Self-Assembly PEGylation Retaining Activity (SPRA) Technology via a

Dec 29, 2016 - Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto. Univers...
0 downloads 8 Views 3MB Size
Subscriber access provided by University of Newcastle, Australia

Article

Self-assembly PEGylation Retaining Activity (SPRA) Technology via a Host– guest Interaction Surpassing Conventional PEGylation Methods of Proteins Tatsunori Hirotsu, Taishi Higashi, Irhan Ibrahim Abu Hashim, Shogo Misumi, Koki Wada, Keiichi Motoyama, and Hidetoshi Arima Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.6b00678 • Publication Date (Web): 29 Dec 2016 Downloaded from http://pubs.acs.org on December 30, 2016

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

Molecular Pharmaceutics 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 34

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

1

Molecular Pharmaceutics

Article

Revised

2

3

Self-assembly PEGylation Retaining Activity (SPRA) Technology via a Host–

4

guest Interaction Surpassing Conventional PEGylation Methods of Proteins

5

Tatsunori Hirotsu †, ‡, #, Taishi Higashi †, #, Irhan Ibrahim Abu Hashim †, §, Shogo Misumi †, Koki Wada ǁ,

6

Keiichi Motoyama †, Hidetoshi Arima †, ‡, *

7



8

Kumamoto 862-0973, Japan

9



Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku,

Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal

10

Oriented) Program”, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan

11

§

Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt

12

ǁ

Nihon Shokuhin Kako Co., Ltd., 30 Tajima, Fuji, Shizuoka 417-8539, Japan

13 14

#

15

* Corresponding author.

These authors contributed equally to this work.

16

Hidetoshi Arima

17

Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku,

18

Kumamoto 862-0973, Japan.

19

E-mail: [email protected], TEL: +81 96 371 4160, FAX: +81 96 371 4420

20 21

1

ACS Paragon Plus Environment

Molecular Pharmaceutics

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

22

ABSTRACT

23

Polyethylene glycol (PEG) modification (PEGylation) is one of the best approaches to improve the

24

stabilities and blood half-lives of protein drugs; however, PEGylation dramatically reduces the

25

bioactivities of protein drugs. Here, we present “self-assembly PEGylation retaining activity” (SPRA)

26

technology via a host–guest interaction between PEGylated β-cyclodextrin (PEG-β-CyD) and

27

adamantane-appended (Ad)-proteins. PEG-β-CyD formed stable complexes with Ad-insulin and Ad-

28

lysozyme to yield SPRA-insulin and SPRA-lysozyme, respectively. Both SPRA-proteins showed high

29

stability against heat and trypsin digest, comparable with that of covalently PEGylated protein equivalents.

30

Importantly, the SPRA-lysozyme possessed ca. 100% lytic activity, whereas the activity of the covalently

31

PEGylated lysozyme was ca. 23%. Additionally, SPRA-insulin provided a prolonged and peakless blood

32

glucose profile when compared with insulin glargine. It also showed no loss of activity. In contrast, the

33

covalently PEGylated insulin showed a negligible hypoglycemic effect. These findings indicate that

34

SPRA technology has the potential as a generic method, surpassing conventional PEGylation methods for

35

proteins.

36 37

KEYWORDS : cyclodextrin, adamantine, polyethylene glycol, protein, supramolecular host–guest

38

interaction

39 40

Table of contents graphic

41 42 2

ACS Paragon Plus Environment

Page 2 of 34

Page 3 of 34

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

Molecular Pharmaceutics

43

1. INTRODUCTION

44

Protein drugs have been used widely as valuable therapeutic agents, with worldwide sales in 2014

45

showing that protein drugs represent 7 out of the top 10 pharmaceuticals used.1 However, protein drugs

46

often show low physicochemical stability, low enzymatic stability, immunogenicity and short circulating

47

half-lives.2 Therefore, pharmaceutical techniques are needed to develop protein drug formulations that

48

show superior stability and activity.

49

Covalent polyethylene glycol (PEG) modification (PEGylation) of proteins is one of the best

50

approaches to improve pharmaceutical properties of protein drugs, and a number of PEGylated proteins

51

have been used in the clinical field.3, 4 However, bioactivities of proteins are dramatically reduced by

52

PEGylation, because PEGylated proteins bind weakly to their cognate receptors or substrates due to steric

53

hindrance of PEG or structural changes to the PEGylated proteins.3, 5 For example, PEGylated interferon

54

α-2a (branched PEG, M.W. 40 kDa) and PEGylated erythropoietin (linear PEG, M.W. 30 kDa), both of

55

which are commercially available products, show only 7% and 1–2% bioactivities, respectively,

56

compared with those of native interferon and erythropoietin.6, 7

57

Steric hindrance by the PEG chain is due to the bulky hydrated PEG, which forms a layer on the

58

surface of the protein. Here, PEG entangles around the protein surface through hydrophobic interactions

59

and concurrently forms hydrogen bonds with the surrounding water molecules.8 The hydrodynamic radius

60

of PEG (M.W. 2–30 kDa) is ca. 1.36–5.98 nm.9 Although these features of PEGylated proteins provide

61

crucial stability and avoid glomerular filtration (slit-pores are 3.5 nm), they reduce the bioactivity of the

62

protein drug considerably.

63

To develop PEGylated proteins, the benefits and disadvantages of PEGylation must be considered, and

64

an acceptable M.W. of PEG should be determined.5 For example, mono- and multi-substituted PEGylated

65

lysozyme (PEG 20 kDa) showed high stabilities; however, their lytic activities were only 1.4% and 1.1%,

66

respectively.10 In addition, PEGylation of insulin with PEG 2 kDa improved its physicochemical stability,

67

but it did not prolong its hypoglycemic effect (the activity was 85%).11, 12 In our previous report, the 3

ACS Paragon Plus Environment

Molecular Pharmaceutics

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

68

enzymatic stability of insulin was not improved by PEGylation with PEG 2 kDa.13, 14 Torosantucci et al.

69

also reported that PEGylation with PEG 5 kDa does not protect insulin against forced aggregation.15 In

70

addition, a PEGylated insulin analog (PEG-lispro, linear PEG, M.W. 20 kDa) was in Phase III of clinical

71

trials as a once-daily treatment for type-1 and type-2 diabetes, although it was recently stopped.16-18 PEG-

72

lispro has a >10-fold longer blood half-life than insulin lispro, leading to a prolonged hypoglycemic effect,

73

compared with that of insulin glargine. However, PEG-lispro possesses only 6% of the bioactivity of

74

intact insulin lispro.16,

75

protein drugs’ pharmaceutical properties and bioactivity. Hence, PEGylation that can improve the

76

pharmaceutical properties of proteins without loss of activity will lead to development of a large number

77

of PEGylated proteins.

18

Thus, PEGylation of proteins represents a serious dilemma with respect to

78

Recently, site-specific PEGylation19-21 and reversible PEGylation22-24 technologies have been developed.

79

These approaches improve the pharmaceutical properties of protein drugs without significant loss of

80

bioactivity. Nonetheless, these PEGylated proteins showed only 10–40% bioactivity, compared with

81

unmodified proteins. Thus, improving the pharmaceutical properties of proteins by PEGylation without

82

significant loss of activity is challenging.

83

Cyclodextrins (CyDs), cyclic oligosaccharides, are acknowledged to form inclusion complexes with

84

hydrophobic compounds spontaneously through a host–guest interaction.25, 26 Recently, a large number of

85

CyD-based supramolecular drug carriers have been developed for low-molecular weight drugs, protein

86

drugs, genes and oligonucleotides.27-29 However, little is known about the fabrication of reversible

87

PEGylated proteins utilizing CyD-based supramolecular properties.

88

Based on these studies, we propose “Self-assembly PEGylation Retaining Activity (SPRA)”

89

technology via a host–guest interaction between β-CyD and adamantane (Ad). We prepared PEGylated

90

proteins through supramolecular inclusion complexation rather than covalent bond linkage and evaluated

91

their pharmaceutical properties. These SuPRAmolecular PEGylated proteins (SPRA-proteins) are based

92

on a reversible host–guest interaction, and are expected to show prolonged and high bioactivities. We 4

ACS Paragon Plus Environment

Page 4 of 34

Page 5 of 34

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

Molecular Pharmaceutics

93

employed β-CyD as a host molecule and Ad as a guest molecule because they form a stable complex,30

94

and they are also used as a part of siRNA carrier in human.31 Additionally, we used insulin as a model

95

protein because PEG-lispro shows only 6% bioactivity of unmodified insulin.16 Also, lysozyme as a

96

model protein was used to demonstrate the utility of SPRA technology for other protein drugs.

97 98

2. MATERIAL AND METHODS

99

2.1. Materials. Bovine Zn-insulin (27.5 IU/mg, approximately 0.5% Zn) and hen egg lysozyme (58,100

100

IU/mg) were obtained from Sigma Chemicals (St. Louis, MO, USA). β-CyD and 6-O-α-(4-O-α-D-

101

glucuronyl)-D-glucosyl-β-CyD (GUG-β-CyD) were donated by Nihon Shokuhin Kako (Tokyo, Japan)

102

and Ensuiko Sugar Refining (Tokyo, Japan), respectively. Both mono- and per-NH2-β-CyDs were

103

prepared according to the previous method.32, 33 SUNBRIGHT® ME-200 CS (mPEG-NHS) was obtained

104

from NOF (Tokyo, Japan). All other chemicals and solvents were of analytical reagent grade and

105

deionized double-distilled water was used throughout the study.

106 107

2.2. Preparation of Ad-proteins. To obtain Ad conjugate with protein (Ad-protein), adamantane acetic

108

acid (190 mg), N-hydroxysuccinimide (NHS) (230 mg) and N,N'-dicyclohexylcarbodiimide (DCC) (820

109

mg) were dissolved in 6 mL of DMSO. The solution was stirred at room temperature for 1 h and

110

centrifuged at 12,890 G for 15 min. The supernatant was added to 300 mL of water to precipitate

111

activated Ad and resulting precipitates were dried under reduced pressure. Insulin (28.5 mg) and activated

112

Ad (20 mg) were dissolved in 5 mL of dimethylformamide (DMF)/water (3:2 v/v, pH 10) and stirred at

113

room temperature for 10 min. Water (40 mL) was added, and the pH adjusted to 2 with 1.0 M

114

hydrochloric acid to terminate the reaction. The reaction solution was dialyzed using a membrane filter

115

(Spectra/Por® membrane MWCO: 3,500) and lyophilized. Finally, crude product was purified by HPLC

116

with the following conditions: GL Pack NUCLEOSIL 100–10C18 (4.6 mm i.d. × 150 mm), a mobile

117

phase of acetonitrile/water/trifluoroacetic acid (30:69.9:0.1) and acetonitrile/water/trifluoroacetic acid 5

ACS Paragon Plus Environment

Molecular Pharmaceutics

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

118

(95:4.9:0.1), a gradient flow increasing the ratio of the latter solution (0–100%/60 min), a flow rate of 1.0

119

mL/min and detection at 280 nm. Likewise, Ad-lysozyme was prepared by mixing lysozyme (58.8 mg)

120

and activated Ad (7.2 mg) in 5 mL of DMF/PBS (2:3 v/v, pH 8) solution at 4 °C for 24 h. After dialysis

121

using a membrane filter (Spectra/Por® membrane MWCO: 8,000), the sample was lyophilized and

122

purified by HPLC.

123 124

2.3. Preparation of PEG-β β-CyDs. To obtain mono-substituted PEGylated β-CyD (mono-PEG-β-CyD),

125

SUNBRIGHT® ME-200 CS (mPEG-NHS, 1,000 mg) and mono-NH2-β-CyD (590 mg) were reacted in 20

126

mL of DMF/water (3:2 v/v) at room temperature for 24 h. The reaction solution was dialyzed using a

127

membrane filter (Spectra/Por® membrane MWCO: 8,000) until free NH2-β-CyD was completely removed.

128

The resulting sample was lyophilized. Likewise, multi-substituted PEGylated-β-CyD (multi-PEG-β-CyD)

129

was prepared by mixing mPEG-NHS (1,000 mg) and per-NH2-β-CyD (5.64 mg) in 20 mL of DMF/water

130

(3:2 v/v) at room temperature for 24 h. After dialysis using a membrane filter (Spectra/Por® membrane

131

MWCO: 50,000), the sample was lyophilized.

132 133

2.4. Formation of Supramolecular Complexes. Isothermal titration calorimetry (ITC) was measured at

134

a titration rate of 2.5 µL/2 min and stirring speed of 350 rpm at 25 °C using a Nano ITC LV (Tokyo,

135

Japan). Ad-proteins and PEG-β-CyDs were dissolved in PBS. The mono-PEG-β-CyD (0.1 or 0.5 mM)

136

and multi-PEG-β-CyD (0.1 or 0.5 mM) solutions were loaded into the sample cell. After the titrations of

137

the Ad-insulin solution (1 or 2 mM) or Ad-lysozyme solution (1 or 2 mM) into the sample cell, the

138

stability constants (Kc) were determined from the titration curve using an independent model. Particle

139

sizes of the Ad-proteins/mono-PEG-β-CyD (mono-SPRA-proteins) and Ad-proteins/multi-PEG-β-CyD

140

(multi-SPRA-proteins) were determined by dynamic light scattering using a Zetasizer Nano (Malvern

141

Instruments, Worcestershire, UK). The solvent was PBS, and the concentration of Ad-insulin or Ad6

ACS Paragon Plus Environment

Page 6 of 34

Page 7 of 34

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

Molecular Pharmaceutics

142

lysozyme was 0.025 mM. The molar ratios of Ad-proteins and PEG-β-CyDs were 1:0.1, 1:0.5, 1:1, 1:2,

143

1:5 and 1:10.

144 145

2.5. Thermal Stability.34 Ad-proteins (0.05 mM) and unmodified proteins (0.05 mM) were mixed with

146

PEG-β-CyDs in PBS (pH 7.4) at a molar ratio of 1:10. Proteins alone, Ad-proteins alone and covalently

147

PEGylated proteins (PEG-proteins) (0.05 mM) were also dissolved in PBS. These solutions were

148

incubated at 37 °C (the insulin system) or 70 °C (the lysozyme system). At appropriate intervals, 0.7 mL

149

of the samples were collected and centrifuged at 8,952 G for 10 min. The absorbance of the supernatant

150

was measured by a spectrophotometer (V-630, JASCO, Tokyo, Japan) at 280 nm.

151 152

2.6. Enzymatic Stability.34 Ad-proteins (5 mg) and unmodified proteins (5 mg) were mixed with PEG-β-

153

CyDs in 5 mL of PBS (pH 7.4) at a molar ratio of 1:10. Proteins alone, Ad-proteins alone and PEG-

154

proteins (5 mg) were also dissolved in 5 mL of PBS. PBS (100 µL) including 1.0 mg/mL trypsin was

155

added to 300 µL of protein solutions. These solutions were incubated at 37 °C and 50 rpm for 6 h, and the

156

non-degraded insulin or lysozyme was measured by HPLC.

157 158

2.7. In Vitro Lytic Activity.35 Ad-lysozyme (0.4 µM) or lysozyme (0.4 µM) was mixed with PEG-β-

159

CyDs in 5 mL of PBS (pH 7.4) at a molar ratio of 1:5. Lysozyme, Ad-lysozyme and PEG-lysozyme (0.4

160

µM) were also dissolved in 5 mL of PBS. The suspension of M. lysodeikticus cells (1 mg/mL, 50 µL) was

161

added to the lysozyme solutions (900 µL), and then incubated at 25 °C for 2 min. The turbidity of the

162

suspension was measured at 540 nm using a spectrophotometer. The lytic activity was estimated as a

163

relative activity (% activity) based on a decrease in turbidity relative to the lysozyme alone system.

164

7

ACS Paragon Plus Environment

Molecular Pharmaceutics

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

165

2.8. In Vivo Serum Insulin Level, Hypoglycemic Effect and Blood Chemistry Values.13 All animal

166

procedures were carried out in accordance with the approved guidelines and with the approval of the

167

Ethics Committee for Animal Care and Use of Kumamoto University (Approval ID: A27-139). Insulin,

168

Ad-insulin, PEG-insulin, multi-SPRA-insulin (molar ratio = 1:5), insulin/multi PEG-β-CyD mixture

169

(molar ratio = 1:5) and insulin glargine were dissolved in PBS (pH 7.4). The samples (insulin 2 U/kg)

170

were subcutaneously injected into male Wistar rats (200–250 g), and at appropriate intervals, blood

171

samples were taken from the jugular veins. The serum insulin and glucose levels of rats were determined

172

by the enzyme immunoassay using a Rat Insulin ELISA Kit (Shibayagi, Gunma, Japan) and mutarotase-

173

glucose oxidase method using the Glucose-CII-Test Wako (Wako Pure Chemical Industries, Osaka,

174

Japan), respectively. Blood chemistry values were measured by a clinical chemistry analyzer, JCA-

175

BM2250 (JEOL, Tokyo, Japan) after injection of the samples.

176 177

3. RESULTS

178

3.1. Design and Preparation of Components of SPRA-insulin and SPRA-lysozyme. We previously

179

prepared the insulin conjugate with GUG-β-CyD.34 Therefore, we initially employed this conjugate as a

180

component of SPRA-proteins. However, 89% of the hypoglycemic effect of insulin was lost by

181

conjugation with GUG-β-CyD, when assuming that the upper area of the blood glucose level–time curve

182

(AUCG) corresponds with in vivo bioactivity of insulin (Figure S1). Thus, we prepared an insulin

183

conjugate with Ad; a guest molecule (Figure 1). The Ad-conjugation reaction was performed at pH 10,

184

because an electrophilic moiety selectively reacts with the amino group of LysB29 of insulin at pH 10 and

185

LysB29 is also employed to prepare commercially available bioconjugated insulins, such as insulin

186

detemir, insulin degludec and PEG-lispro.18, 36, 37 According to the results of HPLC of the reaction mixture

187

(Figure S2), 47.2% of mono-substituted Ad-insulin was included in the reaction mixture. After the

188

purification of crude products with HPLC, the purity of Ad-insulin was determined as >99% by HPLC

189

and single peak derived from mono-substituted Ad-insulin was observed in the MALDI-TOF MS 8

ACS Paragon Plus Environment

Page 8 of 34

Page 9 of 34

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

Molecular Pharmaceutics

190

spectrum (Figure 2A, B). Importantly, Ad-insulin retained ca. 100% of the hypoglycemic effect of insulin

191

as described later, suggesting negligible adverse effects of Ad-conjugation on the bioactivity of insulin.

192

Likewise, mono-substituted lysozyme conjugate with Ad was successfully prepared with 15.2% of crude

193

yield (Figure S2) and purified with >99% of purity (Figures 1 and 2C, D). Hereafter, the exact position of

194

Ad moiety in the Ad-proteins should be determined by the measurement of MALDI-TOF MS after the

195

trypsin digestion.

196

Next, we prepared mono-PEG-β-CyD (Figure 3A) and multi-PEG-β-CyD (Figure 3B); the PEGylated

197

host molecules. The results of the MALDI-TOF MS (Figure 3C-E), FT-IR (Figure S3) and 1H-NMR

198

(Figure S4) spectra indicated successful preparation of these PEG-β-CyDs, and average degrees of

199

substitution of the PEG chain per a β-CyD molecule were 1.0 and 2.8 in mono-PEG-β-CyD and multi-

200

PEG-β-CyD, respectively. Hereafter, gel permeation chromatography of multi-PEG-β-CyD should be

201

measured to determine its distribution, heterogeneity and accurate degree of substitution of PEG.

202

9

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

203 204

Figure 1. Preparation pathways of SPRA-insulin and SPRA-lysozyme. Kc represents the stability constant

205

of SPRA-proteins, and was determined by ITC.

206

207 208

Figure 2. MALDI-TOF MS spectra of (A) insulin, (B) Ad-insulin, (C) lysozyme and (D) Ad-lysozyme. 10

ACS Paragon Plus Environment

Page 11 of 34

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

Molecular Pharmaceutics

209 210

Figure 3. (A, B) Preparation pathways and (C, D, E) MALDI-TOF MS spectra of (C) PEG (20 kD), (A,

211

D) mono-PEG-β-CyD and (B, E) multi-PEG-β-CyD. 11

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

212

3.2. Preparation of SPRA-insulin and SPRA-lysozyme. As shown in Figure 1, four kinds of SPRA-

213

proteins, consisting of Ad-insulin/mono-PEG-β-CyD (mono-SPRA-insulin), Ad-insulin/multi-PEG-β-

214

CyD

215

lysozyme/multi-PEG-β-CyD (multi-SPRA-lysozyme), were prepared by mixing both components in

216

aqueous solution. According to the results of ITC, exothermic peaks were observed by the addition of

217

mono-PEG-β-CyD or multi-PEG-β-CyD solutions (Figure S5), indicating complexation between the Ad

218

moiety of Ad-proteins and the β-CyD moiety of PEG-β-CyDs. Additionally, Kc values of SPRA-proteins

219

were determined as 2.7 × 104–4.8 × 105 M−1 by ITC (Figure 1). Meanwhile, the Kc value of adamantane

220

acetic acid and multi-PEG-β-CyD was determined as 1.1 × 105 M−1 (Figure S6), and was higher than that

221

of multi-SPRA-insulin (2.7 × 104 M−1) or multi-SPRA-lysozyme (9.3 × 104 M−1), indicating that protein-

222

modification somewhat reduces the interaction between Ad and multi-PEG-β-CyD.

223

proteins and PEG-β-CyDs are highly likely to form SPRA-proteins with a certain level of Kc values.

(multi-SPRA-insulin),

Ad-lysozyme/mono-PEG-β-CyD

(mono-SPRA-lysozyme)

and

Ad-

Anyhow, Ad-

224

In addition, particle sizes of Ad-insulin and Ad-lysozyme were found to increase by the addition of

225

mono-PEG-β-CyD and multi-PEG-β-CyD in a concentration-dependent manner due to complexation of

226

both components (Figure 4). Hence, these results indicate the formation of SPRA-proteins. Covalently

227

PEGylated insulin (PEG-insulin) and PEGylated lysozyme (PEG-lysozyme) were prepared as controls

228

with activated PEG (M.W. 20 kDa) (Figure 1).

229

12

ACS Paragon Plus Environment

Page 13 of 34

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

Molecular Pharmaceutics

230 231

Figure 4. Particle sizes of SPRA-insulin and SPRA-lysozyme determined by dynamic light scattering.

232 233

3.3. Physicochemical and Enzymatic Stabilities of SPRA-insulin and SPRA-lysozyme. Protein drugs

234

often form aggregates during long-term storage, resulting in loss of bioactivity, immunogenicity and

235

alteration of pharmacokinetics. In addition, enzyme degradation of protein drugs also causes loss of

236

bioactivity. In this context, PEGylation is a powerful approach to improve the physicochemical and

237

enzymatic stabilities of protein drugs. Thus, thermal and enzymatic stabilities of SPRA-proteins were

238

examined (Figures 5 and 6). Here, to evaluate the thermal stabilities of SPRA-insulin and SPRA-

239

lysozyme, the temperature was set at 37 °C and 70 °C, respectively, according to results of preliminary

240

experiment, which showed that insulin or lysozyme were certainly degraded. As shown in Figure 5,

241

insulin and lysozyme gradually formed aggregates at 37 °C and 70 °C, respectively. Also, the addition of

242

multi-PEG-β-CyD to insulin or lysozyme did not improve their thermal stability, suggesting the

243

negligible stabilizing effects of multi-PEG-β-CyD (Figure S7). In contrast, mono- and multi-SPRA-

244

proteins showed high thermal stability compared with the results of protein-only samples. In addition, the

245

stability of SPRA-insulin was equivalent to that of PEG-insulin and SPRA-lysozyme showed higher

246

stability than PEG-lysozyme after 6.5 h of incubation. 13

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

247

The stability of SPRA-proteins against trypsin digest, especially multi-SPRA-proteins, was also

248

superior to protein-only samples or mixtures of proteins/multi-PEG-β-CyD and equivalent to covalent

249

PEG-proteins (Figure 6, Figure S8). These results show that SPRA technology dramatically improves the

250

thermal and enzymatic stabilities of protein drugs.

251

252 253

Figure 5. Thermal stability of SPRA-insulin at 37 °C and SPRA-lysozyme at 70 °C. Each point

254

represents the mean ± S.E. of 4–7 experiments. *p < 0.05 versus insulin or lysozyme.

255 256

Figure 6. Enzymatic stability of SPRA-insulin and SPRA-lysozyme against trypsin treatment for 6 h.

257

Each value represents the mean ± S.E. of 3–6 experiments. *p < 0.05 versus insulin or lysozyme.

258 259

3.4. In Vitro Enzymatic Activities of SPRA-lysozyme. To evaluate the in vitro enzymatic activity of

260

SPRA-lysozyme, we measured the lytic activity of lysozyme against M. lysodeikticus cells (Figure 7).

261

PEG-lysozyme showed only 23% lytic activity, compared with the lytic activity of native unmodified 14

ACS Paragon Plus Environment

Page 15 of 34

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

Molecular Pharmaceutics

262

lysozyme. Moreover, the mixture of lysozyme with multi-PEG-β-CyD showed ca. 100% of the lytic

263

activity (Figure S9), suggesting that the addition of multi-PEG-β-CyD to lysozyme does not affect the

264

activity. Importantly, the lytic activities of mono- and multi-SPRA-lysozymes were 90% and 100%,

265

respectively, compared with those of unmodified lysozyme, indicating near complete preservation of the

266

in vitro activity of SPRA-lysozyme.

267

268 269

Figure 7. Lytic activities of SPRA-lysozyme to M. Lysodeikticus cells. Each value represents the mean ±

270

S.E. of 5–13 experiments. *p < 0.05 versus lysozyme.

271 272

3.5. In Vivo Serum Insulin Level and Hypoglycemic Effect of SPRA-insulin. To achieve optimum

273

treatment of diabetes, rapid- and long-acting insulins have been utilized in the clinical field. In the case of

274

long-acting insulin, a flat blood glucose profile resulting from the prolonged hypoglycemic effect of

275

insulin is strongly required. Thus, the serum insulin and glucose levels were evaluated after subcutaneous

276

administration of SPRA-insulin in rats (Figure 8). Here, we chose multi-SPRA-insulin because of its

277

higher physicochemical and enzymatic stabilities and larger molecular weight than mono-SPRA-insulin.

278

High serum insulin levels were retained after subcutaneous administration of multi-SPRA-insulin

279

compared to that of insulin alone (Figure 8A). In addition, area under the blood concentration-time curve

280

(AUC) of SPRA-insulin (81.3 ng/mL·h) was higher than that of intravenously administrated insulin alone 15

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

281

(Table S1). As a result, absolute bioavailability of multi-SPRA-insulin after subcutaneous administration

282

was 195%, and was higher than that of insulin. This is probably due to the high enzymatic stability and

283

high retention in the blood of multi-SPRA-insulin. As shown in Figure 8B, duration of the hypoglycemic

284

effect of Ad-insulin was equivalent to that of insulin alone. On the other hand, multi-SPRA-insulin

285

provided the prolonged and flat blood glucose level for 24 h (Figure 8B, left). In fact, the mean residence

286

time of serum glucose levels (MRTG) of multi-SPRA-insulin was significantly higher than that of insulin

287

(Figure 8C). Besides, the cumulative percentage of change in plasma glucose levels up to 24 h post-

288

administration (AUCG) of multi-SPRA-insulin was also superior to that of insulin alone, indicating higher

289

in vivo activity of multi-SPRA-insulin (Figure 8D). In sharp contrast, PEG-insulin showed the negligible

290

hypoglycemic effect, and its AUCG was only 10% versus insulin alone. Importantly, a mixture of insulin

291

and multi-PEG-β-CyD did not show the prolonged the hypoglycemic effect (data not shown). Next, to

292

evaluate the utility of multi-SPRA-insulin as long-acting insulin preparation, the hypoglycemic effect of

293

multi-SPRA-insulin was compared with insulin glargine, a commercially available long-acting insulin

294

analog and the best-selling diabetes drug in 2014. Multi-SPRA-insulin showed higher AUCG and MRTG

295

than insulin glargine (Figure 8C, D). Most importantly, the blood glucose profile after administration of

296

multi-SPRA-insulin was flat when compared with that of insulin glargine (Figure 8B, right). Thus, multi-

297

SPRA-insulin is a promising long-acting insulin.

298 299

16

ACS Paragon Plus Environment

Page 17 of 34

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

Molecular Pharmaceutics

300 301

Figure 8. (A) Serum insulin levels after subcutaneous or intravenous administration of insulin (2 U/kg)

302

and multi-SPRA-insulin (2 U/kg) to rats. (B) Serum glucose levels after subcutaneous administration of

303

insulin (2 U/kg), Ad-insulin (2 U/kg), PEG-insulin (2 U/kg) and multi-SPRA-insulin (2 U/kg) to rats. To

304

compare the effects of multi-SPRA-insulin (2 U/kg) and insulin glargine (2 U/kg), the same results of

305

multi-SPRA-insulin are described in the right figure. (C) MRTG and (D) AUCG of multi-SPRA-insulin.

306

Each point represents the mean ± S.E. of 5–13 experiments. *p < 0.05 versus insulin. † p < 0.05 versus

307

insulin glargine. 17

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

308 To evaluate a safety profile of multi-SPRA-insulin, blood

309

3.6. Safety Profile of SPRA-insulin.

310

chemistry values, such as creatinine (CRE), blood urea nitrogen (BUN), aspartate aminotransferase (AST)

311

and alanine aminotransferase (ALT) were measured 24 h after subcutaneous administration of multi-

312

SPRA-insulin to rats (Table 1). A negligible change in blood chemistry values was observed after

313

subcutaneous administration of multi-SPRA-insulin, suggesting a high safety profile. Additionally, lower

314

hemolytic activity of multi-PEG-β-CyD than β-CyD (Figure S10), and negligible hemolytic activity of

315

multi-SPRA-insulin (Figure S11) supported that high safety of multi-SPRA-insulin after subcutaneous

316

administration.

317 318

319

Table 1. Blood Chemistry Values 24 h after Subcutaneous Administration of Multi-SPRA-insulin to Rats. System

CRE (mg/dL)

BUN (mg/dL)

AST (U/L)

ALT (U/L)

Saline

0.37 ± 0.01

23.1 ± 0.9

73 ± 11

44 ± 3

Insulin

0.35 ± 0.01

19.5 ± 0.8

77 ± 21

62 ± 5

Ad-insulin

0.31 ± 0.02

15.5 ± 1.3

69 ± 12

42 ± 3

multi-SPRA-insulin

0.33 ± 0.01

20.3 ± 1.1

81 ± 23

36 ± 6

Each value represents the mean ± S.E. of three experiments.

320 321

4. DISCUSSION

322

The development of a long-acting insulin drug without loss of the activity has proven to be challenging.

323

Currently, commercially available long-acting insulin products such as insulin/myristic acid (insulin

324

detemir) and insulin/hexadecanedioic acid (insulin degludec) conjugates are used in the clinical field, but

325

these drugs possess only 46% and 13–15% of the bioactivity versus native, unmodified insulin,

326

respectively.36, 37 In addition, PEG-lispro which was in Phase III of clinical trials only possesses 6% 18

ACS Paragon Plus Environment

Page 19 of 34

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

Molecular Pharmaceutics

327

activity when compared with that of insulin lispro.16, 18 To achieve advanced PEGylation of insulin that

328

does not lead to significant reduction in activity, a number of reversible PEGylation techniques have been

329

developed. Shechter et al. prepared reversible PEGylated insulin (PEG 40 kDa) through the 9-

330

hydroxymethyl-2-aminofluorene moiety and the resulting PEGylated insulin showed a prolonged

331

hypoglycemic effect.23 However, its in vitro bioactivity was 11% compared with the activity of insulin,

332

and a dose > 100-fold was required for in vivo study. Reversible PEGylation using complexation between

333

insulin and PEGylated basic oligopeptides through an electrostatic interaction38 and using lysine

334

modification through boronic acid39 have also been reported. However, both in vitro and in vivo

335

bioactivities of these PEGylated insulins were not examined.

336

In this context, SPRA technology dramatically improved the stabilities and prolonged the bioactivities

337

of insulin and lysozyme without their loss, highly probably owing to the reversible interaction between

338

Ad-proteins and PEG-β-CyD. Importantly, the addition of multi-PEG-β-CyD negligibly affected thermal

339

stability, enzymatic stability and hypoglycemic effect of insulin or lysozyme, suggesting that the

340

interaction of PEG-β-CyDs with Ad moiety of Ad-proteins is important for improvement of the

341

pharmaceutical properties of the proteins.

342

To achieve long and high bioactivity in vivo, the SPRA-insulin and SPRA-lysozyme should maintain

343

the supramolecular structure in blood and dissociate near the insulin receptor on cells or the rigid layer of

344

bacterial cell walls as its substrate, respectively. Herein, Stella et al. reported that a Kc > 104 M−1 for a

345

CyD/guest molecule complex is required to exist as an inclusion complex in vivo40. In addition, Leong et

346

al.41 demonstrated that sulfobutylether β-CyD is able to alter pharmacokinetics of Ad derivatives when

347

intravenously administered, resulting from complexation with a Kc value equal to ca. 2 × 104 M−1.

348

Moreover, Davis and coworkers developed the CyD nanoparticle containing siRNA in which transferrin

349

is modified through the host-guest interaction between β-CyD and Ad. This particle is stable in the blood,

350

and can accumulate in tumor after intravenous administration.31 Therefore, SPRA-proteins (Kc ~105 M−1)

351

should retain supramolecular structures following subcutaneous administration. 19

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

352

Meanwhile, the binding constants of insulin/insulin receptor and lysozyme/substrate are ca. ~109 M−1

353

and ~106 M−1, respectively,42, 43 evoking a competitive dissociation of SPRA-proteins in the presence of

354

their receptors or substrates, and subsequently leading to targeted activities. Thus, SPRA technology is

355

based on a reversible host–guest interaction and the Kc values were moderate (~105 M−1). This moderate

356

association could be suitable for complexation both in vitro and in vivo and for dissociation in the

357

presence of the receptor and substrate.

358

As the other possible mechanism for the in vivo prolonged effects of multi-SPRA-insulin, its absorption

359

via lymph could be attributed. In general, the absorption of high-molecular-weight drug into the blood is

360

restricted after subcutaneous administration by their limited permeability across the vascular endothelia.

361

In this case, the lymphatics provide an alternative absorption pathway from the interstitial space. Lymph

362

flows more slowly than blood, resulting in prolonged high blood drug levels.44 Therefore, multi-SPRA-

363

insulin could be absorbed through the lymph. Hereafter, we should investigate the pharmacokinetics of

364

multi-SPRA-insulin after subcutaneous administration.

365

In the present study, we fabricated reversible PEGylated proteins through the host-guest interaction

366

between PEG-β-CyDs and Ad-proteins. Future efforts should examine the complexation between Ad-

367

proteins and PEG-β-CyD under in vivo conditions or in the presence of receptor and substrate. In

368

addition, to exhibit whether SPRA-insulins retain the supramolecular structure in the blood, after this we

369

should investigate the pharmacokinetics of both PEG-β-CyDs and Ad-insulin.

370

covalently PEGylated insulin prepared by ourselves as a control. Hereafter, we should compare stability

371

and hypoglycemic effects of SPRA-insulins with PEG-lispro produced under GMP.

However, we used

372 373

5. CONCLUSION

374

In this study, we proposed a novel PEGylation technology, SPRA technology, via a host–guest interaction

375

between β-CyD and Ad. We believe that SPRA technology is one of the most successful PEGylation

376

approaches to improve the pharmaceutical properties of proteins without loss of bioactivity. In addition, 20

ACS Paragon Plus Environment

Page 21 of 34

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

Molecular Pharmaceutics

377

SPRA technology could be theoretically applied to a large number of peptides and proteins. Currently, we

378

are examining the utility of SPRA technology using various protein drugs. Moreover, SPRA technology

379

should be useful for the PEGylation of low molecular weight drugs, biomaterials and drug carriers.

380

Hereafter, detailed safety profiles, including immunogenicity of SPRA-proteins should be clarified. We

381

believe that SPRA technology has the potential as a generic method, surpassing conventional PEGylation

382

methods of proteins.

383 384

ASSOCIATED CONTENT

385

Supporting Information

386

The Supporting Information is available free of charge on the ACS Publications website at DOI:

387 388

AUTHOR INFORMATION

389

T.H. and T.H. contributed equally to this work.

390

Corresponding Author

391

*Address: Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-

392

ku, Kumamoto 862-0973, Japan. TEL: +81 96 371 4160, FAX: +81 96 371 4420, E mail:

393

[email protected].

394

Notes

395

This study was funded by Nihon Shokuhin Kako Co., Ltd. (Tokyo, Japan). K. Wada is researcher with

396

Nihon Shokuhin Kako Co., Ltd.

397 398

ACKNOWLEDGMENTS

399

The authors thank Nihon Shokuhin Kako Co., Ltd. (Tokyo, Japan) for providing β-CyD. This work was

400

partially supported by a Program for Leading Graduate Schools “HIGO (Health life science:

401

Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, a Grant-in-Aid for Young 21

ACS Paragon Plus Environment

Molecular Pharmaceutics

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 34

402

Scientists (Start-up) from the Ministry of Education, Science and Culture of Japan (23890161), Adaptable

403

and Seamless Technology Transfer Program through target-driven R&D, JST (AS262Z02650P) and

404

Kumayaku Research Support from the KUMAYAKU Alumni Research Foundation.

405 406

REFERENCES

407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445

(1) Morrison, C.; Lahteenmaki, R. Public biotech in 2014-the numbers. Nat. Biotechnol. 2015, 33, 703709. (2) Frokjaer, S.; Otzen, D. E. Protein drug stability: a formulation challenge. Nat. Rev. Drug Discov. 2005, 4, 298-306. (3) Harris, J. M.; Chess, R. B. Effect of pegylation on pharmaceuticals. Nat. Rev. Drug Discov. 2003, 2, 214-221. (4) Kolate, A.; Baradia, D.; Patil, S.; Vhora, I.; Kore, G.; Misra, A. PEG-a versatile conjugating ligand for drugs and drug delivery systems. J. Control. Release 2014, 192, 67-81. (5) Fishburn, C. S. The pharmacology of PEGylation: balancing PD with PK to generate novel therapeutics. J. Pharm. Sci. 2008, 97, 4167-4183. (6) Jarsch, M.; Brandt, M.; Lanzendorfer, M.; Haselbeck, A. Comparative erythropoietin receptor binding kinetics of C.E.R.A. and epoetin-β determined by surface plasmon resonance and competition binding assay. Pharmacology 2008, 81, 63-69. (7) Bailon, P.; Palleroni, A.; Schaffer, C. A.; Spence, C. L.; Fung, W. J.; Porter, J. E.; Ehrlich, G. K.; Pan, W.; Xu, Z. X.; Modi, M. W.; Farid, A.; Berthold, W.; Graves, M. Rational design of a potent, longlasting form of interferon: a 40 kDa branched polyethylene glycol-conjugated interferon α-2a for the treatment of hepatitis C. Bioconjug. Chem. 2001, 12, 195-202. (8) Yang, C.; Lu, D.; Liu, Z. How PEGylation enhances the stability and potency of insulin: a molecular dynamics simulation. Biochemistry 2011, 50, 2585-2593. (9) Armstrong, J. K.; Wenby, R. B.; Meiselman, H. J.; Fisher, T. C. The hydrodynamic radii of macromolecules and their effect on red blood cell aggregation. Biophys. J. 2004, 87, 4259-4270. (10) Zhao, H.; Yang, K.; Martinez, A.; Basu, A.; Chintala, R.; Liu, H. C.; Janjua, A.; Wang, M.; Filpula, D. Linear and branched bicin linkers for releasable PEGylation of macromolecules: controlled release in vivo and in vitro from mono- and multi-PEGylated proteins. Bioconjug. Chem. 2006, 17, 341-351. (11) Hinds, K.; Koh, J. J.; Joss, L.; Liu, F.; Baudys, M.; Kim, S. W. Synthesis and characterization of poly(ethylene glycol)-insulin conjugates. Bioconjug. Chem. 2000, 11, 195-201. (12) Hinds, K. D.; Kim, S. W. Effects of PEG conjugation on insulin properties. Adv. Drug Deliv. Rev. 2002, 54, 505-530. (13) Higashi, T.; Hirayama, F.; Misumi, S.; Arima, H.; Uekama, K. Design and evaluation of polypseudorotaxanes of pegylated insulin with cyclodextrins as sustained release system. Biomaterials 2008, 29, 3866-3871. (14) Higashi, T.; Hirayama, F.; Misumi, S.; Motoyama, K.; Arima, H.; Uekama, K. Polypseudorotaxane formation of randomly-pegylated insulin with cyclodextrins: slow release and resistance to enzymatic degradation. Chem. Pharm. Bull. 2009, 57, 541-544. (15) Torosantucci, R.; Kukrer, B.; Mero, A.; Van Winsen, M.; Tantipolphan, R.; Jiskoot, W. Plain and mono-pegylated recombinant human insulin exhibit similar stress-induced aggregation profiles. J. Pharm. Sci. 2011, 100, 2574-2585. (16) Madsbad, S. LY2605541-a preferential hepato-specific insulin analogue. Diabetes 2014, 63, 390392. 22

ACS Paragon Plus Environment

Page 23 of 34

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

446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494

Molecular Pharmaceutics

(17) Moore, M. C.; Smith, M. S.; Sinha, V. P.; Beals, J. M.; Michael, M. D.; Jacober, S. J.; Cherrington, A. D. Novel PEGylated basal insulin LY2605541 has a preferential hepatic effect on glucose metabolism. Diabetes 2014, 63, 494-504. (18) Caparrotta, T. M.; Evans, M. PEGylated insulin Lispro, (LY2605541)-a new basal insulin analogue. Diabetes Obes. Metab. 2014, 16, 388-395. (19) Danial, M.; van Dulmen, T. H.; Aleksandrowicz, J.; Potgens, A. J.; Klok, H. A. Site-specific PEGylation of HR2 peptides: effects of PEG conjugation position and chain length on HIV-1 membrane fusion inhibition and proteolytic degradation. Bioconjug. Chem. 2012, 23, 1648-1660. (20) Basu, A.; Yang, K.; Wang, M.; Liu, S.; Chintala, R.; Palm, T.; Zhao, H.; Peng, P.; Wu, D.; Zhang, Z.; Hua, J.; Hsieh, M. C.; Zhou, J.; Petti, G.; Li, X.; Janjua, A.; Mendez, M.; Liu, J.; Longley, C.; Zhang, Z.; Mehlig, M.; Borowski, V.; Viswanathan, M.; Filpula, D. Structure-function engineering of interferon-β-1b for improving stability, solubility, potency, immunogenicity, and pharmacokinetic properties by site-selective mono-PEGylation. Bioconjug. Chem. 2006, 17, 618-630. (21) Shaunak, S.; Godwin, A.; Choi, J. W.; Balan, S.; Pedone, E.; Vijayarangam, D.; Heidelberger, S.; Teo, I.; Zloh, M.; Brocchini, S. Site-specific PEGylation of native disulfide bonds in therapeutic proteins. Nat. Chem. Biol. 2006, 2, 312-313. (22) Kim, T. H.; Swierczewska, M.; Oh, Y.; Kim, A.; Jo, D. G.; Park, J. H.; Byun, Y.; Sadegh-Nasseri, S.; Pomper, M. G.; Lee, K. C.; Lee, S. Mix to validate: a facile, reversible PEGylation for fast screening of potential therapeutic proteins in vivo. Angew. Chem. Int. Ed. Engl. 2013, 52, 6880-6884. (23) Shechter, Y.; Mironchik, M.; Rubinraut, S.; Tsubery, H.; Sasson, K.; Marcus, Y.; Fridkin, M. Reversible pegylation of insulin facilitates its prolonged action in vivo. Eur. J. Pharm. Biopharm. 2008, 70, 19-28. (24) Nesher, M.; Vachutinsky, Y.; Fridkin, G.; Schwarz, Y.; Sasson, K.; Fridkin, M.; Shechter, Y.; Lichtstein, D. Reversible pegylation prolongs the hypotensive effect of atrial natriuretic peptide. Bioconjug. Chem. 2008, 19, 342-348. (25) Uekama, K.; Hirayama, F.; Irie, T. Cyclodextrin drug carrier systems. Chem. Rev. 1998, 98, 20452076. (26) Davis, M. E.; Brewster, M. E. Cyclodextrin-based pharmaceutics: past, present and future. Nat. Rev. Drug Discov. 2004, 3, 1023-1035. (27) Simoes, S. M.; Rey-Rico, A.; Concheiro, A.; Alvarez-Lorenzo, C. Supramolecular cyclodextrinbased drug nanocarriers. Chem. Commun. 2015, 51, 6275-6289. (28) Zhang, J.; Ma, P. X. Cyclodextrin-based supramolecular systems for drug delivery: recent progress and future perspective. Adv. Drug Deliv. Rev. 2013, 65, 1215-1233. (29) Arima, H.; Hayashi, Y.; Higashi, T.; Motoyama, K. Recent advances in cyclodextrin delivery techniques. Expert Opin. Drug Deliv. 2015, 12, 1425-1441. (30) Cromwell, W. C.; Byström, K.; Eftink, M. R. Cyclodextrin-adamantanecarboxylate inclusion complexes: Studies of the variation in cavity size. J. Phys. Chem. 1985, 89, 326-332. (31) Davis, M. E.; Zuckerman, J. E.; Choi, C. H.; Seligson, D.; Tolcher, A.; Alabi, C. A.; Yen, Y.; Heidel, J. D.; Ribas, A. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010, 464, 1067-1070. (32) Uekama, K.; Minami, K.; Hirayama, F. 6A-O-[(4-biphenylyl)acetyl]-α-, -β-, and -γ-cyclodextrins and 6A-deoxy-6A-[[(4-biphenylyl)acetyl]amino]-α-, -β-, and -γ-cyclodextrins: potential prodrugs for colon-specific delivery. J. Med. Chem. 1997, 40, 2755-2761. (33) Okamatsu, A.; Motoyama, K.; Onodera, R.; Higashi, T.; Koshigoe, T.; Shimada, Y.; Hattori, K.; Takeuchi, T.; Arima, H. Folate-appended β-cyclodextrin as a promising tumor targeting carrier for antitumor drugs in vitro and in vivo. Bioconjug. Chem. 2013, 24, 724-733. (34) Hirotsu, T.; Higashi, T.; Motoyama, K.; Hirayama, F.; Uekama, K.; Arima, H. Improvement of pharmaceutical properties of insulin through conjugation with glucuronylglucosyl-β-cyclodextrin. J. Incl. Phenom. Macrocycl. Chem. 2014, 80, 107-112. 23

ACS Paragon Plus Environment

Molecular Pharmaceutics

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

495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518

Page 24 of 34

(35) Higashi, T.; Hirayama, F.; Yamashita, S.; Misumi, S.; Arima, H.; Uekama, K. Slow-release system of pegylated lysozyme utilizing formation of polypseudorotaxanes with cyclodextrins. Int. J. Pharm. 2009, 374, 26-32. (36) Markussen, J.; Havelund, S.; Kurtzhals, P.; Andersen, A. S.; Halstrom, J.; Hasselager, E.; Larsen, U. D.; Ribel, U.; Schaffer, L.; Vad, K.; Jonassen, I. Soluble, fatty acid acylated insulins bind to albumin and show protracted action in pigs. Diabetologia 1996, 39, 281-288. (37) Gough, S. C.; Harris, S.; Woo, V.; Davies, M. Insulin degludec: overview of a novel ultra longacting basal insulin. Diabetes Obes. Metab. 2013, 15, 301-309. (38) Tsiourvas, D.; Sideratou, Z.; Sterioti, N.; Papadopoulos, A.; Nounesis, G.; Paleos, C. M. Insulin complexes with PEGylated basic oligopeptides. J. Colloid. Interface Sci. 2012, 384, 61-72. (39) Cal, P. M.; Frade, R. F.; Cordeiro, C.; Gois, P. M. Reversible lysine modification on proteins by using functionalized boronic acids. Chemistry 2015, 21, 8182-8187. (40) Stella, V. J.; Rao, V. M.; Zannou, E. A.; Zia, V. V. Mechanisms of drug release from cyclodextrin complexes. Adv. Drug Deliv. Rev. 1999, 36, 3-16. (41) Leong, N. J.; Prankerd, R. J.; Shackleford, D. M.; McIntosh, M. P. The effect of intravenous sulfobutylether7-β-cyclodextrin on the pharmacokinetics of a series of adamantane-containing compounds. J. Pharm. Sci. 2015, 104, 1492-1498. (42) Gammeltoft, S.; Gliemann, J. Binding and degradation of 125I-labelled insulin by isolated rat fat cells. Biochim. Biophys. Acta. 1973, 320, 16-32. (43) Malcolm, B. A.; Rosenberg, S.; Corey, M. J.; Allen, J. S.; de Baetselier, A.; Kirsch, J. F. Sitedirected mutagenesis of the catalytic residues Asp-52 and Glu-35 of chicken egg white lysozyme. Proc. Natl. Acad. Sci. U S A 1989, 86, 133-137. (44) McLennan, D. N.; Porter, C. J.; Charman, S. A. Subcutaneous drug delivery and the role of the lymphatics. Drug Discov. Today Technol. 2005, 2, 89-96.

519 520

24

ACS Paragon Plus Environment

Page 25 of 34

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

Molecular Pharmaceutics

mono-PEG-b-CyD

mono-SPRA-insulin (Kc = 4.8 x 105 M-1)

Ad-insulin

multi-PEG-b-CyD

multi-SPRA-insulin (PEG : ave. 2.8)

(Kc = 2.7 x 104 M-1)

PEG-insulin

mono-PEG-b-CyD

mono-SPRA-lysozyme (Kc = 6.4 x 104 M-1)

Ad-lysozyme

multi-PEG-b-CyD

(PEG : ave. 2.8)

multi-SPRA-lysozyme (Kc = 9.3 x 104 M-1)

PEG-lysozyme

ACS Paragon Plus Environment

Figure 1

Molecular Pharmaceutics

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

A. Insulin

Page 26 of 34

C. Lysozyme

14,308

5,734

B. Ad-insulin

D. Ad-lysozyme 5,910

5000

6000

14,483

7000

m/z

14000

16000

m/z

ACS Paragon Plus Environment

Figure 2

Page 27 of 34

Molecular Pharmaceutics

A. mono-PEG-b-CyD (OH)6

p-Toluenesulfonyl chloride

= =

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

O O-SO

-CH3

(OH)6

NaN3

N3

H2O NaOH a.q.

Azido b-CyD

Tosylated b-CyD

(OH)7

Activated PEG (20 kD)

b-CyD

Ph3P (OH)6

O COON O

NH3 a.q. NH2

DMF/H2O

Amino b-CyD

mono-PEG-b-CyD

B. multi-PEG-b-CyD (Cl)7

(N3)7

NaN3

CH3SO2Cl DMAc/H2O

DMF

(OH)7

per-Chloro b-CyD

per-Azido b-CyD Activated PEG (20 kD)

b-CyD

Ph3P

NH3 a.q. (NH2)7

O COON O

DMF/H2O

per-Amino b-CyD

multi-PEG-b-CyD

C. PEG (20 kD)

E. multi-PEG-b-CyD

D. mono-PEG-b-CyD

18

20

x

103

22

24

20

40

m/z

60

x

103

80

100

120

m/z

ACS Paragon Plus Environment

Figure 3

Molecular Pharmaceutics

Number (%)

A. mono-SPRA-insulin

PEG-b-CyD/Ad-Protein 0.1 0.5

30

1 2 10

20 10

Ad-insulin

0 1

10

100

10

100

10

100

10

100

Number (%)

B. multi-SPRA-insulin 30 20 10

Ad-insulin

0 1

Number (%)

C. mono-SPRA-lysozyme 30 20 10

Ad-lysozyme

0 1

D. multi-SPRA-lysozyme Number (%)

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

Page 28 of 34

30 20 10

Ad-lysozyme

0 1

Size (nm)

ACS Paragon Plus Environment

Figure 4

Page 29 of 34

A. SPRA-insulin

B. SPRA-lysozyme

: multi-SPRA-insulin : PEG-insulin : mono-SPRA-insulin : Ad-insulin : Insulin

* * * *

120 100

: multi-SPRA-lysozyme : PEG-lysozyme : mono-SPRA-lysozyme : Ad-lysozyme : Lysozyme

* * *

* * * *

80

*

60 40 20 0

2

4

6

Lysozyme remaining (%)

Insulin remaining (%)

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

Molecular Pharmaceutics

120

*

*

100

*

80

* * *

60 40

*

* * *

20 0

2

4

6

Time (h)

Time (day)

ACS Paragon Plus Environment

Figure 5

Molecular Pharmaceutics

B. SPRA-lysozyme

100 80 60 40 20 0

*

*

Lysozyme undegraded (%)

A. SPRA-insulin Insulin undegraded (%)

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

Page 30 of 34

100

*

80 60

*

*

40 20 0

ACS Paragon Plus Environment

Figure 6

Page 31 of 34

120

Relative activity (%)

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

Molecular Pharmaceutics

100 80 60 40

*

20 0

ACS Paragon Plus Environment

Figure 7

Molecular Pharmaceutics

Serum insulin level (ng/mL)

A. Serum insulin level

30 : Insulin (i.v., 2 U/kg) : Insulin (s.c., 2 U/kg) : multi-SPRA-insulin (s.c., 2 U/kg)

* 20 10

*

0

2

*

4

6

8

Time (h)

Serum glucose level (%)

B. Serum glucose level 120

120

100

100

: PEG-insulin (2 U/kg) 40 : Ad-insulin (2 U/kg) : Insulin (2 U/kg) 20 : multi-SPRA-insulin (2 U/kg)

40 20 2

4

6

8 10 Time (h)

12

: Insulin glargine (2 U/kg) : multi-SPRA-insulin (2 U/kg)

0

24

C. MRTG

2

4

6

8 10 Time (h)

12

24

D. AUCG *

*† * AUCG (%・h)

6

† †

60

60

0

80

*

80

MRTG (h)

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

Page 32 of 34

4

2

*

600

400

200

* 0

0

ACS Paragon Plus Environment

Figure 8

Page 33 of 34

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

Molecular Pharmaceutics

Adamantane-protein conjugate

Dissociation

PEGylated b-cyclodextrin

SPRA-protein

ACS Paragon Plus Environment

Molecular Pharmaceutics

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

Adamantane-protein conjugate Dissociation

PEGylated b-cyclodextrin

SPRA-protein

ACS Paragon Plus Environment

Page 34 of 34