Nucleus-targeted, echogenic polymersomes for delivering a cancer

Aug 31, 2018 - Search; Citation; Subject. Search in: ..... Published online 31 August 2018. + .... This website uses cookies to improve your user expe...
0 downloads 0 Views 3MB Size
Subscriber access provided by Kaohsiung Medical University

Article

Nucleus-targeted, echogenic polymersomes for delivering a cancer stemness inhibitor to pancreatic cancer cells Fataneh Karandish, Babak Mamnoon, Li Feng, Manas K. Haldar, Lang Xia, Kara N Gange, Seungyong You, Yongki Choi, Kausik Sarkar, and Sanku Mallik Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.8b01133 • Publication Date (Web): 31 Aug 2018 Downloaded from http://pubs.acs.org on August 31, 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 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

Nucleus-targeted,

echogenic

polymersomes

for

2

delivering a cancer stemness inhibitor to pancreatic

3

cancer cells

4

Fataneh Karandish†, Babak Mamnoon†, Li Feng†, Manas K. Haldar†, Lang Xiaᶩ, Kara N.

5

Gangeᶨ, Seungyong You‡, Yongki Choi‡, Kausik Sarkarᶩ, and Sanku Mallik†*

6



7



8

Washington DC 20052

9

ᶨDepartment of Health, Exercise, and Nutrition Sciences, North Dakota State University, Fargo,

Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108

Department of Mechanical and Aerospace Engineering, The George Washington University,

10

ND 58108

11



12

AUTHOR ADDRESS

13

Corresponding Author

14

*E-mail: [email protected]. Phone: 701-231-7888. Fax: 701-231-7831.

15

Present Address

16

Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota

17

58108.

Department of Physics, North Dakota State University, Fargo, ND 58108

ACS Paragon Plus Environment

1

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 35

18

KEYWORDS: Nuclear-delivery, targeted drug delivery, dexamethasone, BBI608, napabucasin,

19

echogenic, polymersomes.

20 21

ABSTRACT

22

Chemotherapeutic agents for treating cancers show considerable side effects, toxicity, and drug

23

resistance. To mitigate the problems, we designed nucleus-targeted, echogenic, stimuli-responsive

24

polymeric vesicles (polymersomes) to transport and subsequently release the encapsulated anti-

25

cancer drugs within the nuclei of pancreatic cancer cells. We synthesized an alkyne-

26

dexamethasone derivative and conjugated it to N3–polyethylene glycol (PEG)–polylactic acid

27

(PLA) copolymer employing the Cu2+ catalyzed “Click” reaction. We prepared polymersomes

28

from the dexamethasone-PEG-PLA conjugate along with a synthesized stimuli-responsive

29

polymer PEG–S–S–PLA. The dexamethasone group dilates the nuclear pore complexes and

30

transports the vesicles to the nuclei. We designed the polymersomes to release the encapsulated

31

drugs in the presence of a high concentration of reducing agents in the nuclei of pancreatic cancer

32

cells. We observed that the nucleus-targeted, stimuli-responsive polymersomes released 70% of

33

encapsulated contents in the nucleus-mimicking environment in 80 minutes. We encapsulated the

34

cancer stemness inhibitor BBI608 in the vesicles and observed that the BBI608 encapsulated

35

polymersomes reduced the viability of the BxPC3 cells to 43% in three-dimensional spheroid

36

cultures. The polymersomes were prepared following a special protocol so that they scatter

37

ultrasound, allowing imaging by a medical ultrasound scanner. Therefore, these echogenic,

38

targeted, stimuli-responsive, drug-encapsulated polymersomes have the potential for trackable,

39

targeted carrier of chemotherapeutic drugs to cancer cell nuclei.

40

ACS Paragon Plus Environment

2

Page 3 of 35 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

41

Biomacromolecules

INTRODUCTION

42

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of death in the United

43

States with the approximate five-year survival rate of less than 10%.1 Pancreatic cancer treatment

44

is complicated because of invasiveness, rapid metastasis, and the complex nature of the disease.

45

Alteration of different genes, aberrant biochemical pathways, epithelial to mesenchymal transition

46

(EMT), hypoxic tumor microenvironment, and the subpopulation of cancer stem cells lead to

47

recurrence and drug resistance.2 Considering that pancreatic cancer shows early invasion and

48

metastasis, there is an urgent need for developing new treatment of this devastating disease. In the

49

solid tumor tissue, EMT leads to a subpopulation of cancer stem cells, which can initiate a tumor,

50

self-renew, and increase resistance to chemotherapeutic drugs.3 The small molecule BBI608

51

(napabucasin) inhibits signal transducer and activator of transcription 3 (STAT3), reduces the

52

expression of cancer stemness markers, inhibits cell proliferation, blocks spherogensis and

53

apoptosis in both cancer stem cells, and non-stem cells.2-4 BBI608 is currently in the Phase III

54

clinical trials as an adjuvant therapy for a variety of solid tumors, including pancreatic cancer

55

(www.clinicaltrials.gov). STAT3 controls many oncogenic pathways and is activated in different

56

type of cancers such as breast, ovarian, colorectal, etc.4

57

Targeted, stimuli-responsive carriers enhance therapeutic efficacy and reduce toxicity by

58

selectively delivering the cytotoxic chemotherapeutic drugs to cancerous tissues.5-7 Polymersomes

59

are vesicles prepared from synthetic, amphiphilic block copolymers. Because of higher molecular

60

weights, polymersomes have enhanced stability and mechanical robustness compared to

61

liposomes, micelles, and polymer micelles.8

62

hydrophobic drugs, and the aqueous core incorporates the hydrophilic molecules, enabling

63

simultaneous delivery of both drugs.9 Conjugation of antibodies, small molecules or peptides

The bilayer of the polymersomes encapsulate

ACS Paragon Plus Environment

3

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 35

64

enables active targeting of the vesicles to the cancer cells.10, 11 However, due to increased stability,

65

the polymer bilayer usually requires a stimulus to rapidly release the encapsulated anticancer

66

drugs.12 Polymersomes responsive to pH 13, heat 14, hypoxia 15, and light 16 has been used to deliver

67

chemotherapeutic drugs to the cancer cells. Multifunctional polymersomes with drug delivery and

68

simultaneous imaging capability are also reported.17, 18 The polymeric nanoparticles accumulate in

69

the tumor tissues by the enhanced permeation and retention effect 19; however, for efficient cellular

70

internalization, specific ligands are necessary on the vesicle surface.

71

Nuclear pore complex is a large multi-protein assembly, which spans the nuclear envelope.

72

Approximately 2000 nuclear pore complexes exist in the nuclear envelope. Dexamethasone is a

73

synthetic steroid that dilates the nuclear pore complex from 38 nm to 300 nm.20, 21 Glutathione

74

(GSH), the abundant cellular reducing agent, is a tripeptide consisting of glutamic acid, cysteine,

75

and glycine. GSH is involved in several important processed in the cell nuclei, such as

76

transcription, DNA replication, nuclear protein import and export, and chromatin stability.

77

High levels of GSH is related to increased cell proliferation. 23 24

22, 23

78

Herein, we report nucleus-targeted, echogenic, redox-sensitive polymersomes to deliver the

79

cancer stemness inhibitor BBI608 to pancreatic cancer cells. We used dexamethasone as a

80

targeting group to dilate the nuclear pore complexes

81

nucleus. In the synthesized copolymer, we used polyethylene glycol (PEG) as the hydrophilic

82

block and polylactic acid (PLA) as the hydrophobic block. Incorporation polyethylene glycol

83

polymer (PEGylation) in the polymersomes composition reduces the interactions of nanoparticle

84

with circulating proteins.25 Polylactic acid (PLA) is the hydrophobic part of the polymer and has

85

a degradation time of more than one week in aqueous solutions at 37 oC. 26, 27 The two polymer

86

blocks were linked by the reduction sensitive disulfide linker.

19

and deliver the polymersomes inside the

Several literature reports

ACS Paragon Plus Environment

4

Page 5 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

87

demonstrate the effectiveness of the disulfide bonds to deliver the encapsulated payloads under

88

increased cellular reducing agent concentration

89

dexamethasone on the surface, internalize in the nuclei of pancreatic cancer cells. The high

90

reducing agent concentration in the nuclei

91

compromises the polymersome structure, and rapidly releases the encapsulated drug. We observed

92

that nucleus-targeted polymersomes encapsulating BBI608 significantly (p < 0.05) decreased the

93

viability of the BxPC3 cells compared to control and the non-targeted vesicles. The polymersomes

94

were shown to be responsive to ultrasound offering possibility of concurrent ultrasound imaging

95

of the cancerous tumors. We have used a preparation protocol incorporating lyophilization in the

96

presence of mannitol that has proved previously effective in rendering liposomes

97

polymersomes 40 echogenic.

28-31

32, 33

We observed that the vesicles, presenting

cleaves the disulfide bonds of the polymers,

34-39

and

98 99

MATERIALS AND METHODS

100

All chemicals and solvents were purchased either from VWR International or TCI America and

101

used as received.

102

Synthesis of alkyne-dexamethasone. Dexamethasone (100 mg, 0.26 mmol) was dissolved in

103

anhydrous pyridine (2 mL) in a round bottom flask and stirred on ice (0 °C) with methane sulfonyl

104

chloride (250 µL, 3.2 mmol) under nitrogen for four hours. Then, an additional amount of methane

105

sulfonyl chloride (18 µL, 0.23 mmol) was added, and the reaction continued for an additional hour.

106

After five hours of stirring under nitrogen, 40 mL of ice water was added to precipitate the product.

107

The precipitate was filtered and washed with 40 mL of additional ice water. The crude product

108

was purified by recrystallization from tetrahydrofuran (THF) twice to afford the pure product. The

109

product (0.026g, 0.06 mmol) and propargylamine (0.10 mL, 1.561 mmol), was stirred in 800 µL

ACS Paragon Plus Environment

5

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 35

110

dimethylformamide (DMF) at 65 °C under nitrogen gas for two hours. After cooling to room

111

temperature, the reaction mixture was added dropwise to cold water and centrifuged at 425 g for

112

10 minutes. The clear supernatant was decanted, and the precipitate was washed again with water,

113

dried under vacuum. The pure product (16 mg, 62%) was characterized by 1H NMR spectroscopy

114

(Supporting Information). 1H NMR (400 MHz, chloroform-d) δ ppm: 7.41 (d, 1H), 6.40 (d, 1H),

115

6.20 (d, 1H), 4.37 (d, 2H), 4.32 (t, 1H), 3.41 (s, 1H), 2.43 (t, 2H), 2.38 (t, 2H), 1.70 (d, 2H), 1.56

116

(s, 3H), 1.06 (s, 3H), 0.95 (s, 3H). HRMS calcd. for C25 H32FNO4: 429.2315. Observed: 429.2306.

117 118

Figure 1. Structure of the synthesized alkyne conjugated dexamethasone

119

Synthesis of dexamethasone–PEG1900–PLA8000 polymer conjugate. We synthesized the N3–

120

PEG1900–PLA8000 polymer employing a protocol developed in our laboratory (Supporting

121

Information). The alkyne-dexamethasone was reacted with the N3–PEG1900–PLA8000 polymer

122

using the [2+3]-cycloaddition reaction.41 Briefly, we prepared the copper (II) complex by mixing

123

the CuSO4 (71.3 mg in 3 mL water, 0.53 mmol) and pentamethyl diethylenetriamine (440 µL in 3

124

mL water, 2 mmol). The N3–PEG1900–PLA8000 polymer (20 mg) and alkyne-dexamethasone (2

125

mg) were dissolved in 6 mL tetrahydrofuran (THF), 400 L of the 53 mM copper complex and

126

400 L of 53 mM aqueous sodium ascorbate solution were added, and the reaction mixture was

127

stirred at room temperature for 24 hours. The reaction mixture was added dropwise to cold ether

128

and centrifuged at 425 g for 2 minutes. The clear supernatant was decanted, and the precipitate

ACS Paragon Plus Environment

6

Page 7 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

129

was washed with water three times by vortex and centrifuge, then dried under vacuum. 1H NMR

130

(400 MHz, chloroform-d) δ ppm: 6.34 (d, 1H), 6.15 (d, 1H), 5.18 (q, 1 H), 4.37 (d, 2H), 3.66 (t, 4

131

H), 2.52 (t, 2H), 2.30 (t, 2H), 1.60 (d, 3 H). From the 1H NMR spectrum, we calculated the

132

conversion yield for the dexamethasone coupling reaction to be 39% (Supporting Information).

133

134 135

Scheme 1. [2+3]-Cycloaddition reaction of N3–PEG1900–PLA8000 polymer and alkyne-

136

dexamethasone.

137 138

Preparation of nucleus targeted polymersomes encapsulating BBI608. Polymersomes were

139

prepared by the solvent exchange method42 with PEG1900–S–S–PLA6000

140

PEG1900–PLA8000 polymer, and 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-lissamine

141

rhodamine B sulfonyl ammonium salt (fluorescent dye, LR, Avanti Polar Lipids) with a molar

43

, dexamethasone–

ACS Paragon Plus Environment

7

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 35

142

ratio of 35:60:5, respectively (Figure 2). The polymers were dissolved in THF (9 mg/mL), BBI608

143

in THF (3 mg/mL), and LR in chloroform (0.01 mg/mL). First, a rotary evaporator was used to

144

evaporate the chloroform from LR lipid to form a thin layer film. The THF solutions of the

145

polymers and BBI608 were added to the thin film. The resultant fluorescent THF solution was

146

added dropwise to an aqueous HEPES buffer (10 mM, pH 7.4) and stirred for 45 minutes. To

147

remove the THF, a gentle stream of air was passed through the mixture for 45 minutes. The

148

polymersomes formed were bath sonicated for 60 minutes (Symphony 117 V, 60 Hz, Power level

149

9). The polymersomes (1 mg/mL) were passed through a SephadexTM G-100 size exclusion

150

column to remove the unencapsulated drug. Drug loading efficacies (DLE) of the polymersomes

151

were determined using UV−Vis spectroscopy. After passing through the size-exclusion column,

152

the absorption of the polymersomes was recorded at 235 nm to calculate the loading efficiency.

ACS Paragon Plus Environment

8

Page 9 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

153 154

Figure 2. The structure of synthesized polymers PEG1900–S–S–PLA6000, dexamethasone–

155

PEG1900–PLA8000 polymer conjugate, and the commercially available fluorescent lipid 1,2-

156

dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-lissamine rhodamine B sulfonyl ammonium

157

salt.

158

Preparation of control polymersomes. The control polymersomes were prepared following the

159

same method as the nucleus-targeted vesicles. PEG1900–S–S–PLA6000, N3–PEG1900–PLA8000, and

160

lissamine rhodamine (LR) were used in the molar ratio of 35:60:5, respectively. The PEG1900–S–

161

S–PLA6000 and N3–PEG1900–PLA8000 polymers were dissolved in THF. The polymer solution was

162

added slowly to the thin film of the LR dye, and then the mixture was added dropwise to a stirred

163

10 mM HEPES buffer (pH 7.4). The polymersome solutions were stirred for 45 minutes at room

ACS Paragon Plus Environment

9

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 35

164

temperature, and then air was passed for 45 minutes through the mixture. The polymersomes were

165

sonicated for 60 minutes (Symphony 117 V, 60 Hz). Subsequently, the polymersomes were passed

166

through a Sephadex G100 (GE Healthcare) size exclusion column to collect lissamine rhodamine

167

dye incorporated polymersomes. These polymersomes were used as a control for the cell viability

168

assays.

169 170

Polymersomes Size Analysis. The nucleus-targeted polymersomes encapsulating BBI608 and

171

control polymersomes were characterized by dynamic light scattering at 90o using a Zeta Sizer

172

Nano ZS 90 (Malvern Instrument). Polymersomes were equilibrated for 120 seconds, and five

173

measurements were recorded with 10 repeats each.

174 175

Transmission Electron Microscopy (TEM). Samples were prepared on 300 mesh copper grids

176

with a formvar-carbon support film. Grids were pretreated with 1% poly-L-lysine and air-dried;

177

5 µL of the polymersome suspension was added and allowed to stand 1 min, then wicked off with

178

a filter paper. Negative staining was performed using 0.1% phosphotungstic acid for 2 minutes,

179

then wicking off and air-dried before observation and imaging in a JEOL JEM-2100 LaB6

180

transmission electron microscope.

181

Atomic Force Microscopic (AFM) Imaging. The size and morphology of polymersomes were

182

characterized using atomic force microscopy. Polymersomes (1 mg/mL) were diluted (20X) in

183

HEPES buffer (pH 7.4, 10 mM). Polymersomes were dropped on silica substrates, incubated for a

184

minute and extra liquid dried by an air blowgun. The AFM measurements were performed in non-

185

contact mode (resonance frequency of 145 kHz and a scanning rate of 1.3 Hz) using an NT-MDT

186

INTEGRA instrument (NT-MDT America). The scanning areas were 5 × 5 μm² at the resolution

ACS Paragon Plus Environment

10

Page 11 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

187

of 512 points per line, respectively. The images were flattened to eliminate the background low-

188

frequency noise and tilt from the surface using all unmasked portions of scan lines to calculate

189

individual least-square fit polynomials for each line.

190 191

Redox-triggered Release studies. Polymersomes encapsulating 20 µM calcein dye were

192

prepared to perform the release study. The experiments were conducted by the quenching method

193

44

194

chloride (10 mM) was used to quench the fluorescence from the unencapsulated calcein outside

195

the vesicles. We used 20 μL of calcein-encapsulated polymersomes (total polymer concentration:

196

1 mg/mL) in 10 mM HEPES buffer (180 μL, pH =7.4) in a 96-well plate. The release from the

197

polymersomes were monitored in the presence of 10 mM glutathione for 40 minutes; subsequently,

198

the concentration was increased to 50 mM, and the release was monitored for an additional 40

199

minutes using (excitation: 495 nm, emission: 515 nm) employing a fluorescence microplate reader

200

(Spectramax M5, Molecular Devices). The amount of calcein release form polymersomes were

201

calculated employing this equation:

202

Percent Release = [(Emission Intensity after 80 min – Initial Intensity before treatment)/Initial

203

Intensity before treatment] ×100.

using cobalt chloride as the quencher and glutathione as the reducing agent. The cobalt (II)

204 205

Preparation and characterization of echogenic polymersomes. Polymersomes were prepared

206

by dissolving the PEG1900–S–S–PLA6000 and N3–PEG1900–PLA8000 polymers with the molar ratio

207

of 40:60 (total 5 mg in 1 mL of THF), respectively. Then, the polymer mixture was dropwise added

208

to 0.32 M mannitol (weak cryoprotectant) prepared in HEPES buffer (10 mM, pH 7.4).

209

Subsequently, THF was evaporated for 45 min by passing air through the solution. Then,

ACS Paragon Plus Environment

11

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 35

210

polymersomes were sonicated for 60 minutes (Symphony 117 V, 60 Hz). To make the

211

polymersomes echogenic, they were subjected to three freeze (-80 oC, 24 h) and thaw (60 °C)

212

cycles. Finally, the polymersomes were lyophilized (Labconco freeze dryer) and reconstituted in

213

HEPES buffer (10 mM, pH 7.4) for further experiments.

214 215

Ultrasound Experimental setup to measure scattering in echogenic polymersomes. Two

216

spherically focused transducers (each having a central frequency of 2.25 MHz/5 MHz/10 MHz,

217

Figure 3) with the same specifications (V310-SU, Olympus NDT) were employed for scattering

218

measurements. The transmitting and receiving transducers were placed perpendicularly by two

219

separate linear stages (433 series, 360-90, Newport) and immersed in a bigger water tank filled

220

with DI water. A 20-mL syringe served as a sample chamber, in which polymersome suspension

221

was injected. A function generator (Model AFG 3251; Tektronix) was utilized to generate a 32-

222

cycle sinusoidal pulse of 5 MHz frequency at a PRF of 100 Hz. These signals were then amplified

223

using a 55dB power amplifier (Model A-300, ENI) and sent to the transmitting transducer. The

224

input signals were scattered by the polymersomes inside the focal volume of the transducer. The

225

scattered signals were received by the receiving transducer connected to a pulser/receiver

226

(DPR300, 475v, JSR) in the through mode with a 27-dB gain. The output signals were then

227

transmitted to an oscilloscope (TDS2012, Tektronix) for real-time visualization. The output

228

voltage-time RF signals were obtained by the oscilloscope by averaging over every 64 sequences.

229

The data from the oscilloscope were finally transmitted and saved onto a desktop computer using

230

the software Signal Express Tektronix Edition (version 2.5.1, Labview NI). In time-dependent

231

scattering experiments, all the setups and procedures were the same except that the data were

232

recorded over a 20-minute period (corresponding to about 344 acquisitions). As for the degassed

ACS Paragon Plus Environment

12

Page 13 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

233

experiment, all the setups and procedures were still the same, only difference being that the PBS

234

solution had been degassed using a vacuum pump.

235

236 237

Figure 3. Experimental setups for the ultrasound scattering measurements from the echogenic

238

polymersomes.

239 240

Echogenic polymersomes’ experimental procedure and data reduction. In a scattering

241

experiment, the suspension was made by reconstituting the dry powder in phosphate buffered

242

saline (PBS) solution to obtain a concentration of 10 μg polymer/mL. We injected 20 mL of the

243

resulting suspension into the sample chamber. The measurement was repeated five times to

244

guarantee the reliability of the experimental data. The measurement of the control signal, i.e.,

245

without polymersomes and the responses due to the polymersomes were acquired by the

ACS Paragon Plus Environment

13

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 35

246

procedures above. The Fourier transform of the signal was performed using a Matlab program to

247

get the average scattered power spectra in the frequency domain (50 voltage time acquisitions were

248

used for averaging). The scattered response was converted into a dB scale by taking a unit

249

reference. Fundamental, second- and sub-harmonic scattered responses were extracted from the

250

power spectrum. The final data is reported as an enhancement over the control. We also checked

251

the scattered response in the presence of 50 mM of glutathione.

252 253

Ultrasound Imaging. Dried polymersomes were reconstituted in 10 mM HEPES buffer, pH 7.4

254

with the concentration of 10 µg/mL. In a 96-well plate, 0.2 mL of polymersomes were dispensed

255

into each well; then the plate was covered with parafilm. Subsequently, an ultrasound gel

256

(Aquasonic 100, Parker Laboratories) was applied, and a 15 MHz linear ultrasound transducer was

257

used for the imaging experiments employing a Terason t3200 instrument (MediCorp LLC).

258 259

Cell Culture. The pancreatic cancer cell line BxPC3 was purchased from the American Tissue

260

Culture Consortium (ATCC). The cells were maintained in RPMI 1640 medium (without phenol

261

red) supplemented with 1% v/v antibiotics (penicillin and streptomycin) and 10% v/v fetal bovine

262

serum. The cell culture flasks were maintained in an incubator at 37 °C in a 5% CO2 atmosphere.

263

Nuclear Uptake Studies. The BxPC3 cells (3×10³) were seeded in a 12-well tissue culture plate

264

for 24 hours before the experiment. Once the culture is 80-90% confluent, the nucleus-targeted (20

265

µL) and non-targeted (20 µL) polymersomes were incubated with the cells for 3 hours.

266

Subsequently, the cell culture media and polymersomes were removed, and the cells were washed

267

twice with Hanks’ balanced salt solution (HBSS) to remove the non-internalized vesicles. The cell

ACS Paragon Plus Environment

14

Page 15 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

268

nuclei were stained with HOESCHT 33342 dye (Enzo Life Sciences, 1:1000 dilution) and imaged

269

using the 20X objective of a Leica fluorescence microscope.

270 271

Cell Viability in Monolayer Cultures. To evaluate the efficacy of the nucleus-targeted

272

polymersomes on the human pancreatic cancer cells (BxPC3), the Alamar Blue assay was

273

performed. The BxPC3 cells were seeded at a density of 10³/well in a 96-well tissue culture plate

274

and were allowed to grow until 80−95% confluent. The plate was divided into four groups: control,

275

free drug (BBI608), non-targeted polymersomes, and nucleus-targeted polymersomes

276

encapsulating BBI608. The control group did not receive any treatment. Cells treated with BBI608

277

received 1, 4, and 8 μM of free and an equivalent amount of encapsulated drug in nucleus-targeted

278

polymersomes. The cells were treated for 48 hours at 37 °C, in a 5% CO2 atmosphere.

279

Subsequently, the cells were washed with sterile HBSS and replaced with 200 μL of fresh media.

280

Then 20 μL Alamar Blue was added to all the wells and fluorescence were measured after 4 hours.

281

The data presented are normalized to the control.

282 283

Cell Viability in three-dimensional (3D) spheroid cultures. The 24-well 3D petri dishes

284

(Microtissues) were used to prepare BxPC3 spheroids. Briefly, 2% w/v agarose solution was

285

prepared and autoclaved. The BxPC3 cell suspension (104 cells in 60 μL media) was then added

286

to each 3D scaffold. The spheroids were allowed to grow for 7 days. Then scaffolds were divided

287

into four groups: control, non-targeted polymersomes, nucleus-targeted polymersomes

288

encapsulating BBI608, and free drug. Spheroids were treated for 48 hours with the same

289

concentration of drug as used for the monolayer studies. Subsequently, the spheroids were washed

290

with sterile HBSS and then incubated with 100 μL TryPLE (recombinant trypsin, Life

ACS Paragon Plus Environment

15

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 35

291

Technologies) for 10 minutes. The spheroids were removed and subjected to the Alamar Blue

292

assay. The data presented are normalized to the control.

293 294

Results and Discussion

295

Polymer synthesis and polymersome preparation. To prepare nucleus targeted polymersomes,

296

we synthesized an alkyne-dexamethasone conjugate (Figure 1) and linked it to the N3-–PEG1900–

297

PLA8000 polymer (Figure 2 and Supporting Information). The redox-sensitive polymer PEG1900–

298

S–S–PLA6000 (Figure 2) was synthesized as previously reported by our group.

299

dexamethasone–PEG1900–PLA8000 polymer conjugate was synthesized using the Click chemistry

300

[2+3 cycloaddition]. We anticipated that the slightly higher molecular weight of the polymer

301

conjugate would protrude the dexamethasone group from the surface of the vesicles and facilitate

302

the interactions with its receptor. The polymersomes were prepared by the solvent exchange

303

method

304

microscopy (Figure 5), and atomic force microscopy (Figure 6). We encapsulated the stemness

305

gene transcription inhibitor BBI608 (napabucasin) 3 in the polymersomes with an efficiency of 68

306

+ 5%. We observed that the nucleus-targeted polymersomes had a hydrodynamic diameter of 200

307

± 2 nm, with a polydispersity index (PDI) of 0.2 ± 0.02. The average hydrodynamic diameter for

308

the non-targeted polymersomes was 140 ± 3 nm with a PDI of 0.2 ± 0.03. The nucleus-targeted

309

polymersomes were slightly larger than nontargeted polymersomes. We hypothesize that the

310

encapsulation of hydrophobic BBI608 in the polymer bilayer of the vesicles increases the size of

311

vesicles.

312

The dexamethasone–PEG1900–PLA8000 polymer was incorporated into polymersome composition

313

to target the nanoparticles to the cell nucleus. We expected that the dexamethasone on the

42

15, 43, 45

The

and characterized by dynamic light scattering (Figure 4), transmission electron

ACS Paragon Plus Environment

16

Page 17 of 35

21, 46

314

polymersomes would dilate the nuclear pore complex

315

nucleus. The disulfide bond in the redox-sensitive polymer will subsequently be reduced in the

316

nucleus, releasing the encapsulated BBI608.

A

10 8

Intensity%

Intensity%

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

6 4 2 0

0

100

200

300

Size(nm)

317

400

500

16 14 B 12 10 8 6 4 2 0 0 100

and transport the vesicles into the

200 300 Size (nm)

400

500

318

Figure 4. The hydrodynamic diameters of the (A) non-targeted and (B) nucleus-targeted

319

polymersomes encapsulating the stemness inhibitor BBI608, as determined by dynamic light

320

scattering.

321 322

Figure 5. Transmission electron microscopy images (scale bar: 50 nm) of the non-targeted

323

polymersomes.

324

ACS Paragon Plus Environment

17

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 35

325

326 327

Figure 6. Atomic force microscopy (AFM) images of the non-targeted polymersomes. (A, B)

328

Polymersomes, (C, D) Polymersomes treated with 10 mM glutathione for 5 minutes, and (E, F)

329

polymersomes treated with 50 mM GSH for 5 minutes.

330 331

Demonstration of reduction-triggered contents release from the polymersomes and

332

structural characterization. We studied the reduction-triggered release of the dye calcein from

333

the polymersomes as a function of time in the presence of different concentrations of added

334

glutathione (GSH). GSH is an important intracellular reducing agent, comprising of glutamic acid,

335

cysteine, and glycine.47 In tumor cells, the concentration of GSH is about 100-1000 time higher

336

than the extracellular fluids.32, 48 It's increased level is correlated with progression and proliferation

337

of various cancers, such as breast 3, colon

338

We observed that the polymersomes released 45% the encapsulated dye in the presence of 10 mM

339

(mimicking the cytosol) GSH. However, the released increased to 70% with 50 mM GSH

49

, lung 49, pancreas

47

, resistance to chemotherapy.47

ACS Paragon Plus Environment

18

Page 19 of 35

340

(mimicking the nucleus, Figure 7).24 We also observed that both of the releases were rapid. Our

341

atomic force microscopic studies indicated that 10 mM GSH slightly changed the morphology of

342

the polymersomes (Figure 8A); however, 50 mM GSH completely disrupted the vesicle structure

343

(Figure 8B). We checked the size of disruption of polymersomes (with dynamic light scattering)

344

and observed that 50 mM GSH decreased the average diameter of the vesicles from 200 nm (Figure

345

4B) to 100 nm (Figure 8C).

80 [GSH] = 50 mM

Percent Release

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

70 60 50

[GSH] = 10 mM

40 30 20 10 0 0

346 347

20

40

60

80

Time (min) Figure 7. The glutathione-triggered release of the encapsulated calcein from the polymersomes.

348

ACS Paragon Plus Environment

19

Biomacromolecules

C 40 35 30

Intensity%

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 35

25 20 15 10 5 0 0

349

100

200

300

400

500

Size(nm)

350

Figure 8. Structural characterization of the polymersomes after release study employing atomic

351

force microscopy. (A) Polymersomes treated with 10 mM GSH. (B) Polymersomes treated with

352

50 mM GSH. (C) The hydrodynamic diameters of polymersomes after release study, as determined

353

by dynamic light scattering.

354 355

Demonstration of polymersomes’ echogenicity. Polymersomes’ echogenicity was confirmed by

356

a Terason medical ultrasonic imaging system (t3200) using a 15 MHz transducer. We observed

357

that the echogenic polymersomes reflected ultrasound even after a week in the aqueous solution.

358

The ultrasound reflection suggests the presence of air pockets (detailed discussion below) in

359

polymersomes (Figure 9B and C) while control (buffer without any polymersomes) did not show

360

any contrast (Figure 9A).

361

ACS Paragon Plus Environment

20

Page 21 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

362 363

Figure 9. Ultrasound contrast images of the polymersomes at 15 MHz. (A) Control, (B) freshly

364

reconstituted echogenic polymersomes, and (C) echogenic polymersomes after one week in

365

aqueous solution.

366

The scattered power spectra of the polymersomes at three excitation frequencies (2.25, 5,

367

and 10 MHz) are displayed in Figure 10. Power spectra show the responses of the polymersomes

368

as a function of frequencies. Contributions at frequencies other than the excitation frequency

369

indicate nonlinear responses of the polymersomes, which in turn offers the possibility of a

370

nonlinear imaging modality with a potentially better signal to noise ratio 50, 51. Polymersomes are

371

echogenic in aqueous phosphate buffered saline (PBS, pH = 7.4), but not in the degassed solution

372

(Figure 10D). There is a significant nonlinear response, specifically subharmonic, only at 2.25

373

MHz (Figure 10A).

374

Note that although echogenicity of echogenic liposomes (ELIPs), prepared following a

375

particular protocol, has been demonstrated by others and us

376

echogenicity remains unknown. For ELIPs, repeated freeze-thaw cycles followed by lyophilization

377

in the presence of mannitol in the preparation protocol has proved crucial for ensuring echogenicity

378

36, 61

379

that during rehydration generate lipid-coated air-bubbles either inside liposomes or in the bilayer

380

62, 63

36, 52-60

, the exact mechanism of the

. It has been speculated that mannitol, being a weak cryoprotectant, leaves defects in the bilayer

. The bubbles, stabilized against dissolution by the coating64-66, produce strong ultrasound

ACS Paragon Plus Environment

21

Biomacromolecules

381

echoes. The lack of echogenicity in degassed solution as well as the strong nonlinear response

382

verifies the role of bubbles in the acoustic response of echogenic polymersomes.

-50

-80

2.25 MHz

A

Control Polymersomes

-60

Power Spectrum (dB)

Power Spectrum (dB)

-40

-70 -80 -90 -100

-85

B

5 MHz Control Polymersomes

-90 -95 -100 -105

-110 2

4

6

8

-110

10

5

Frequency (MHz)

383

10

15

20

Frequency (MHz)

-80

-90

C

10 MHz

-95

Power Spectrum (dB)

Power Spectrum (dB)

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 35

Control Polymersomes

-100

-105

-85 -90

5 MHz

D

Control polymersomes in degassed solution

-95 -100 -105

-110

384

10

20

30

40

-110

5

10

15

20

Frequency (MHz)

Frequency (MHz)

385

Figure 10. Scattered responses of echogenic polymersomes in PBS at 500 kPa excitation pressure

386

and excitation frequencies of (A) 2.25 MHz, (B) 5 MHz, and (C) 10 MHz. Control is without

387

polymersomes. (D) Scattered response in degassed solution at 500 kPa and 5 MHz.

388 389

Figure 11(A) shows the enhancements at different excitation frequencies. The

390

enhancement in fundamental response is strongest at 5 MHz excitation, whereas the highest

391

subharmonic enhancement appears at 2.25 MHz. The latter indicates the possibility of

ACS Paragon Plus Environment

22

Page 23 of 35

392

polymersome aided subharmonic imaging. 36, 67 We also examined the scattered responses over a

393

20-minute period in Figure 11(B) to investigate the long-term stability of their echogenicity crucial

394

for clinical applications. During the 20-minute sonication, the polymersomes produced an almost

395

constant signal indicating their suitability for use in contrast-enhanced imaging. Polymersomes in

396

degassed solution expectedly did not generate any scattered response.

20

-50 Fundamental Subharmonic 2nd-Harmonic

15

10

5

0

2.25 MHz

5 MHz

10 MHz

Fundamental Response (dB)

Enhancment (dB)

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

B -60

Polymersomes Polymersomes in degassed solution

-70 -80 -90 -100 -110

0

5

10

15

20

Time (min)

397 398

Figure 11. (A) Enhancements in fundamental, sub– and second harmonic scattered responses from

399

the echogenic polymersomes at 500 kPa and 2.25, 5, and 10 MHz.

400

fundamental responses of the echogenic polymersomes in normal (blue trace) and degassed PBS

401

(black trace) at 5 MHz and 500 kPa (N = 5).

(B) Time-dependent

402 403

Glutathione cleaves the disulfide bond destabilizing the bilayer of the polymersomes. We

404

repeated the scattering experiments in the presence of 50 mM GSH. As expected, we observed that

405

the fundamental enhancement decreased by 10 dB, and the subharmonic enhancement decreased

406

by 4 dB (Figure 12) due to the loss of structural integrity of the polymersomes.

ACS Paragon Plus Environment

23

Biomacromolecules

35 Fundamental Sub-Harmonics

30

Enhancement (dB)

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 35

25 20 15 10 5 0

Without Glutathione

With Glutathione

407 408

Figure 12. Fundamental (black bars) and sub-harmonic (red bars) enhancements of echogenic

409

polymersomes at 5 MHz and 500 kPa with and without glutathione.

410 411

Nuclear Uptake Studies. To determine the localization within the pancreatic cancer cells, we

412

prepared the polymersomes incorporating 5% of the DSPE–lissamine rhodamine lipid (structure

413

shown in Figure 2) into the bilayer. We anticipated that the dexamethasone would dilate the nuclear

414

pore complexes and transport the polymersomes inside the nucleus.68 We incubated the BxPC3

415

pancreatic cancer cells with nucleus-targeted and non-targeted polymersomes for 3 hours.

416

Subsequently, the cell nuclei were stained with the HOESCHT 33342 dye and imaged employing

417

a fluorescence microscope (Figure 13). We observed localization of the targeted polymersomes in

418

the nuclei of the BxPC3 cells. We also observed similar results employing the breast cancer cells

419

MCF-7 (Supporting Information).

ACS Paragon Plus Environment

24

Page 25 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

420 421

Figure 13. Cellular uptake studies with the BxPC3 cells. The non-targeted polymersomes (top

422

panel) did not enter the cell nucleus. The targeted polymersomes (bottom panel) were present in

423

the cell nuclei after 3 hours of incubation (indicated by the overlapping blue and red colors in the

424

Merged panel; scale bar: 50 µm).

425

Viability studies in monolayer cultures of pancreatic cancer cells. After validating efficient

426

nuclear localization, we proceeded to determine the effectiveness of the nucleus-targeted, drug-

427

encapsulated polymersomes in the pancreatic cancer cells. The monolayer culture of the BxPC3

428

cells was treated with the nucleus-targeted polymersomes encapsulating BBI608, the non-targeted

429

vesicles, and the free drug for 48 hours. The cell viability was determined by the Alamar Blue

430

assay (Figure 14). We observed that the control polymersomes without the dexamethasone group

431

decreased the cell viability to 50% at 4 M concentration of encapsulated BBI608 (red columns).

432

At this concentration of encapsulated drug, the targeted polymersomes were more effective,

433

decreasing the viability of the BxPC3 cells to 21% (blue column), similar to the un-encapsulated

434

free BBI608 (pink columns). At lower concentrations of encapsulated BBI608 (1 M and 2 M),

ACS Paragon Plus Environment

25

Biomacromolecules

435

both encapsulated and free BBI608 were less effective in killing the cancer cells. We determined

436

the IC50 values of the nucleus-targeted polymersomes encapsulating BBI608, the non-targeted

437

vesicles, and the free drug for the BxPC3 cells (48 hours) to be 1.75 + 0.12 M, 3.20 + 0.47 M,

438

and 0.77 + 0.48 M, respectively (Supporting Information).

Control Non-targeted polymersomes encapsulating BBI608 Nucleus-targeted polymersomes encapsulating BBI608 Free BBI608 in DMSO

100

Percent 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

Page 26 of 35

80 60 40 20 0 1µM

439

2µM

4µM

[BBI608]

440

Figure 14. The viability of the BxPC3 cells in monolayer cultures at three different concentrations

441

of encapsulated BBI608. (A) Cell viability with media only (control, cyan bar), non-targeted

442

polymersomes (red bar), nucleus-targeted polymersomes encapsulating BBI608 (blue bar), and

443

free BBI608 (magenta bar, N = 4).

444

Viability studies in three-dimensional spheroid cultures of pancreatic cancer cells. Compared

445

to the monolayer cultures, the spheroids demonstrate cellular heterogeneity, cell-cell interactions,

446

and better mimic real tumors.45, 69 To evaluate the effectiveness of the polymersome formulations,

447

BxPC3 spheroids were prepared using the 24-well 3D petri dishes (Microtissues). We treated the

448

7-day old spheroids of the BxPC3 cells (Figure 15A) with nucleus-targeted polymersomes

ACS Paragon Plus Environment

26

Page 27 of 35

449

encapsulating BBI608, non-targeted polymersomes encapsulating BBI608, and free drug for 48

450

hours. The cell viability was determined by the Alamar Blue assay. We observed that the nucleus-

451

targeted polymersomes encapsulating BBI608 decreased (p ≤ 0.05) the cell viability to 43% in

452

compared to non-targeted polymersomes (84%) and the control (Figure 15B). We speculate that

453

dexamethasone in the composition of polymersomes opens the nucleus pores, the vesicles enter

454

the nuclei and release the cancer stemness inhibitor, leading to the enhanced toxicity in the BxPC3

455

cells. Control Non-targeted polymersomes encapsulating BBI608 Nucleus-targeted polymersomes encapsulating BBI608

B 100

Free BBI608 in DMSO

*

80 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

Biomacromolecules

60 40 20 0 8 µM

456

[BBI608]

457

Figure 15. (A) Optical microscopic image of the 7- day old three-dimensional spheroids of the

458

BxPC3 cells (scale bar: 25 µm). (B) The viability of the BxPC3 cells in spheroid cultures. Viability

459

with media only (control, cyan bar), non-targeted polymersomes (red bar), nucleus-targeted

460

polymersomes encapsulating BBI608 (blue bar), and the free BBI608 (magenta bar, N = 4).

461 462 463 464

ACS Paragon Plus Environment

27

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 35

465

Conclusions

466

We successfully synthesized alkyne dexamethasone and conjugated it to the N3 – PEG1900–PLA8000

467

polymer. The nucleus-targeted polymer was combined with a redox-sensitive polymer to form

468

stable polymeric vesicles, which encapsulate air bubbles and a stemness inhibitor. Our echogenic

469

nucleus-targeted polymersomes respond to ultrasound and release the encapsulated BBI608 to the

470

nucleus of pancreatic cancer cells. The nucleus-targeted drug-encapsulated polymersomes reduced

471

the viability of pancreatic cancer cells in monolayer and spheroidal cultures to 30% and 43%,

472

respectively. The polymersomes scatter ultrasound and respond to medical ultrasound imager,

473

confirming the echogenicity. They have the potential to image and deliver chemotherapeutic drugs

474

to the tumors tissues simultaneously. This is a non-invasive strategy to monitor targeted drug

475

delivery to improve the therapeutic outcome of chemotherapy. The results of this research will

476

pave the way for other ultrasound reflective nanoparticles for targeted drug delivery and

477

simultaneous imaging capability.

478 479

ASSOCIATED CONTENT

480

Supporting Information. The Supporting Information is available for readers on the ACS

481

Publications website at DOI:

482

The following files are available free of charge.

483

NMR spectra of the alkyne dexamethasone and the dexamethasone–PEG1900–PLA8000 polymer,

484

NMR spectra, and Gel permeation chromatography of the N3-PEG1900–PLA8000 polymer (file

485

type: PDF).

486

ACS Paragon Plus Environment

28

Page 29 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

487

FUNDING SOURCE

488

This research was supported by the NSF grant DMR 1306154, and the NIH grant 1 R01GM

489

114080 to SM and KS. SM also acknowledges support from the Grand Challenge Initiative and

490

the Office of the Vice President for Research and Creative Activity, North Dakota State University.

491

We acknowledge Dr. Michael D. Scoot for recording and interpreting the NMR spectrum of the

492

alkyne-dexamethasone conjugate.

493 494 495

REFERENCES

496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521

1. Karandish, F.; Mallik, S., Biomarkers and targeted therapy in pancreatic cancer. Biomarkers in cancer 2016, 8, (Suppl 1), 27. 2. Li, Y.; Rogoff, H. A.; Keates, S.; Gao, Y.; Murikipudi, S.; Mikule, K.; Leggett, D.; Li, W.; Pardee, A. B.; Li, C. J., Suppression of cancer relapse and metastasis by inhibiting cancer stemness. Proc. Natl. Acad. Sci. 2015, 112, (6), 1839-1844. 3. Zhang, Y.; Jin, Z.; Zhou, H.; Ou, X.; Xu, Y.; Li, H.; Liu, C.; Li, B., Suppression of prostate cancer progression by cancer cell stemness inhibitor napabucasin. Cancer Med. 2016, 5, (6), 12511258. 4. Hubbard, J. M.; Grothey, A., Napabucasin: An Update on the First-in-Class Cancer Stemness Inhibitor. Drugs 2017, 77, (10), 1091-1103. 5. Ganta, S.; Devalapally, H.; Shahiwala, A.; Amiji, M., A review of stimuli-responsive nanocarriers for drug and gene delivery. J. Controlled Rel. 2008, 126, (3), 187-204. 6. Mohammadi, M.; Ramezani, M.; Abnous, K.; Alibolandi, M., Biocompatible polymersomes-based cancer theranostics: Towards multifunctional nanomedicine. Int. J. Pharm. 2017, 519, (1-2), 287-303. 7. Leong, J.; Teo, J. Y.; Aakalu, V. K.; Yang, Y. Y.; Kong, H., Engineering Polymersomes for Diagnostics and Therapy. Adv. Healthc. Mater. 2018, 7, (8), e1701276. 8. Guan, L.; Rizzello, L.; Battaglia, G., Polymersomes and their applications in cancer delivery and therapy. Nanomedicine (Lond) 2015, 10, (17), 2757-80. 9. Martin, C.; Aibani, N.; Callan, J. F.; Callan, B., Recent advances in amphiphilic polymers for simultaneous delivery of hydrophobic and hydrophilic drugs. Ther. Deliv. 2016, 7, (1), 15-31. 10. Qin, Y.; Zhang, Z.; Huang, C.; Fan, F.; Liu, L.; Lu, L.; Wang, H.; Liu, Z.; Yang, J.; Wang, C.; Yang, H.; Sun, H.; Leng, X.; Kong, D.; Zhang, L.; Zhu, D., Folate-Targeted Redox-Responsive Polymersomes Loaded with Chemotherapeutic Drugs and Tariquidar to Overcome Drug Resistance. J. Biomed. Nanotechnol. 2018, 14, (10), 1705-1718.

ACS Paragon Plus Environment

29

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567

Page 30 of 35

11. Swain, S.; Sahu, P. K.; Beg, S.; Babu, S. M., Nanoparticles for Cancer Targeting: Current and Future Directions. Curr. Drug Deliv. 2016, 13, (8), 1290-1302. 12. Thambi, T.; Park, J. H.; Lee, D. S., Stimuli-responsive polymersomes for cancer therapy. Biomater. Sci. 2016, 4, (1), 55-69. 13. Chen, W.; Meng, F.; Cheng, R.; Zhong, Z., pH-Sensitive degradable polymersomes for triggered release of anticancer drugs: a comparative study with micelles. J. Controlled Rel. 2010, 142, (1), 40-46. 14. Amstad, E.; Kim, S. H.; Weitz, D. A., Photo‐and thermoresponsive polymersomes for triggered release. Angew. Chem. 2012, 124, (50), 12667-12671. 15. Kulkarni, P.; Haldar, M. K.; You, S.; Choi, Y.; Mallik, S., Hypoxia-Responsive Polymersomes for Drug Delivery to Hypoxic Pancreatic Cancer Cells. Biomacromolecules 2016, 17, (8), 2507-2513. 16. Kamat, N. P.; Robbins, G. P.; Rawson, J.; Therien, M. J.; Dmochowski, I. J.; Hammer, D. A., A generalized system for photoresponsive membrane rupture in polymersomes. Adv. Functional Mater. 2010, 20, (16), 2588-2596. 17. Zhu, D.; Fan, F.; Huang, C.; Zhang, Z.; Qin, Y.; Lu, L.; Wang, H.; Jin, X.; Zhao, H.; Yang, H.; Zhang, C.; Yang, J.; Liu, Z.; Sun, H.; Leng, X.; Kong, D.; Zhang, L., Bubble-generating polymersomes loaded with both indocyanine green and doxorubicin for effective chemotherapy combined with photothermal therapy. Acta Biomater. 2018. 18. Sarkar, B.; Paira, P., Theranostic Aspects: Treatment of Cancer by Nanotechnology. Mini Rev Med Chem 2018, 18, (11), 969-975. 19. Xu, P.; Van Kirk, E. A.; Zhan, Y.; Murdoch, W. J.; Radosz, M.; Shen, Y., Targeted Charge‐ Reversal Nanoparticles for Nuclear Drug Delivery. Angew. Chem. Int. Ed. 2007, 46, (26), 49995002. 20. Shahin, V.; Albermann, L.; Schillers, H.; Kastrup, L.; Schafer, C.; Ludwig, Y.; Stock, C.; Oberleithner, H., Steroids dilate nuclear pores imaged with atomic force microscopy. J. Cell. Physiol. 2005, 202, (2), 591-601. 21. Kastrup, L.; Oberleithner, H.; Ludwig, Y.; Schafer, C.; Shahin, V., Nuclear envelope barrier leak induced by dexamethasone. J. Cell. Physiol 2006, 206, (2), 428-34. 22. Conrad, M.; Moreno, S.; Sinowatz, F.; Ursini, F.; Kölle, S.; Roveri, A.; Brielmeier, M.; Wurst, W.; Maiorino, M.; Bornkamm, G., The nuclear form of phospholipid hydroperoxide glutathione peroxidase is a protein thiol peroxidase contributing to sperm chromatin stability. Mol. Cell. Biol. 2005, 25, (17), 7637-7644. 23. Go, Y.-M.; Jones, D. P., Redox Control Systems in the Nucleus: Mechanisms and Functions. Antioxidants Redox Signaling 2010, 13, (4), 489-509. 24. Anajafi, T.; Scott, M. D.; You, S.; Yang, X.; Choi, Y.; Qian, S. Y.; Mallik, S., Acridine orange conjugated polymersomes for simultaneous nuclear delivery of gemcitabine and doxorubicin to pancreatic cancer cells. Bioconjugate Chem. 2016, 27, (3), 762-771. 25. Molineux, G., Pegylation: engineering improved pharmaceuticals for enhanced therapy. Cancer Treatment Rev. 2002, 28, 13-16. 26. Chen, W. L.; Liu, S. J.; Leng, C. H.; Chen, H. W.; Chong, P.; Huang, M. H., Disintegration and cancer immunotherapy efficacy of a squalane-in-water delivery system emulsified by bioresorbable poly(ethylene glycol)-block-polylactide. Biomaterials 2014, 35, (5), 1686-95. 27. Shah, S. S.; Zhu, K. J.; Pitt, C. G., Poly-DL-lactic acid: polyethylene glycol block copolymers. The influence of polyethylene glycol on the degradation of poly-DL-lactic acid. .J Biomater. Sci. Polym. Ed. 1994, 5, (5), 421-31.

ACS Paragon Plus Environment

30

Page 31 of 35 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

568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611

Biomacromolecules

28. Yang, W.; Zou, Y.; Meng, F.; Zhang, J.; Cheng, R.; Deng, C.; Zhong, Z., Efficient and Targeted Suppression of Human Lung Tumor Xenografts in Mice with Methotrexate Sodium Encapsulated in All-Function-in-One Chimeric Polymersomes. Adv. Mater. 2016, 28, (37), 82348239. 29. Wu, J.; Deng, C.; Meng, F.; Zhang, J.; Sun, H.; Zhong, Z., Hyaluronic acid coated PLGA nanoparticulate docetaxel effectively targets and suppresses orthotopic human lung cancer. J. Controlled Rel. 2017, 259, (Supplement C), 76-82. 30. Fang, Y.; Jiang, Y.; Zou, Y.; Meng, F.; Zhang, J.; Deng, C.; Sun, H.; Zhong, Z., Targeted glioma chemotherapy by cyclic RGD peptide-functionalized reversibly core-crosslinked multifunctional poly(ethylene glycol)-b-poly(ε-caprolactone) micelles. Acta Biomaterialia 2017, 50, (Supplement C), 396-406. 31. Zhu, Y.; Wang, X.; Zhang, J.; Meng, F.; Deng, C.; Cheng, R.; Feijen, J.; Zhong, Z., Exogenous vitamin C boosts the antitumor efficacy of paclitaxel containing reduction-sensitive shell-sheddable micelles in vivo. J. Controlled Rel. 2017, 250, (Supplement C), 9-19. 32. Cheng, R.; Feng, F.; Meng, F.; Deng, C.; Feijen, J.; Zhong, Z., Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery. J. Controlled Rel. 2011, 152, (1), 2-12. 33. García-Giménez, J. L.; Markovic, J.; Dasí, F.; Queval, G.; Schnaubelt, D.; Foyer, C. H.; Pallardó, F. V., Nuclear glutathione. Biochim. Biophys. Acta - General Subjects 2013, 1830, (5), 3304-3316. 34. Huang, S.-L., Liposomes in ultrasonic drug and gene delivery. Adv. Drug Deliv. Rev. 2008, 60, (10), 1167-1176. 35. Nahire, R.; Haldar, M. K.; Paul, S.; Mergoum, A.; Ambre, A. H.; Katti, K. S.; Gange, K. N.; Srivastava, D.; Sarkar, K.; Mallik, S., Polymer-coated echogenic lipid nanoparticles with dual release triggers. Biomacromolecules 2013, 14, (3), 841-853. 36. Paul, S.; Russakow, D.; Nahire, R.; Nandy, T.; Ambre, A. H.; Katti, K.; Mallik, S.; Sarkar, K., In vitro measurement of attenuation and nonlinear scattering from echogenic liposomes. Ultrasonics 2012, 52, (7), 962-969. 37. Huang, S.-L., Ultrasound-responsive liposomes. Liposomes: Methods and Protocols, Volume 1: Pharmaceutical Nanocarriers 2010, 113-128. 38. Nahire, R.; Hossain, R.; Patel, R.; Paul, S.; Meghnani, V.; Ambre, A. H.; Gange, K. N.; Katti, K. S.; Leclerc, E.; Srivastava, D., pH-triggered echogenicity and contents release from liposomes. Mol. Pharm. 2014, 11, (11), 4059-4068. 39. Paul, S.; Nahire, R.; Mallik, S.; Sarkar, K., Encapsulated microbubbles and echogenic liposomes for contrast ultrasound imaging and targeted drug delivery. Computational Mech. 2014, 53, (3), 413-435. 40. Nahire, R.; Haldar, M. K.; Paul, S.; Ambre, A. H.; Meghnani, V.; Layek, B.; Katti, K. S.; Gange, K. N.; Singh, J.; Sarkar, K., Multifunctional polymersomes for cytosolic delivery of gemcitabine and doxorubicin to cancer cells. Biomaterials 2014, 35, (24), 6482-6497. 41. Zhang, S.; Zhao, Y., Controlled release from cleavable polymerized liposomes upon redox and pH stimulation. Bioconjugate Chem. 2011, 22, (4), 523-528. 42. Discher, B. M.; Won, Y. Y.; Ege, D. S.; Lee, J. C.; Bates, F. S.; Discher, D. E.; Hammer, D. A., Polymersomes: tough vesicles made from diblock copolymers. Science 1999, 284, (5417), 1143-6.

ACS Paragon Plus Environment

31

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656

Page 32 of 35

43. Nahire, R.; Haldar, M. K.; Paul, S.; Ambre, A. H.; Meghnani, V.; Layek, B.; Katti, K. S.; Gange, K. N.; Singh, J.; Sarkar, K.; Mallik, S., Multifunctional polymersomes for cytosolic delivery of gemcitabine and doxorubicin to cancer cells. Biomaterials 2014, 35, (24), 6482-97. 44. Kopechek, J. A.; Abruzzo, T. M.; Wang, B.; Chrzanowski, S. M.; Smith, D. A.; Kee, P. H.; Huang, S.; Collier, J. H.; McPherson, D. D.; Holland, C. K., Ultrasound‐Mediated Release of Hydrophilic and Lipophilic Agents From Echogenic Liposomes. J. Ultrasound Med. 2008, 27, (11), 1597-1606. 45. Karandish, F.; Haldar, M. K.; You, S.; Brooks, A. E.; Brooks, B. D.; Guo, B.; Choi, Y.; Mallik, S., Prostate-Specific Membrane Antigen Targeted Polymersomes for Delivering Mocetinostat and Docetaxel to Prostate Cancer Cell Spheroids. ACS omega 2016, 1, (5), 952-962. 46. Mi Bae, Y.; Choi, H.; Lee, S.; Ho Kang, S.; Tae Kim, Y.; Nam, K.; Sang Park, J.; Lee, M.; Sig Choi, J., Dexamethasone-conjugated low molecular weight polyethylenimine as a nucleustargeting lipopolymer gene carrier. Bioconjugate Chem. 2007, 18, (6), 2029-36. 47. Schnelldorfer, T.; Gansauge, S.; Gansauge, F.; Schlosser, S.; Beger, H. G.; Nussler, A. K., Glutathione depletion causes cell growth inhibition and enhanced apoptosis in pancreatic cancer cells. Cancer 2000, 89, (7), 1440-1447. 48. Schafer, F. Q.; Buettner, G. R., Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Rad. Biol. Med. 2001, 30, (11), 11911212. 49. Redmond, S. M.; Joncourt, F.; Buser, K.; Ziemiecki, A.; Altermatt, H.-J.; Fey, M.; Margison, G.; Cerny, T., Assessment of P-glycoprotein, glutathione-based detoxifying enzymes and O6-alkylguanine-DNA alkyltransferase as potential indicators of constitutive drug resistance in human colorectal tumors. Cancer Res. 1991, 51, (8), 2092-2097. 50. Forsberg, F.; Shi, W. T.; Goldberg, B., Subharmonic imaging of contrast agents. Ultrasonics 2000, 38, (1), 93-98. 51. Sarkar, K.; Shi, W. T.; Chatterjee, D.; Forsberg, F., Characterization of ultrasound contrast microbubbles using in vitro experiments and viscous and viscoelastic interface models for encapsulation. J. Acoustic. Soc. Am. 2005, 118, (1), 539-550. 52. Huang, S. L.; Hamilton, A. J.; Nagaraj, A.; Tiukinhoy, S. D.; Klegerman, M. E.; Mcpherson, D. D.; Macdonald, R. C., Improving ultrasound reflectivity and stability of echogenic liposomal dispersions for use as targeted ultrasound contrast agents. J. Pharm. Sci. 2001, 90, (12), 1917-1926. 53. Huang, S. L., Liposomes in ultrasonic drug and gene delivery. Adv. Drug Delivery Rev. 2008, 60, (10), 1167-1176. 54. Kopechek, J. A.; Haworth, K. J.; Raymond, J. L.; Mast, T. D.; Perrin, S. R.; Klegerman, M. E.; Huang, S. L.; Porter, T. M.; McPherson, D. D.; Holland, C. K., Acoustic characterization of echogenic liposomes: Frequency-dependent attenuation and backscatter. J. Acoust. Soc. Am. 2011, 130, (5), 3472-3481. 55. Coussios, C. C.; Holland, C. K.; Jakubowska, L.; Huang, S. L.; MacDonald, R. C.; Nagaraj, A.; McPherson, D. D., In vitro characterization of liposomes and Optison (R) by acoustic scattering at 3.5 MHz. Ultrasound Med. Biol. 2004, 30, (2), 181-190. 56. Kee, P.; Abruzzo, T.; Smith, D. A. B.; Wang, B.; Huang, S. L.; MacDonald, R. C.; Holland, C. K.; McPherson, D. D., Synthesis and acoustic characterization of a novel ultrasound controlled drug delivery system based on echogenic liposomes. J. Am. Coll. Cardiolog. 2007, 49, (9), 120a120a.

ACS Paragon Plus Environment

32

Page 33 of 35 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

657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690

Biomacromolecules

57. Paul, S.; Nahire, R.; Mallik, S.; Sarkar, K., Encapsulated microbubbles and echogenic liposomes for contrast ultrasound imaging and targeted drug delivery. Computational Mechanics 2014, 53, (3), 413-435. 58. Nahire, R.; Paul, S.; Scott, M. D.; Singh, R. K.; Muhonen, W. W.; Shabb, J.; Gange, K. N.; Srivastava, D. K.; Sarkar, K.; Mallik, S., Ultrasound Enhanced Matrix Metalloproteinase-9 Triggered Release of Contents from Echogenic Liposomes. Mol. Pharm. 2012, 9, (9), 2554-2564. 59. Nahire, R.; Hossain, R.; Patel, R.; Paul, S.; Meghnani, V.; Ambre, A. H.; Gange, K. N.; Katti, K. S.; Leclerc, E.; Srivastava, D. K.; Sarkar, K.; Mallik, S., pH-Triggered Echogenicity and Contents Release from Liposomes. Molecular Pharmaceutics 2014, 11, (11), 4059-4068. 60. Nahire, R.; Haldar, M. K.; Paul, S.; Mergoum, A.; Ambre, A. H.; Katti, K. S.; Gange, K. N.; Srivastava, D. K.; Sarkar, K.; Mallik, S., Polymer-Coated Echogenic Lipid Nanoparticles with Dual Release Triggers. Biomacromolecules 2013, 14, (3), 841-853. 61. Huang, S.-L.; Hamilton, A. J.; Pozharski, E.; Nagaraj, A.; Klegerman, M. E.; McPherson, D. D.; MacDonald, R. C., Physical correlates of the ultrasonic reflectivity of lipid dispersions suitable as diagnostic contrast agents. Ultrasound Med. Biol. 2002, 28, (3), 339-348. 62. Huang, S. L.; McPherson, D. B.; MacDonald, R. C., A method to co-encapsulate gas and drugs in liposomes for ultrasound-controlled drug delivery. Ultrasound Med. Biol. 2008, 34, (8), 1272-1280. 63. Kopechek, J. A.; Haworth, K. J.; Raymond, J. L.; Douglas Mast, T.; Perrin, S. R.; Klegerman, M. E.; Huang, S.; Porter, T. M.; McPherson, D. D.; Holland, C. K., Acoustic characterization of echogenic liposomes: Frequency-dependent attenuation and backscatter. J. Acoust. Soc. Am. 2011, 130, (5), 3472. 64. Sarkar, K.; Katiyar, A.; Jain, P., Growth and dissolution of an encapsulated contrast microbubble Ultrasound Med. Biol. 2009, 35, (8), 1385-1396. 65. Katiyar, A.; Sarkar, K.; Jain, P., Effects of Encapsulation Elasticity on the stability of an Encapsulated Microbubble. J. Coll. Interfac. Sci. 2009, 336, 519-525. 66. Katiyar, A.; Sarkar, K., Stability analysis of an encapsulated microbubble against gas diffusion. J. Coll. Interfac. Sci. 2010, 343, (1), 42-47. 67. Shi, W. T.; Forsberg, F., Ultrasonic characterization of the nonlinear properties of contrast microbubbles. Ultrasound Med. Biol. 2000, 26, (1), 93-104. 68. Kastrup, L.; Oberleithner, H.; Ludwig, Y.; Schafer, C.; Shahin, V., Nuclear envelope barrier leak induced by dexamethasone. J. Cell. Physiology 2006, 206, (2), 428-434. 69. Desoize, B.; Gimonet, D.; Jardiller, J., Cell culture as spheroids: an approach to multicellular resistance. Anticancer Res. 1997, 18, (6A), 4147-4158.

691 692 693 694 695 696

ACS Paragon Plus Environment

33

Biomacromolecules 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

697

Page 34 of 35

TOC graphic:

698

ACS Paragon Plus Environment

34

Page 35 of 35 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

TOC Graphic: The polymersomes enter the nuclei of pancreatic cancer cells and deliver a stemness inhibitor.

ACS Paragon Plus Environment