9

HEK293 or HaCaT cells treated with intermediate structures, including OCMCh, TP, TOC1,

10

and TOC3, is shown in Fig. S5 (see Supplementary Information). The cytotoxicity of DOX-

11

conjugated polymeric micelles (TOC1-DOX, TOC3-DOX, HPTOC1-DOX, and HPTOC3-

12

DOX) was lower than that of free DOX at equivalent DOX concentrations ranging from

13

0.078125 to 10 µg/mL in both HEK293 and HaCaT cells (Fig. 5A and B). The viability of

14

HEK293 cells treated with DOX-conjugated polymeric micelles at equivalent DOX

15

concentrations ranging from 0.078125 to 5 µg/mL was higher than 85% (Fig. 5A). However,

16

the viability of HaCaT cells treated with DOX-conjugated polymeric micelles at equivalent

17

DOX concentrations ranging from 2.5 to 10 µg/mL was below 85% (Fig. 5B). The slight

18

toxicity of polymeric micelles against normal cell lines is because of the toxicity of DOX.

19

A hemolysis assay was performed to determine whether TOC-DOX and HPTOC-DOX

20

micelles are safe for systemic administration. The results showed that TOC1-DOX, TOC3-

21

DOX, HPTOC1-DOX, and HPTOC3-DOX did not induce hemolysis at DOX concentrations

22

ranging from 0.625 to 1 µg/mL due to the low positive charge of OCMCh (Fig. 5C). These

23

results demonstrated that the prepared polymeric micelles can be used for systemic

24

administration.

Ac ce p

te

d

M

an

us

7

17

Page 16 of 36

3.5. Anti-tumor effect of polymeric micelles in vitro

2

To investigate the anti-cancer effect of polymeric micelles we measured the viability of

3

HER2/neu-overexpressing SK-BR-3 cells treated with TOC-DOX and HPTOC-DOX

4

micelles with the MTT assay. Fig. 6 shows the anti-cancer effect of intermediate structures

5

(OCMCh, TP, TOC1, and TOC3) and final products (TOC-DOX and HPTOC-DOX) at

6

equivalent intermediate structure or DOX concentrations ranging from 0.078125 to 10 µg/mL.

7

The viability of TP-, TOC1-, and TOC3-treated cells decreased with increasing intermediate

8

structure concentration, but OCMCh-treated cells were 100% viable at all concentrations

9

tested (Fig. 6A). The 50% inhibitory concentration (IC50) of TP, TOC1, and TOC3 was above

10

0.625 µg/mL (Table S1). Treatment with the DOX-conjugated polymeric micelles TOC-

11

DOX and HPTOC-DOX decreased cell viability to below 60% at all equivalent DOX

12

concentrations (Fig. 6B). The IC50 of TOC-DOX and HPTOC-DOX was approximately

13

0.078125 µg/mL (Table S1). TOC-DOX and HPTOC-DOX decreased cell viability to a

14

similar extent as free DOX at concentrations ranging from 2.5 to 10 µg/mL. In addition,

15

treatment with targeting ligand-modified HPTOC-DOX micelles decreased cell viability to a

16

greater extent than unmodified TOC-DOX. However, the amount of TP did not have a

17

significant effect on cell viability. Therefore, to examine the anti-tumor effect of polymeric

18

micelles in vivo, we selected TOC3-DOX and HPTOC3-DOX.

Ac ce p

te

d

M

an

us

cr

ip t

1

19

3.6. Anti-tumor effect and immunohistological analysis in HER2/neu-positive SK-Br3

20

tumor-bearing mice

21

To evaluate the anti-tumor effect of TOC3-DOX or HPTOC3-DOX micelles, the tumor

22

growth rate was examined after systemic administration of polymeric micelles to HER2/neu-

23

positive SK-BR-3 tumor-bearing mice (Fig. 7A). Twenty days after the polymeric micelles

18

Page 17 of 36

were administered; the average tumor volume was 4092.3 mm3, 3142.6 mm3, and 1650.7

2

mm3 in PBS, TOC3-DOX, and HPTOC3-DOX-treated mice, respectively. The tumor volume

3

of polymeric micelle-treated group was decreased as compared with the PBS-treated group

4

due to the effect of DOX conjugated to the polymeric micelles. In particular, the targeting

5

ligand-modified HPTOC3-DOX showed a higher therapeutic efficacy than the other

6

treatments (P < 0.05 for TOC3-DOX; P < 0.001 for PBS). These results show that treatment

7

with the targeting ligand (anti-HER2/neu)-modified HPTOC-DOX micelles led to an

8

impressive anti-tumor effect through the specific receptor-mediated interaction between the

9

targeting ligand on the HPTOC-DOX micelle surface and the HER2/neu receptor on the SK-

cr

us

an

10

ip t

1

BR-3 cell surface.

To confirm the therapeutic efficacy of TOC3-DOX and HPTOC3-DOX in SK-BR-3 tumor-

12

bearing mice, histological observation of TOC3-DOX and HPTOC3-DOX micelle-treated

13

tumor tissues was carried out after H&E staining. Tumors of PBS-treated mice had a high cell

14

density. In contrast, the tumors of TOC3-DOX- and HPTOC3-DOX-treated mice contained

15

necrotic cells. Furthermore, the necrotic area in tumors of HPTOC3-DOX-treated mice

16

dramatically increased as compared to the control and TOC3-DOX-treated groups (Fig. 7B).

17

PCNA is a marker of proliferating cells and PCNA immunostaining was used to assess tumor

18

cell proliferation (Fig. 7B). Tumors in the PBS-treated group (control) were highly positive

19

for PCNA, whereas tumors in the TOC3-DOX- or HPTOC3-DOX- treated groups were not.

20

In particular, PCNA expression was considerably reduced in the HPTOC3-DOX-treated

21

group as compared with the control and TOC3-DOX groups. These in vivo results clearly

22

demonstrate that the polymeric micelles can suppress tumor growth. In addition, the targeting

23

ligand can enhance the ability of polymeric micelles to deliver anti-cancer drugs.

Ac ce p

te

d

M

11

24 25 19

Page 18 of 36

1

4. Discussion In this study, we synthesized a novel polymeric micelle to develop a targeted and redox-

3

sensitive system for delivery of chemotherapeutic drugs such as TP and DOX. HPTOC-DOX

4

micelles were synthesized in three steps using chemical conjugation with a targeting ligand

5

(anti-HER2/neu targeting peptide), anti-cancer drugs (TP and DOX), and a redox-sensitive

6

bond (DTSP) to the OCMCh backbone (Fig. S1). This novel polymeric micelle has a

7

hydrophilic group and a hydrophobic group. Therefore, it can form micelles in aqueous

8

solution. Our results show that hydrophobic group-conjugated OCMCh formed particles. The

9

particle size of TOC1 and TOC3 was 1096.8 nm and 1347.1 nm, respectively. The

10

conjugation ratio of TP was increased 1% to 3% (molar ratio), but particle size increased

11

from 1096.8 nm to 1347.1 nm. The explanation for this phenomenon could be that TOC

12

formed micelles, but did not form strong hydrophobic interactions due to very low

13

hydrophobicity. We used OCMCh with an MW of 12,000 Da in this study. It consisted of

14

approximately 54.5 monomers (MW 220 Da). TP was conjugated to OCMCh monomers in

15

ratios of 0.5 (TOC1) and 1.6 (TOC3). Therefore, TOC has very low hydrophobicity and

16

relatively large particle sizes. The TOC micelles aggregated when TP was conjugated to

17

OCMCh with a molar ratio higher than 3%. However, when DOX was conjugated to TOC,

18

the particle size of TOC1-DOX and TOC3-DOX significantly decreased from 1096.8 nm to

19

127.4 and 1347.1 nm to 244.9 nm, respectively, because tight micelles were formed by strong

20

hydrophobic interactions. Several studies reported that the particle size decreased when

21

hydrophobic drugs were associated with polymeric micelles [38-40]. In addition, we

22

modified TOC-DOX micelles with a targeting ligand to allow site-specific delivery of anti-

23

cancer drugs. The particle size of HPTOC-DOX was slightly increased because of the

24

addition of the hydrophilic group (HER2/neu targeting peptide-PEG; HP).

25

Ac ce p

te

d

M

an

us

cr

ip t

2

In this study, DTSP was used as a linker to conjugate DOX to the micelle and to confer 20

Page 19 of 36

redox-sensitivity. DTSP has a disulfide bond and DTSP-based drug carriers allow drugs to be

2

accumulated in target tissues at higher concentrations than they would with drug carriers

3

without disulfide bond-containing linkers [41]. As mention above, DTSP is is cleavable in the

4

cytoplasm by an intracellular reducing agent such as glutathione (GSH, 0.5 – 10 mM) [1, 2,

5

31, 32]. The disulfide bond in HPTOC-DOX micelles allows DOX release at the target site

6

due to the reducing agent after cellular uptake.

cr

ip t

1

The DOX release profile from polymeric micelles at different pH values indicated that the

8

synthesized polymeric micelles (TOC-DOX and HPTOC-DOX) released DOX at the tumor

9

site. TOC-DOX and HPTOC-DOX retained the conjugated DOX at pH 7.4. This means that

10

the TOC-DOX and HPTOC-DOX micelles are stable in blood plasma (pH 7.4). Drug carriers

11

consisting of polycationic polymers such as chitosan, polyethylenimine (PEI), and polylysine

12

(PLL) may not be considered as drug delivery systems for systemic administration due to

13

their positive charge. Positively charged polycationic polymers can interact with negatively

14

charged erythrocyte membranes and lead to hemolysis. Therefore, the stability of the

15

synthesized polymeric micelles is very important for RBC stability. Our results show that

16

TOC-DOX and HPTOC3-DOX micelles are stable in blood plasma and can be used as a

17

systemic drug delivery system.

Ac ce p

te

d

M

an

us

7

18

In general, targeted delivery systems are divided into two kinds: passive targeting systems

19

and systems with covalently conjugated targeting molecules on the surface of drug carriers.

20

The targeting ligand conjugated to drug carriers can bind to receptors expressed specifically

21

in cancer cells [11, 14]. The target receptor, human epidermal growth factor receptor 2

22

(HER2), which is also known as Neu, CD340, ErbB-2, or p185, is overexpressed in

23

approximately 30% of breast cancers and 20% of ovarian cancers, while its expression in

24

normal tissues is low [14, 42]. The HPTOC-DOX micelle, containing an anti-HER2 targeting

25

peptide, showed a better therapeutic effect better than TOC-DOX both in vitro in SK-BR-3 21

Page 20 of 36

cells and in vivo in SK-BR-3 tumor-bearing mice (Fig. 6B and Fig. 7). These results clearly

2

demonstrated that HPTOC-DOX cellular uptake was through receptor-mediated endocytosis

3

via an interaction between the anti-HER2 targeting peptide and the overexpressed HER2/neu

4

receptor on the SK-BR-3 cell surface. The receptor-mediated endocytosis enhanced cellular

5

uptake efficacy of micelles and led to increased therapeutic efficacy. In addition, we designed

6

multi-drug-conjugated polymeric micelles. TP was used to not only enhance hydrophobicity

7

but also as an anti-cancer drug. TP has anti-cancer activity against various cancer cells and

8

can induce apoptosis and diminish cell differentiation by blocking the cell cycle [27-30].

9

Therefore, TP-conjugated TOC micelles at concentrations exceeding 0.625 µg/mL

10

significantly decreased the viability of SK-BR-3 cells (Fig. 6A). The synergistic effect of TP

11

and DOX was evident from the low cell viability after treatment with a very low

12

concentration (0.078125 µg/mL) of TOC-DOX and HPTOC-DOX (Fig. 6B). Interestingly,

13

the viability of cells treated with polymeric micelles was higher than or similar to that of cells

14

treated with free DOX at concentrations exceeding 0.625 µg/mL. These results demonstrate

15

that that TP and DOX in multi-drug-conjugated TOC-DOX and HPTOC-DOX have a

16

synergistic effect in tumor therapy.

Ac ce p

te

d

M

an

us

cr

ip t

1

17

In conclusion, we synthesized novel polymeric micelles with a targeting ligand, two anti-

18

cancer drugs (TP and DOX), and a redox-sensitive function. The synthesized TOC-DOX and

19

HPTOC-DOX formed polymeric micelles in aqueous solution through hydrophobic

20

interactions between the conjugated TP and DOX. The particle size of TOC-DOX and

21

HPTOC-DOX micelles was in the range of 124.7-244.9 nm and 151.9-311.2 nm, respectively.

22

The cumulative drug release of HPTOC-DOX of around 90% within 6 h after the pH changed

23

from 7.4 to 5.2. The anti-cancer drugs of TOC-DOX and HPTOC-DOX had a synergistic

24

effect against SK-BR-3 cells. The anti-HER2/neu targeting peptide (epitope form,

25

LTVSPWY) was used as the targeting moiety. It enhanced not only cellular uptake by 22

Page 21 of 36

HER2/neu overexpressing SK-BR-3 cells, but also therapeutic efficacy in SK-BR-3 tumor-

2

bearing mice. The targeting peptide-modified HPTOC-DOX had lower cytotoxicity than free

3

DOX and enhanced therapeutic efficacy as evidenced by results from both in vitro and in vivo

4

assays.

ip t

1

cr

5

Ac ce p

te

d

M

an

us

6

23

Page 22 of 36

Acknowledgements

2

This work was supported by the National Research Foundation of Korea (NRF) grant funded

3

by the Ministry of Science, ICT & Future Planning (NRF-2014R1A2A1A10053027).

ip t

1

Ac ce p

te

d

M

an

us

cr

4

24

Page 23 of 36

Reference

2

[1] Y.C. Wang, F. Wang, T.M. Sun, J. Wang, Redox-responsive nanoparticles from the single

3

disulfide bond-bridged block copolymer as drug carriers for overcoming multidrug resistance

4

in cancer cells, Bioconjugate chemistry, 22 (2011) 1939-1945.

5

[2] Y. Su, Y. Hu, Y. Du, X. Huang, J. He, J. You, H. Yuan, F. Hu, Redox-Responsive

6

Polymer-Drug Conjugates Based on Doxorubicin and Chitosan Oligosaccharide-g-stearic

7

Acid for Cancer Therapy, Molecular pharmaceutics, 12 (2015) 1193-1202.

8

[3] A.A. Barba, A. Dalmoro, M. d'Amore, G. Lamberti, In vitro dissolution of pH sensitive

9

microparticles for colon-specific drug delivery, Pharmaceutical development and technology,

us

cr

ip t

1

(2012).

11

[4] Z. Amoozgar, J. Park, Q. Lin, Y. Yeo, Low molecular-weight chitosan as a pH-sensitive

12

stealth coating for tumor-specific drug delivery, Molecular pharmaceutics, 9 (2012) 1262-

13

1270.

14

[5] V. Balamuralidhara, T.M. Pramodkumar, N. Srujana, M.P. Venkatesh, N. Vishal Gupta,

15

K.L. Krishna, H.V. Gangadharappa, pH Sensitive drug delivery systems: A review, American

16

Journal of Drug Discovery and Development, 1 (2011) 24-48.

17

[6] G.S. Kwon, T. Okano, Polymeric micelles as new drug carriers, Advanced drug delivery

18

reviews, 21 (1996) 107-116.

19

[7] M. Hruby, C. Konak, K. Ulbrich, Polymeric micellar pH-sensitive drug delivery system

20

for doxorubicin, Journal of controlled release : official journal of the Controlled Release

21

Society, 103 (2005) 137-148.

22

[8] K. Kataoka, T. Matsumoto, M. Yokoyama, T. Okano, Y. Sakurai, S. Fukushima, K.

23

Okamoto, G.S. Kwon, Doxorubicin-loaded poly(ethylene glycol)-poly(beta-benzyl-L-

24

aspartate) copolymer micelles: their pharmaceutical characteristics and biological

25

significance, Journal of controlled release : official journal of the Controlled Release Society,

26

64 (2000) 143-153.

27

[9] J. Yue, S. Liu, R. Wang, X. Hu, Z. Xie, Y. Huang, X. Jing, Transferrin-Conjugated

28

Micelles: Enhanced Accumulation and Antitumor Effect for Transferrin-Receptor-

29

Overexpressing Cancer Models, Molecular pharmaceutics, (2012).

30

[10] D. Chen, P. Song, F. Jiang, X. Meng, W. Sui, C. Shu, L.J. Wan, pH-responsive

31

mechanism of a deoxycholic acid and folate comodified chitosan micelle under cancerous

32

environment, The journal of physical chemistry. B, 117 (2013) 1261-1268.

Ac ce p

te

d

M

an

10

25

Page 24 of 36

[11] Y.Z. Du, L.L. Cai, J. Li, M.D. Zhao, F.Y. Chen, H. Yuan, F.Q. Hu, Receptor-mediated

2

gene delivery by folic acid-modified stearic acid-grafted chitosan micelles, International

3

journal of nanomedicine, 6 (2011) 1559-1568.

4

[12] R. Yang, F. Meng, S. Ma, F. Huang, H. Liu, Z. Zhong, Galactose-decorated cross-linked

5

biodegradable poly(ethylene glycol)-b-poly(epsilon-caprolactone) block copolymer micelles

6

for enhanced hepatoma-targeting delivery of paclitaxel, Biomacromolecules, 12 (2011) 3047-

7

3055.

8

[13] S.S. Yu, C.M. Lau, W.J. Barham, H.M. Onishko, C.E. Nelson, H. Li, C.A. Smith, F.E.

9

Yull, C.L. Duvall, T.D. Giorgio, Macrophage-specific RNA interference targeting via "click",

us

cr

ip t

1

mannosylated polymeric micelles, Molecular pharmaceutics, 10 (2013) 975-987.

11

[14] P. Yousefpour, F. Atyabi, E. Vasheghani-Farahani, A.A. Movahedi, R. Dinarvand,

12

Targeted delivery of doxorubicin-utilizing chitosan nanoparticles surface-functionalized with

13

anti-Her2 trastuzumab, International journal of nanomedicine, 6 (2011) 1977-1990.

14

[15] F. Hoppe-Seiler, Chitin and chitosan, Ber Dtsch Chem Ges, 27 (1994) 3329-3331.

15

[16] A. Domard, N. Cartier, Glucosamine oligomers: 4. Solid state-crystallization and

16

sustained dissolution, International journal of biological macromolecules, 14 (1992) 100-106.

17

[17] R. Hejazi, M. Amiji, Chitosan-based gastrointestinal delivery systems, Journal of

18

controlled release : official journal of the Controlled Release Society, 89 (2003) 151-165.

19

[18] L. Chen, Y. Du, Z. Tian, L. Sun, Effect of the degree of deacetylation and the

20

substitution of carboxymethyl chitosan on its aggregation behavior, Journal of Polymer

21

Science Part B: Polymer Physics, 43 (2005) 296-305.

22

[19] X.-G. Chen, H.-J. Park, Chemical characteristics of O-carboxymethyl chitosans related

23

to the preparation conditions, Carbohydrate polymers, 53 (2003) 355-359.

24

[20] X. Fei Liu, Y. Lin Guan, D. Zhi Yang, Z. Li, K. De Yao, Antibacterial action of chitosan

25

and carboxymethylated chitosan, Journal of Applied Polymer Science, 79 (2001) 1324-1335.

26

[21] F. Arcamone, Doxorubicin Academic Press, New York, 1981.

27

[22] D.D. Von Hoff, M. Rozencweig, M. Piccart, The cardiotoxicity of anticancer agents,

28

Seminars in oncology, 9 (1982) 23-33.

29

[23] L.J. Goldstein, H. Galski, A. Fojo, M. Willingham, S.L. Lai, A. Gazdar, R. Pirker, A.

30

Green, W. Crist, G.M. Brodeur, et al., Expression of a multidrug resistance gene in human

31

cancers, Journal of the National Cancer Institute, 81 (1989) 116-124.

32

[24] L. Qiu, X. Wu, Y. Jin, Doxorubicin-Loaded Polymeric Micelles Based on Amphiphilic

33

Polyphosphazenes with Poly(N-isopropylacrylamide-co-N,N-dimethylacrylamide) and Ethyl

Ac ce p

te

d

M

an

10

26

Page 25 of 36

Glycinate as Side Groups: Synthesis, Preparation and In Vitro Evaluation, Pharmaceutical

2

research, 26 (2009) 946-957.

3

[25] D. Kim, Z.G. Gao, E.S. Lee, Y.H. Bae, In vivo evaluation of doxorubicin-loaded

4

polymeric micelles targeting folate receptors and early endosomal pH in drug-resistant

5

ovarian cancer, Molecular pharmaceutics, 6 (2009) 1353-1362.

6

[26] S. Cammas, T. Matsumoto, T. Okano, Y. Sakurai, K. Kataoka, Design of functional

7

polymeric micelles as site-specific drug vehicles based on poly (α-hydroxy ethylene oxide-

8

co-β-benzyl l-aspartate) block copolymers, Materials Science and Engineering: C, 4 (1997)

9

241-247.

us

cr

ip t

1

[27] K.N. Prasad, B. Kumar, X.D. Yan, A.J. Hanson, W.C. Cole, Alpha-tocopheryl succinate,

11

the most effective form of vitamin E for adjuvant cancer treatment: a review, Journal of the

12

American College of Nutrition, 22 (2003) 108-117.

13

[28] J.M. Turley, F.W. Ruscetti, S.J. Kim, T. Fu, F.V. Gou, M.C. Birchenall-Roberts,

14

Vitamin E succinate inhibits proliferation of BT-20 human breast cancer cells: increased

15

binding of cyclin A negatively regulates E2F transactivation activity, Cancer research, 57

16

(1997) 2668-2675.

17

[29] J.M. Turley, T. Fu, F.W. Ruscetti, J.A. Mikovits, D.C. Bertolette, 3rd, M.C. Birchenall-

18

Roberts, Vitamin E succinate induces Fas-mediated apoptosis in estrogen receptor-negative

19

human breast cancer cells, Cancer research, 57 (1997) 881-890.

20

[30] M.W. Fariss, M.B. Fortuna, C.K. Everett, J.D. Smith, D.F. Trent, Z. Djuric, The

21

selective antiproliferative effects of alpha-tocopheryl hemisuccinate and cholesteryl

22

hemisuccinate on murine leukemia cells result from the action of the intact compounds,

23

Cancer research, 54 (1994) 3346-3351.

24

[31] M. Muthiah, H.L. Che, S. Kalash, J. Jo, S.Y. Choi, W.J. Kim, C.S. Cho, J.Y. Lee, I.K.

25

Park, Formulation of glutathione responsive anti-proliferative nanoparticles from thiolated

26

Akt1 siRNA and disulfide-crosslinked PEI for efficient anti-cancer gene therapy, Colloids

27

and surfaces. B, Biointerfaces, 126 (2015) 322-327.

28

[32] T.I. Kim, T. Rothmund, T. Kissel, S.W. Kim, Bioreducible polymers with cell

29

penetrating and endosome buffering functionality for gene delivery systems, Journal of

30

controlled release : official journal of the Controlled Release Society, 152 (2011) 110-119.

31

[33] J.P. Nam, D.G. Kim, Y.B. Kim, Y.I. Jeong, M.K. Jang, J.W. Nah, Preparation and NMR

32

spectroscopic characterization of low molecular water soluble O-carboxymethyl chitosan, J

33

Chitin Chitosan, 13 (2008) 105-109.

Ac ce p

te

d

M

an

10

27

Page 26 of 36

[34] H.M. Burt, X. Zhang, P. Toleikis, L. Embree, W.L. Hunter, Development of copolymers

2

of poly(d,l-lactide) and methoxypolyethylene glycol as micellar carriers of paclitaxel,

3

Colloids and Surfaces B: Biointerfaces, 16 (1999) 161-171.

4

[35] S.C. Park, J.Y. Kim, C. Jeong, S. Yoo, K.S. Hahm, Y. Park, A plausible mode of action

5

of pseudin-2, an antimicrobial peptide from Pseudis paradoxa, Biochimica et biophysica acta,

6

1808 (2011) 171-182.

7

[36] J.W. Choi, E. Kang, O.J. Kwon, T.J. Yun, H.K. Park, P.H. Kim, S.W. Kim, J.H. Kim,

8

C.O. Yun, Local sustained delivery of oncolytic adenovirus with injectable alginate gel for

9

cancer virotherapy, Gene therapy, (2013).

us

cr

ip t

1

[37] G. Arya, M. Vandana, S. Acharya, S.K. Sahoo, Enhanced antiproliferative activity of

11

Herceptin (HER2)-conjugated gemcitabine-loaded chitosan nanoparticle in pancreatic cancer

12

therapy, Nanomedicine : nanotechnology, biology, and medicine, 7 (2011) 859-870.

13

[38] Y. Tao, J. Han, H. Dou, Paclitaxel-loaded tocopheryl succinate-conjugated chitosan

14

oligosaccharide nanoparticles for synergistic chemotherapy, Journal of Materials Chemistry,

15

22 (2012) 8930.

16

[39] H. Yuan, L.J. Lu, Y.Z. Du, F.Q. Hu, Stearic acid-g-chitosan polymeric micelle for oral

17

drug delivery: in vitro transport and in vivo absorption, Molecular pharmaceutics, 8 (2011)

18

225-238.

19

[40] F.Q. Hu, Y.Y. Zhang, J. You, H. Yuan, Y.Z. Du, pH triggered doxorubicin delivery of

20

PEGylated glycolipid conjugate micelles for tumor targeting therapy, Molecular

21

pharmaceutics, 9 (2012) 2469-2478.

22

[41] K.R. West, S. Otto, Reversible covalent chemistry in drug delivery, Current drug

23

discovery technologies, 2 (2005) 123-160.

24

[42] W. Tai, R. Mahato, K. Cheng, The role of HER2 in cancer therapy and targeted drug

25

delivery, Journal of Controlled Release, 146 (2010) 264-275.

27

M

d

te

Ac ce p

26

an

10

28

28

Page 27 of 36

1

Figure Legends

2

10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32

ip t

cr

9

Figure 3. DOX release profile of TOC-DOX and HPTOC-DOX micelles against various pH conditions. The pH conditions maintained at pH 7.4 during 24 h from 30 min and then decreased at pH 6.2 (A) or pH 5.2 (B) until 30 h. The results are presented as mean ± SD (n=3).

us

8

Figure 4. Localization of various polymeric micelles in SK-Br3 cells. SK-Br3 cells were incubated with DOX, TOC-DOX, and HPTOC-DOX and then were observed in confocal laser scanning microscopy. SK-Br3 cells were pre incubated with LysoTracker and Hoechst 33258 to confirm intracellular localization.

an

7

M

6

Figure 2. Particle size and distribution of various polymeric micelles. A) Particle size of TOC1, TOC1-DOX, HPTOC1-DOX, TOC3-DOX, TOC3-DOX, and HPTOC3-DOX. The results are presented as mean ± SD (n=3). B) Particle size distribution of HPTOC3-DOX.

Figure 5. A) In vitro cell cytotoxicity of DOX, TOC-DOX, and HPTOC-DOX against HEK 293 cells (a) or HaCa T cells (b). B) In vitro hemolytic assay of TOC-DOX and HPTOCDOX with different micelles concentration (0.625 to 5 µg/mL) against rat red blood cells (RBCs) from an inbred rat. D-PBS and 0.1% triton X were used the negative control and positive control. The results are presented as mean ± SD (n=5)

d

5

te

4

Figure 1. 1H-NMR spectrum of HPTOC-DOX. Concentration of HPTOC-DOX is 5 mg/0.5 mL in D2O.

Figure 6. In vitro anti-tumor effect of intermediate structure or final products against SK-Br3 cells. The anti-tumor levels were expressed by percentage of cell viability. A) cell viability of OCMCh, TP, TOC1, and TOC3; B) cell viability of DOX, TOC1-DOX, HPTOC1-DOX, TOC3-DOX, HPTOC3-DOX. The results are presented as mean ± SD (n=5)

Ac ce p

3

Figure 7. Therapeutic efficacy and characterization of systemically injected TOC-DOX or HPTOC-DOX micelles in HER2 overexpressed SK-Br3 subcutaneous tumor models. A) Inhibition of tumor growth by intravenous injection of TOC-DOX or HPTOC-DOX micelles (20 mg/kg).*P

Targeting delivery of tocopherol and doxorubicin grafted-chitosan polymeric micelles for cancer therapy: In vitro and in vivo evaluation.

In this study, we report the development of a novel, redox-sensitive chitosan-based targeted drug delivery system, containing two drugs. We determined...
3MB Sizes 0 Downloads 7 Views