8
UNCORRECTED PROOF 1 Theranostic nanoparticles for future personalized medicine Ju Hee Q1 Ryu a , Sangmin Lee a , Sohee Son a , Sun Hwa Kim a , James F. Leary b , Kuiwon Choi a , Ick Chan Kwon a,c, 3 a Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, Republic of Korea 4 b Basic Medical Sciences and Biomedical Engineering, Bindley Bioscience Center and Brick Nanotechnology Center, Purdue University, West Lafayette 47907, USA 5 c KU-KIST School, Korea University, 1 Anam-dong, Seongbuk-gu, Seoul 136-701, Republic of Korea abstract 6 article info 7 Article history: 8 Received 11 February 2014 9 Accepted 12 April 2014 10 Available online xxxx 11 Keywords: 12 Theranostics 13 Personalized medicine 14 Molecular imaging 15 Monitoring response 16 The concept of personalized medicine has recently emerged as a promising way to address unmet medical needs. 17 Due to the limitations of standard diagnostic and therapeutic strategies, the disease treatment is moving towards 18 tailored treatment for individual patients, considering the inter-individual variability in therapeutic response. 19 Theranostics, which involves the combination of therapy and diagnostic imaging into a single system, may fulll 20 the promise of personalized medicine. By integrating molecular imaging functionalities into therapy, theranostic 21 approach could be advantageous in therapy selection, treatment planning, objective response monitoring and 22 follow-up therapy planning based on the specic molecular characteristics of a disease. Although the eld of 23 therapy and imaging of its response have been independently developed thus far, developing imaging strategies 24 can be fully exploited to revolutionize the theranostic systems in combination with the therapy modality. In this 25 review, we describe the recent advances in molecular imaging technologies that have been specically developed 26 to evaluate the therapeutic efcacy for theranostic purposes. 27 © 2014 Published by Elsevier B.V. 28 29 30 31 32 1. Introduction 33 The paradigm for the disease treatment is changing; clinicians are 34 gradually going beyond the traditional one-size-ts-allapproach of 35 medicine toward a new era of personalized treatment strategies [1,2]. 36 In such approaches, medical treatments are tailored to the specic 37 characteristics of individual patients [3]. No single therapeutic 38 agent is recognized to produce the same effect in different patients 39 with a single type of disease. Considering the inter-individual variability 40 in therapeutic response, a patient-specic treatment may produce 41 better therapeutic outcomes, while reducing patient discomfort and 42 undesirable side-effects [4]. 43 Theranostics, the integration of therapeutic and diagnostic capability 44 in a single system, may contribute signicantly to the ever-growing 45 eld of personalized medicine [5]. By combining molecular imaging 46 functionalities with therapy, a theranostic approach could be advanta- 47 geous in identifying and selecting a particular subgroup of patients 48 with a specic molecular phenotype. A particular molecular phenotype 49 can be an indication of positive response to a certain treatment. This is 50 especially signicant because treatment using monoclonal antibodies, 51 such as cetuximab and trastuzumab is increasing. In addition, this 52 approach can serve as a valuable tool in the selection of a safe and 53 efcacious dose, recognition of adverse effects at the early stages of 54 therapy, and real-time objective monitoring of the therapeutic re- 55 sponse [6]. For example, clinicians can switch to other therapeutic 56 options, or they can adjust the therapeutic dose for non-responders to 57 the current therapeutic regimen, while continuing the therapy for 58 good responders. 59 In particular, nanotechnology could be applied advantageously to 60 theranostics [7]. The most commonly used nano-sized materials for 61 theranostic purposes are polymeric nanoparticle, lipid-based nano- 62 particle, dendrimer, cage protein and inorganic nanoparticle [810]. 63 These nano-sized particles (NPs) provide numerous advantages for 64 the theranostic applications. One of the greatest advantageous charac- 65 teristics of NPs is their potential to localize in pathological lesions 66 more than normal lesions in vivo, particularly in the case of cancer 67 [11,12]. NPs can easily extravasate from blood vessels into tumor tissues 68 because of leaky and irregularly dilated blood vessels at the tumor site, 69 and they can be retained at tumor sites owing to poor lymphatic 70 drainage. This phenomenon of selective accumulation of NPs near 71 tumor tissues is termed the enhanced permeability and retention 72 (EPR) effect [13,14], allowing their targeted imaging and therapy. 73 In addition to cancer, EPR-like effect is known to be observed in the 74 inammation region because various immunocytes release vasodilators 75 and chemotactic factors in the inammation region [1517]. Another 76 promising characteristic of NPs is their large surface-area-to-volume 77 ratio; this allows for a high loading capacity of imaging probes, thera- 78 peutic drugs, or targeting moieties [18,19]. NPs with multiple functions 79 can remarkably benet monitoring treatment progress and evaluating 80 outcomes, because they can be used to diagnose and track certain Journal of Controlled Release xxx (2014) xxxxxx Corresponding author at: Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, Republic of Korea. Tel.: +82 2 958 5912; fax: +82 2 958 5909. E-mail address: [email protected] (I.C. Kwon). COREL-07137; No of Pages 8 http://dx.doi.org/10.1016/j.jconrel.2014.04.027 0168-3659/© 2014 Published by Elsevier B.V. Contents lists available at ScienceDirect Journal of Controlled Release journal homepage: www.elsevier.com/locate/jconrel Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for future personalized medicine, J. Control. Release (2014), http://dx.doi.org/ 10.1016/j.jconrel.2014.04.027

Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

1

2Q1

345

6

78910

1112131415

27

2829

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

Journal of Controlled Release xxx (2014) xxx–xxx

COREL-07137; No of Pages 8

Contents lists available at ScienceDirect

Journal of Controlled Release

j ourna l homepage: www.e lsev ie r .com/ locate / jconre l

Theranostic nanoparticles for future personalized medicine

F

Ju Hee Ryu a, Sangmin Lee a, Sohee Son a, Sun Hwa Kim a, James F. Leary b, Kuiwon Choi a, Ick Chan Kwon a,c,⁎a Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, Republic of Koreab Basic Medical Sciences and Biomedical Engineering, Bindley Bioscience Center and Brick Nanotechnology Center, Purdue University, West Lafayette 47907, USAc KU-KIST School, Korea University, 1 Anam-dong, Seongbuk-gu, Seoul 136-701, Republic of Korea

O

⁎ Corresponding author at: Center for Theragnosis, BiomInstitute of Science and Technology (KIST), Hwarangno136-791, Republic of Korea. Tel.: +82 2 958 5912; fax:

E-mail address: [email protected] (I.C. Kwon).

http://dx.doi.org/10.1016/j.jconrel.2014.04.0270168-3659/© 2014 Published by Elsevier B.V.

Please cite this article as: J.H. Ryu, et al., Ther10.1016/j.jconrel.2014.04.027

Oa b s t r a c t

a r t i c l e i n f o

16

17

18

19

20

21

22

23

24

Article history:Received 11 February 2014Accepted 12 April 2014Available online xxxx

Keywords:TheranosticsPersonalized medicineMolecular imagingMonitoring response

25

26

ED P

R

The concept of personalizedmedicine has recently emerged as a promisingway to address unmetmedical needs.Due to the limitations of standard diagnostic and therapeutic strategies, the disease treatment ismoving towardstailored treatment for individual patients, considering the inter-individual variability in therapeutic response.Theranostics, which involves the combination of therapy and diagnostic imaging into a single system, may fulfillthe promise of personalizedmedicine. By integrating molecular imaging functionalities into therapy, theranosticapproach could be advantageous in therapy selection, treatment planning, objective response monitoring andfollow-up therapy planning based on the specific molecular characteristics of a disease. Although the field oftherapy and imaging of its response have been independently developed thus far, developing imaging strategiescan be fully exploited to revolutionize the theranostic systems in combination with the therapymodality. In thisreview, we describe the recent advances inmolecular imaging technologies that have been specifically developedto evaluate the therapeutic efficacy for theranostic purposes.

© 2014 Published by Elsevier B.V.

T

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

UNCO

RREC1. Introduction

The paradigm for the disease treatment is changing; clinicians aregradually going beyond the traditional “one-size-fits-all” approach ofmedicine toward a new era of personalized treatment strategies [1,2].In such approaches, medical treatments are tailored to the specificcharacteristics of individual patients [3]. No single therapeuticagent is recognized to produce the same effect in different patientswith a single type of disease. Considering the inter-individual variabilityin therapeutic response, a patient-specific treatment may producebetter therapeutic outcomes, while reducing patient discomfort andundesirable side-effects [4].

Theranostics, the integration of therapeutic and diagnostic capabilityin a single system, may contribute significantly to the ever-growingfield of personalized medicine [5]. By combining molecular imagingfunctionalities with therapy, a theranostic approach could be advanta-geous in identifying and selecting a particular subgroup of patientswith a specific molecular phenotype. A particular molecular phenotypecan be an indication of positive response to a certain treatment. This isespecially significant because treatment using monoclonal antibodies,such as cetuximab and trastuzumab is increasing. In addition, thisapproach can serve as a valuable tool in the selection of a safe and

75

76

77

78

79

80

edical Research Institute, Korea14-gil 6, Seongbuk-gu, Seoul+82 2 958 5909.

anostic nanoparticles for futu

efficacious dose, recognition of adverse effects at the early stages oftherapy, and real-time objective monitoring of the therapeutic re-sponse [6]. For example, clinicians can switch to other therapeuticoptions, or they can adjust the therapeutic dose for non-responders tothe current therapeutic regimen, while continuing the therapy forgood responders.

In particular, nanotechnology could be applied advantageously totheranostics [7]. The most commonly used nano-sized materials fortheranostic purposes are polymeric nanoparticle, lipid-based nano-particle, dendrimer, cage protein and inorganic nanoparticle [8–10].These nano-sized particles (NPs) provide numerous advantages forthe theranostic applications. One of the greatest advantageous charac-teristics of NPs is their potential to localize in pathological lesionsmore than normal lesions in vivo, particularly in the case of cancer[11,12]. NPs can easily extravasate fromblood vessels into tumor tissuesbecause of leaky and irregularly dilated blood vessels at the tumor site,and they can be retained at tumor sites owing to poor lymphaticdrainage. This phenomenon of selective accumulation of NPs neartumor tissues is termed the enhanced permeability and retention(EPR) effect [13,14], allowing their targeted imaging and therapy.In addition to cancer, EPR-like effect is known to be observed in theinflammation region because various immunocytes release vasodilatorsand chemotactic factors in the inflammation region [15–17]. Anotherpromising characteristic of NPs is their large surface-area-to-volumeratio; this allows for a high loading capacity of imaging probes, thera-peutic drugs, or targeting moieties [18,19]. NPs with multiple functionscan remarkably benefit monitoring treatment progress and evaluatingoutcomes, because they can be used to diagnose and track certain

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 2: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

T

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

2 J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

EC

molecular targets of highly heterogeneous diseases such as cancer withmultiple-targeting possibilities and sensitive imaging capabilities [20,21].

For theranostic purposes, a variety of nanomaterials such aspolymers and silica have been extensively investigated. Thesenanomaterials have been utilized for loading both imaging agentsand drugs. Polymer-based NPs are the most widely investigatedtheranostic nanoplatform. Polymer-based NPs, usually fabricated bythe self-assembly of amphiphilic polymers, can load hydrophobicdrugs or imaging agents in the inner cores or functionalize their surfacewith imaging agents or targeting moieties. These encapsulating andsurface-modifying properties of polymer-based NPs mean that they canpotentially serve as prospective theranostic agents [22]. Furthermore,silica-based NPs are advantageous in terms of their well-establishedsiloxane chemistry and well-defined tunable nanostructures. Theseproperties allow for the effective fabrication of a desired surface inthe silica-based NPs for therapeutic and diagnostic applications[11]. Besides classical drug delivery systems such as polymer-basedNPs and silica-based NPs, many nanomaterials including iron oxideor gold-based NPs have intrinsic imaging abilities, which can furtherbe loaded with drugs for theranostics. Iron oxide-based NPs can benoninvasively imaged by magnetic resonance (MR) imaging and uti-lized for drug delivery, which makes them a potential theranosticnanoplatform. To prevent the aggregation of iron oxide-based NPsin biological media, the modification of their surface with hydrophilicpolymers such as dextran is preferred. Gold-based NPs have beenwidely researched as multifunctional agents for theranostic purposesbecause they have distinctive optical characteristics and uniquephotothermal properties along with low toxicity and well-establishedgold chemistry.

Theranostic NPs simultaneously carrying both imaging probes andtherapeutic agents were primarily utilized to investigate drug deliveryprocess and drug release pattern [23]. Theranostic approaches to drugresponse monitoring remain in their developmental stages up to date,and the integration of therapy and the imaging of its response intoone theranostic systemhas not yet been studied in detail. In this review,we describe recent imaging strategies that have been specifically devel-oped for the evaluation of the therapeutic efficacy for theranosticpurposes.

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

UNCO

RR2. Treatment with theranostic NPs

Theranostic NPs can be applied for cancer therapy including chemo-therapy, siRNA therapy, and photodynamic therapy. Some small-molecule drugs have disadvantages including toxic side effects in nor-mal tissues, insufficient specificity to tumor tissues, limited delivery totumor cells owing to their hydrophobicity, and drug-resistance [7].Theranostic NPs have the potential to solve these problems in tradition-al chemotherapy [24,25]. Furthermore, a theranostic NP delivery systemcan be beneficial for siRNA therapy. Theranostic NPs can be effective atenhancing the stability of siRNA in the blood stream after intravenousinjection, minimizing degradation by many enzymes. Considering thatnaked siRNA can be eliminated from the blood within 5 min after intra-venous injection, prolonged circulation time of siRNA in NPs cancontribute to effective therapy. In addition, negatively charged siRNAcannot easily enter the cytosol of target cells, which can be overcomeby theranostic NPs for siRNA therapy [26–28]. In the case of photody-namic therapy, theranostic NP-based photodynamic therapy affordsadvantages including reduced systemic toxicity and improved solubilityin water compared to classical photodynamic therapy. The selectiveaccumulation of photosensitizers contained in NPs inside the targetcan significantly lower the systemic toxicity related to classical photo-dynamic therapy. In addition, most photosensitizers used in photody-namic therapy can aggregate in biological media, leading to a changein their optical properties [29].

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

ED P

RO

OF

3. Imaging modalities for theranostics

In theranostic approaches, diagnostic imaging is essential for deter-mining the presence and degree of molecular targets for a certaindisease [30,31]. Presently, various noninvasive imaging modalities canbe applied to visualize molecular targets in vivo: optical imaging, ultra-sound (US) imaging, MR imaging, computed tomography (CT) and nu-clear imaging (single photon emission computed tomography (SPECT)and positron emission tomography (PET)). These modalities can bebroadly separated into primarily molecular imagingmodalities and pri-marily morphological/anatomical imaging modalities [32]. The former,which are characterized by high sensitivity, include optical imagingand PET/SPECT; on the other hand, the latter, which are featured byhigh spatial resolution, include CT, US and MR [32,33]. The detailed ad-vantages and disadvantages of individual imaging modalities would beleft out because they have been previously discussed inmany literatures[4,34]. In particular, optical imaging and PET/SPECT have great potentialto detectmolecular targets that are important in a disease process; thus,they are more favorable for theranostic approaches. In more recent,integrated molecular/anatomical systems (e.g., SPECT/CT) have beendeveloped to combine the strengths of individual imaging modalities[35,36].

4. Monitoring certain molecular targets involved indisease progression

Targeted molecular imaging, an essential component of a theranosticsystem, can be used to image and trackmolecular targets involved in dis-ease progression. Noninvasive molecular imaging can be used to identifythe presence and degree of molecular targets through a whole-body scanin real time, which is valuable in disease staging and therapy planningsuch as drug dosage and timing. For example, most chemotherapeuticagents, such as doxorubicin (DOX), exert their effects by inducing apopto-sis, which is the process of programmed cell death. Therefore, detectingandquantifying apoptosiswould be useful formonitoring the chemother-apeutic efficacy. A useful approach for assessing apoptosis is to monitorthe specific molecules involved in apoptosis signaling. Caspases are agroupof cysteineproteases that play key roles in apoptosis [37]. They con-vey the signal in apoptotic caspase cascades by cleaving and amplifyingmultiple caspases through various pathways, which ultimately contrib-utes to cell death [38]. Among various caspases, caspase-3 iswidely recog-nized as an important and direct mediator of apoptosis [39]. Our groupdeveloped nanoprobes for detecting caspase-3 activity [40]. This nano-probe consists of hyaluronic acid-based polymeric nanoparticles(HA-NPs) and a caspase-3 substrate peptide-based probe (Fig. 1).The peptide-based probe comprises Cy5.5, a near infrared (NIR)dye, and black hole quencher-3 (BHQ-3), a dark quencher, covalentlylinked to both end parts of a short peptide (Gly-Asp-Glu-Val-Asp-Ala-Pro-Lys-Gly-Cys) that can serve as a hydrolyzable substrate forcaspase-3 (Fig. 1A). The synthesized peptide-based probe is conju-gated onto the surface of HA-NPs to afford Cas3-HA-NP (Fig. 1A, B).The NIR dye is strongly dual-quenched because of the dye–dyequenching and dye–quencher interaction mechanisms, contributingto reduced background signals. In the presence of caspase-3, Cas3-HA-NP showed the linear proportional recovery of NIR fluorescencesignals depending on the caspase-3 concentration in vitro (Fig. 1C).Cas3-HA-NP was activated to produce a strong high-resolution fluo-rescence signal in real time in apoptotic cells induced by the tumornecrosis factor-related apoptosis-inducing ligand (TRAIL). NIR fluo-rescence signals produced by Cas3-HA-NP were detected as early as15min post-treatment of TRAIL; in contrast, annexinV staining, the cur-rent gold standard target for evaluating apoptosis, yielded apoptosissignals 1 h post-treatment (Fig. 1D). In addition, Cas3-HA-NP enabledclear in vivo visualization of apoptosis induced by DOX in tumor-bearing mice (Fig. 1E–G). NIR fluorescence signals produced by Cas3-HA-NP were well-matched with apoptosis signals analyzed by the

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 3: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

CO

RRECTED P

RO

OF

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

E

CA

F

G

Quencher (BHQ-3)

NIR dye(Cy5.5)

caspase-3-specific

substrate

hyaluronic acid-5β-cholanic acid conjugate

NIRF quenched polymeric

nanoprobe for apoptosis imaging

(Cas3-HA-NP)

active caspase -3

0 200 400 600 800 10000

20

40

60

80

100

Rel

ativ

e in

tens

ity (

%)

Size (nm)

300 nm

120

60

30

15

0 nmol /L

Cas-3

Tumor /(+)DOX Tumor / (-) DOX Normal tissue

1 min

15 min

30 min

0 10 20 300

20

40

60

80 Tmour/(+)DOX Tumor/(-)DOX Normal tissue

Flu

ores

cenc

e in

tens

ity(p

hoto

n co

unts

in tu

mor

x10

4 )

Time (min)

Tumor

(+)DOX

Tumor

(-)DOX

Normal

tissue

DIC

NIRF

B

D

10 m

0 min 15 min 30 min 1 h 2 h

Cas3 -HA-NP/(+)Trail

Annexin V-FITC/(+)Trail

NIRF

FITC μ

Fig. 1.Q9 Fluorescence-activatable nanoprobe, and cellular and in vivo caspase-3 imaging. (A) Schematic diagram of caspase-3-sensitive nanoprobe (Cas3-HA-NP). (B) Size distribution andmorphological shape (TEM image) of the Cas3-HA-NPs. (C) Fluorescence image of a 96-wellmicroplate containing the Cas3-HA-NPs in thepresence of various concentrations of caspase-3.(D) Real time video imaging of caspase-3 activity in HCT116 cells using the Cas3-HA-NPs. Cells were incubatedwith the Cas3-HA-NPs (20 μg/mL) for 30min, and then treatedwith tumornecrosis factor-related apoptosis inducing ligand (TRAIL, 100 ng/mL). Apoptosis of cells treated with TRAIL was also evaluated by the annexin V-FITC staining. (E) In vivo near-infraredfluorescence imaging of subcutaneous SCC7 tumor-bearing mice after intratumoral injection of the Cas3-HA-NPs with or without pretreatment of doxorubicin (DOX). (F) Quantitativeanalysis by counting total photons in the Cas3-HA-NP-treated tumor tissues as a function of time (n = 5). (G) Ex vivo tumor tissue images from (E).

3J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNterminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)

assay, which is used to detect apoptosis in dissected tumor tissues.Therefore, this imaging probe (Cas3-HA-NP) can be applied to monitorthe therapeutic response and efficacy of apoptosis-related drugs in cancertherapy. Importantly, this imaging probe platform for detecting proteasesis so flexible that it can be applied to different proteases by simply chang-ing the cleavable substrate peptide [41–44].

Recently, amoremultiplexed system (MultiCas-NP) has been devisedto target three types of caspases (caspase-3, -8 and -9) involved in apo-ptotic signaling cascades (Fig. 2) [45]. This nanoprobe is based on ananoquencher system comprising mesoporous silica NPs incorporatedinto a series of dark quenchers to quench a variety of dyes (Fig. 2A).Different caspase-specific substrate peptides linked with various dyeswere conjugated onto the surface of the nanoquencher system for thesimultaneous detection of caspase-3, -8 and -9. As expected, variouscaspases activated specific substrate peptides linked with the dye,

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

producing the particular signals of the corresponding dye (Fig. 2B, C).MultiCas-NP boostedmultiplexedfluorescence intensities in the presenceof multiple caspases in a single apoptotic cell induced by TRAIL (Fig. 2D).More importantly, MultiCas-NP has great potential for in vivo use tomon-itor the efficacy of apoptosis-related treatment in individual patients inreal time. These imaging probes for monitoring particular molecular tar-gets involved in disease progression can be used to revolutionizetheranostic systems in combination with the therapy modality.

5. Monitoring downstream molecular targets affected by treatment

The efficacy of a treatment can be evaluated by monitoring certainmolecules in a downstream process affected by the treatment as wellas the targeted molecules of the treatment. In addition to chemothera-peutic drugs, small interference RNA (siRNA) has attracted considerableattention as a potential therapy for the treatment of various diseases,

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 4: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

UNCO

RRECTED P

RO

OF

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

A

Cetyltrimethylammonium

bromide micelleHexagonal array Nanoquencher (Q-MSN)

Crosslinker-

nanoquencher Multiquenched

nanosensor Activated nanosensor

(MultiCas-NP)

Dye substrate (e.g. Cy5.5-GDEVDGC) Quenched dye substrate Target proteases (e.g.caspases)

Crosslinker

(NHS-PEG12-maleimide)

Nucleus 8 9 3 Merge

Caspases

F /

F0

Time/min →

C

F →

Wavenumber/nm →

DB

0 h

0.5

h1

h2

hIn

h. 2

h

Fig. 2. Multiplex fluorescence-activatable nanoprobe for caspase cascade imaging. (A) Schematic diagram of various caspases (caspase-3, -8, and -9)-sensitive multiplex nanoprobe(MultiCas-NP). Image: TEM image of the nanoquenchers (Q-MSNs). (B) Fluorescence-quenching properties of Q-MSNs concurrently labeled with FPG-456 (green), FPR-560 (orange),Cy5.5 (dark blue), and FPI-774 (light blue). Upper lines = fluorescence emission spectra of free fluorescence dyes; lower lines in gray zone = emission spectra of fluorescence dyes afterquenching by Q-MSNs. (C) The relative increase in fluorescence (F/F0) of MultiCas-NPs in the presence (upper lines) or absence (lower lines, gray zone) of caspase-3 (blue), caspase-8(green), or caspase-9 (orange). (D) Multiplex imaging of the caspase cascade in HCT116 cells. Blue (DAPI) = nucleus; green (FPG456) = caspase-8; purple (FPG560) = caspase-9; red(Cy5.5) = caspase-3; inh. = with inhibitor. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

4 J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

especially owing to its specific therapeutic effects [46,47]. The actionmechanism of siRNA is post-transcriptional gene silencing by cleavagingthe target mRNA that shares a homologous sequence with the treatedsiRNA [48,49]. This sequence specific suppression of targeted genecan be utilized for efficient therapy through the decrease of targetedmolecules related with the disease [50].

Our group reported thiolated glycol chitosan (tGC) nanoparticlesencapsulating tumor necrosis factor (TNF)-α targeted polymerizedsiRNA (poly-siRNA) for the treatment of rheumatoid arthritis (RA)(Fig. 3) [17]. Among a variety of proinflammatory cytokines associated

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

in the pathogenesis of RA, TNF-α plays amajor role to induce chronic in-flammation via stimulating the release of multiple cytokines and otherinflammatory mediators (Fig. 3A) [51,52]. Therefore, TNF-α inhibitionhas been an important target for RA treatment [53,54]. Poly-siRNA, pre-pared through the self-polymerization of thiol groups at the 5′ end oftheir sense and antisense strands, can be easily encapsulated into tGCpolymers through charge–charge interaction and chemical cross-linkingto produce psi-tGC-NPs (Fig. 3B–D) [55–57]. The TNF-α-targeted siRNAloaded psi-tGC-NPs exhibited excellent in vitro gene silencing for TNF-αand significant in vivo inhibition of inflammation and bone erosion in

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 5: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

ECTED P

RO

OF

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

-SH

S-S

Glycol chitosan

Macrophage

siRNA releaseby Glutathione

TNF- targetedmRNA degradation

TNF -α

MMP

Destruction of bone and cartilage

A B

C D

charge-charge interaction

crosslinkingPoly-siRNA(psi)

Thiolated glycol chitosan (tGC)

psi-tGC-NPs

psi-tGC-NPs

2 5 10 +DTT

200 300 400 500 600 700 8000

20

40

60

80

100

120

Inte

nsity

(a.

u.)

Diameter (nm)

E F GNone

psi-tGC-NPs

2 3 4 5 6 7

0

1

2

3

4

5 None psi-tGC-NPs MTX

Art

hriti

c sc

ore

Time (week)

Normal 3 weeks 4 weeks 5 weeks 7 weeksNone

psi-tGC-NPs

MTX

α

Fig. 3. Polymerized siRNA/thiolated glycol chitosan nanoparticles (psi-tGC-NPs) for rheumatoid arthritis (RA) therapy. (A) Schematic diagram of psi-tGC-NPs for tumor necrosis factor(TNF)-α targeted siRNA delivery in RA therapy. (B) Complexation of poly-siRNA with tGC polymers. (C) Confirmation of poly-siRNA formation by gel electrophoresis of mono- orpoly-siRNA and gel retardation of complexes of thiolated GC with poly-siRNA (psi-tGC-NPs) at different weight ratios (1:2, 1:5, and 1:10 tGC/siRNA). +DTT: Reduction of poly-siRNAby adding 1,4-dithiothreitol (DTT). (D) Size distribution and morphological shape (TEM image) of the psi-tGC-NPs. (E) In vivo near-infrared fluorescence imaging of normal mice andarthritis mice in each group (untreated (none), psi-tGC-NP-treated, and methotrexate (MTX)-treated) after intravenous injection of MMP-3 nanoprobe (MMP-3-NP) at different stagesof disease (at 3, 4, 5, and 7 weeks after the primary immunization) for monitoring of the disease progression in arthritis mice. (F) The paw swelling in each group of (E). (G) Arthriticscore in each group of (E).

5J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RR

collagen-induced arthritis mouse models, as evaluated by microCT orimmunoblot analysis (Fig. 3E–G). Importantly, disease progressionfollowing psi-tGC-NP therapy was evaluated by monitoring of matrixmetalloproteinase-3 (MMP-3) in a downstream process affected by thetherapy using in vivo NIR fluorescence imaging (Fig. 3E). MMPs havebeen known to play a pivotal role in persistent synovial inflammationand articular cartilage degradation, both of which are early symptomsof RA [58]. Among several MMPs related in these processes, MMP-3 isrecognized to be particularly significant in the pathogenesis of RA [59].MMP-3 is known to degrade many proteins of matrices, including colla-gens, proteoglycans, gelatins, laminin andfibronectin [59].More interest-ingly, MMP is known to be downregulated by inhibiting TNF-α inarthritic progression [60,61]. This probe for detecting MMP-3 consists ofthe same platform as Cas3-HA-NP, which is described above. The MMP-3 substrate peptide-based probe comprises cleavable substrate peptide(Gly-Val-Pro-Leu-Ser-Leu-Thr-Met-Gly-Lys-Gly-Gly), Cy5.5, and BHQ-3.These peptide-based probes are then conjugated onto the surface ofglycol chitosan NPs, to afford MMP-3-NP. In our previous study, the NIRfluorescence signal from MMP-3-NP was apparently able to indicate thepresence and severity of arthritic disease [16]. Significantly reduced NIRfluorescence signals from MMP-3-NP were observed in the psi-tGC-NPs-treated group compared with the untreated control group incollagen-induced arthriticmice. Therefore,monitoring amolecular targetin a downstream process affected by the treatment can be a potential ap-proach for assessing the clinical effectiveness of the treatment.

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

6. Using a nanostructure similar to therapeutic vehicles

A nanostructure identical to that used for drug treatment can beloaded with an imaging agent for the real-time visualization of drugefficacy. Following the treatment using a nanostructure loaded with atherapeutic agent, a structurally identical nanostructure labeled withan imaging agent can be applied to monitor the therapeutic response.At first, this theranostic approach has been applied to receptor-targeting radiolabeled peptides and not nanostructures. For instance,somatostatin receptors that are over-expressed in many neuroendo-crine tumors can bind to a specific peptide sequence with high bindingaffinity [62]. Thus, somatostatin receptor-binding peptides labeledwith a therapeutic (β-emitting) radionuclide (177Lu) could eradicatesomatostatin-receptor-positive endocrine tumors. In particular, somato-statin receptor-binding peptides labeled with a diagnostic (γ-emitting)radionuclide (111In) for SPECT could visualize binding sites inreceptor-expressing neuroendocrine tumors in a non-invasive man-ner, allowing the monitoring of the progress and outcome aftersurgery, radiotherapy, or chemotherapy. In addition, patients witha neuroendocrine pancreatic tumor were diagnosed using 111In-octreotide, a radiolabeled somatostatic analogue, after three radio-nuclide therapy cycles using 177Lu-octreotate [63]. They usedoctreotide and octreotate as target peptides with high affinity partic-ularly for somatostatic receptor type 2. In SPECT imaging, no discern-ible uptake was observed in the tumor sites after three radionuclide

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 6: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341Q2

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

6 J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

therapy cycles, revealing the good therapeutic efficacy of radionu-clide therapy.

Recently, our group reported that a nanostructure similar to atherapeutic one can be useful for the real-time visualization of treatmentefficacy (Fig. 4). A PEGylated hyaluronic acid nanoparticle (P-HA-NP)was utilized for the early diagnosis, targeted therapy and chemothera-peuticmonitoring of colon cancer in tumormodel [64]. P-HA-NPs encap-sulating irinotecan (IRT), an anticancer drug, selectively targeted tumorsthrough the EPR effect of NPs and strong receptor-binding to CD44, over-expressing receptors in various tumors (Fig. 4A). As a result, the P-HA-NPencapsulating IRT (P-HA-NP–IRT)-treated group exhibited significantinhibition of tumor growth compared to the free IRT or saline-treatedgroups, as confirmed through measurements of the tumor size (Fig. 4B).To monitor the therapeutic response, Cy5.5 was conjugated onto thesurface of P-HA-NPs (Cy5.5–P-HA-NPs). Indeed, the fluorescence inten-sity of Cy5.5–P-HA-NPs in the P-HA-NP–IRT-treated group was notablylower than that in the free IRT or saline-treated groups, which is attrib-uted to the decreased accumulation of Cy5.5–P-HA-NPs at tumor sitesby a combination of passive targeting (EPR effect) and active targeting(CD44 binding) in the case of suppressed tumor growth (Fig. 4C) [65,66]. Using a nanostructure identical to that used for treatment for diag-nostic purposes can be useful not only formonitoring the therapeutic re-sponse but also for selecting patients who will benefit from an identicalnanostructure loadedwith a therapeutic agent. If the presence of imagingagents (Cy5.5 attachment onto the NP surface) and therapeutic agents(IRT loading into NP) does not affect the shape, size and surface featuresof the nanostructure, specific pharmacokinetics and biodistribution ofdiagnostic nanostructures in individual patients can be identical tothose of therapeutic nanostructures [67]. Therefore, patient-specifictreatment seems possible by varying the biodistribution and pharmaco-kinetics of the therapeutic nanostructure.

The integration of therapy and imaging of its response into onetheranostic system was described in chitosan-based NPs (CNPs)

UNCO

RRECT

Therapeutic NP (P-HA-NP-IRT)

Diagnostic NP (Cy5.5-P-HA-NP)

Tumorigenesis(AOM/DSS treatment)

Diagnostics

Therapeut

84 87 90 9

84 d

Time (day)0 5 10 15 20 25 30

Tu

mo

r V

olu

me

(mm

3 )

200

400

600

800

1000

1200

1400ControlIRT (10 mg/kg)IRT-P-HA-NP(10 mg/kg)

*

Su

rviv

al R

ate

(%)

20

40

60

80

100

A

B

Fig. 4. PEGylated hyaluronic acid nanoparticles (P-HA-NPs) for the diagnosis, therapy, and chemrescence imaging of small-sizedHT29flank tumors (top-left), liver-implantedCT26 colon tumoof Cy5.5 conjugated to P-HA-NPs (Cy5.5–P-HA-NPs). Arrows indicate the sites of tumors. (B)treated with saline, free irinotecan (IRT), and IRT-encapsulated P-HA-NPs (P-HA-NPs–IRT)orthotopic colon cancer models after treatment of saline, IRT, or P-HA-NPs–IRT. NIR fluorescen

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

ED P

RO

OF

(Fig. 5) [68]. Different formulations of PTX–CNPs were prepared byloading with different amounts of PTX (5, 10, or 20 mg PTX/kg) andby labeling with Cy5.5 (Cy5.5–CNP–PTX) (Fig. 5A, B). In these formula-tions, loaded PTX amounts were adjusted to be correlated with the NIRfluorescence intensity of each NP. Therefore, howmuch PTX can be de-livered to the tumor sites in SCC7 tumor-bearing mouse models can bevisualized by NIR fluorescence signals. Strong NIR fluorescence signalsin the tumors were displayed with the frequency of injections atthree-day intervals; thus determining optimal protocol is administrationof Cy5.5–CNP–PTX at three-day intervals (Fig. 5C). PTX–CNPs treatedwith this optimal administration protocol resulted in the improved re-duction in tumor volume compared to treatment of free PTX (Fig. 5D,E). A standard for determining optimal protocol in this study was thedrug concentration in the tumor sites. If therapy was integrated withthe imaging functionalities to evaluate drug efficacy into one theranosticsystem, drug efficacy can be a standard for deciding the optimal drugdosage in question, enhancing both safety and effectiveness of the drug.

7. Conclusions

This review describes recent imaging strategies that have beenspecifically developed for evaluating the therapeutic efficacy fortheranostic purposes. Diagnostic imaging utilizing certain moleculartargets involved in disease progression, downstream molecular targetsaffected by treatment, and a nanostructure similar to a therapeuticone could potentially be used to evaluate the therapeutic responseand efficacy during or after treatment. Unfortunately, the current statusof theranostic technology for drug response monitoring has remainedin the developmental stage; thus far, few studies have focused on the in-tegration of therapy and its response imaging into one theranostic sys-tem. However, the call for a more personalized approach to medicaltreatment and extensive ongoing developments in nanotechnology to-gether with diagnostic imaging strategies discussed in this review

ics

3 96 d

Therapeutic monitoring

125 d

Time (day)0 5 10 15 20 25 30

Control IRT (10 mg/kg)IRT-P-HA-NP(10 mg/kg)

Control(125 d)

IRT(125 d)

P-HA-NP-IRT(125 d)

C

otherapeuticmonitoring of colon cancer. (A) Diagnostics: In vivo near-infrared (NIR) fluo-rs (top-right), and early stage orthotopic colon tumors (bottom) after intravenous injectionTherapeutics: Tumor growth and survival rate of human colon cancer (HT29) xenograftsat a dose of 10 mg IRT/kg. (C) Therapeutic monitoring: Representative colon images ofce images were acquired at 6 h after intravenous injection of Cy5.5–P-HA-NPs.

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 7: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

TED P

RO

OF

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393Q3

394

395396

397398399400401402403404405406407408409410411412413414415416417418419420421422423424425426427428429430431432433434Q4435436

A B

C D

E

Cy5.5 labeled glycol chitosan-5β-cholanic acid conjugate

hydrophilic polymer shell

NIRF Cy5.5

PTX/hydrophobic inner multicores

tumor tissue

1day 2day 3day 4day 5day 6day

7day 8day 9day 10day 11day 12day

13day 14day 15day 16day 17day 18day

2 5 8 11 14 17 200

2000

4000

6000

8000

10000

* *

saline CNPs PTX (20 mg/kg) PTX-CNPs (20 mg/kg)

Tum

or v

olum

e (m

m3 )

Time (days)

Time (days)0 5 10 15 20 25 30

0

20

40

60

80

100

saline CNP PTX (20 mg/kg) PTX-CNPs (20 mg/kg)P

erce

nt S

urvi

val (

%)

(Cy5.5-CNP-PTX)

Fig. 5. Theranostic chitosan-based nanoparticles (CNPs) for simultaneous diagnosis, chemotherapy, and therapeutic monitoring of cancer. (A) Schematic diagram of self-assembled CNPslabeled with Cy5.5 for imaging and therapeutic monitoring, and encapsulated with paclitaxel (PTX) for chemotherapy (Cy5.5–CNPs–PTX). (B) Chemical structure of glycol chitosan–cholanic acid conjugates labeled with Cy5.5. (C) In vivo near-infrared fluorescence imaging of tumor specificity and tumor growth rate in Cy5.5–CNPs–PTX (20 mg PTX/kg)-injectedSCC7 tumor bearing mice. The black arrows indicate intravenous injection of Cy5.5–CNPs–PTX. (D) Therapeutic efficacy of PTX–CNPs in SCC7 tumor-bearing mice. (E) Survival curves.

7J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RRECmight revolutionize to theranostic systems in the near future. Some

important issues, however, must be preferentially resolved to achievesuccessful integration into theranostic systems. In particular, theranosticsystems need to improve the apparent mismatch between the optimalconcentrations of imaging agents and therapeutic agents for potentialclinical use, since the optimal concentration for a desired therapy is gen-erally much higher than that required for imaging. Furthermore, atheranostic system can contain multiple functions within an individualsystem, and thismay increase the complexity of the system. Thus, the de-velopment of reproducible and simplemethods for designing theranosticsystems can help in achieving the successful integration of imaging andtherapy. In addition, to accelerate the clinical translation of theranosticNP, utilizing clinically validated nanomaterials could lower the risk oftranslation [69]. Diagnostic imaging for monitoring therapeutic efficacyhas been shown to play an important role in many ways of personalizedmedicine. The development of diagnostic imaging following treatment,desirably in a single system, could enable therapy selection, treatmentplanning, objective responsemonitoring and follow-up therapy planningbased on the specific molecular characteristics of a disease, thus pavingthe way for personalized medicine.

Acknowledgments

This study was funded by the National Research Foundation ofKorea (NRF-2013K1A1A2032346 and 2012K1A1A2A01056095) andthe Intramural Research Program (Theragnosis) of KIST.

References

[1] N.B. La Thangue, D.J. Kerr, Predictive biomarkers: a paradigm shift towards personal-ized cancer medicine, Nat. Rev. Clin. Oncol. 8 (2011) 587–596.

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

[2] M. Diamandis, N.M.A. White, G.M. Yousef, Personalized medicine: marking a newepoch in cancer patient management, Mol. Cancer Res. 8 (2010) 1175–1187.

[3] M. Kalia, Personalized oncology: recent advances and future challenges, Metabolism62 (Supplement 1) (2013) S11–S14.

[4] J.V. Jokerst, S.S. Gambhir, Molecular imaging with theranostic nanoparticles, Acc.Chem. Res. 44 (2011) 1050–1060.

[5] T.H. Kim, S. Lee, X. Chen, Nanotheranostics for personalized medicine, Expert. Rev.Mol. Diagn. 13 (2013) 257–269.

[6] G. Bartlett, J. Antoun, N.K. Zgheib, Theranostics in primary care: pharmacogenomicstests and beyond, Expert. Rev. Mol. Diagn. 12 (2012) 841–855.

[7] J.H. Ryu, H. Koo, I.-C. Sun, S.H. Yuk, K. Choi, K. Kim, I.C. Kwon, Tumor-targetingmulti-functional nanoparticles for theragnosis: new paradigm for cancer therapy, Adv.Drug Deliv. Rev. 64 (2012) 1447–1458.

[8] A. Puri, R. Blumenthal, Polymeric lipid assemblies as novel theranostic tools, Acc.Chem. Res. 44 (2011) 1071–1079.

[9] B. Sumer, J. Gao, Theranostic nanomedicine for cancer, Nanomedicine 3 (2008)137–140.

[10] H. Koo, M.S. Huh, I.-C. Sun, S.H. Yuk, K. Choi, K. Kim, I.C. Kwon, In vivo targeted de-livery of nanoparticles for theranosis, Acc. Chem. Res. 44 (2011) 1018–1028.

[11] C.M. Wang LS, J.A. Ho, Nanotheranostics—a review of recent publications, Int. J.Nanomedicine 7 (2012) 4679–4695.

[12] I. Brigger, C. Dubernet, P. Couvreur, Nanoparticles in cancer therapy and diagnosis,Adv. Drug Deliv. Rev. 54 (2002) 631–651.

[13] H. Maeda, Macromolecular therapeutics in cancer treatment: the EPR effect andbeyond, J. Control. Release 164 (2012) 138–144.

[14] H. Maeda, H. Nakamura, J. Fang, The EPR effect for macromolecular drug delivery tosolid tumors: improvement of tumor uptake, lowering of systemic toxicity, anddistinct tumor imaging in vivo, Adv. Drug Deliv. Rev. 65 (2013) 71–79.

[15] Y.-N. Chang, H. Guo, J. Li, Y. Song, M. Zhang, J. Jin, G. Xing, Y. Zhao, Adjusting thebalance between effective loading and vector migration of macrophage vehicles todeliver nanoparticles, PLoS ONE 8 (2013) e76024.

[16] J.H. Ryu, A. Lee, J.-U. Chu, H. Koo, C.-Y. Ko, H.S. Kim, S.-Y. Yoon, B.-S. Kim, K. Choi, I.C.Kwon, K. Kim, I. Youn, Early diagnosis of arthritis in mice with collagen-inducedarthritis, using a fluorogenic matrix metalloproteinase 3-specific polymeric probe,Arthritis Rheum. 63 (2011) 3824–3832.

[17] S.J. Lee, A. Lee, S.R. Hwang, J.-S. Park, J. Jang,M.S. Huh, D.-G. Jo, S.-Y. Yoon, Y. Byun, S.H.Kim, I.C. Kwon, I. Youn, K. Kim, TNF-[alpha] gene silencing using polymerized siRNA/thiolated glycol chitosan nanoparticles for rheumatoid arthritis, Mol. Ther. (2013).

[18] M.P. Melancon, R.J. Stafford, C. Li, Challenges to effective cancer nanotheranostics, J.Control. Release 164 (2012) 177–182.

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/

Page 8: Journal of Controlled Release - Purdue Universityjfleary/Nanomedicine... · OOF 81 moleculartargetsof highly heterogeneous diseasessuchascancerwith 82 multiple-targeting possibilities

T

437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468469470471472473474Q5475476477478479480481482483484485486487488489490491492493494495496497498499500501502503504505506507508509

510511512513514515516517518519520521522523524525526527528529Q6530531532533534535536537538539540541542543544545546547548549550Q7551552553554555556557558559560Q8561562563564565566567568569570571572573574575576577578579580581582

583

8 J.H. Ryu et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RREC

[19] J.D. Byrne, T. Betancourt, L. Brannon-Peppas, Active targeting schemes for nanopar-ticle systems in cancer therapeutics, Adv. Drug Deliv. Rev. 60 (2008) 1615–1626.

[20] S. Basu, Personalized versus evidence-based medicine with PET-based imaging, Nat.Rev. Clin. Oncol. 7 (2010) 665–668.

[21] V. Sanna, M. Sechi, Nanoparticle therapeutics for prostate cancer treatment,Nanomedicine: Nanotechnology, Biology and Medicine 8 (Supplement 1) (2012)S31–S36.

[22] S. Mura, P. Couvreur, Nanotheranostics for personalized medicine, Adv. Drug Deliv.Rev. 64 (2012) 1394–1416.

[23] T. Lammers, S. Aime,W.E. Hennink, G. Storm, F. Kiessling, Theranostic nanomedicine,Acc. Chem. Res. 44 (2011) 1029–1038.

[24] A. Khdair, C. Di, Y. Patil, L. Ma, Q.P. Dou, M.P.V. Shekhar, J. Panyam, Nanoparticle-mediated combination chemotherapy and photodynamic therapy overcomestumor drug resistance, J. Control. Release 141 (2010) 137–144.

[25] T. Yoshitomi, Y. Ozaki, S. Thangavel, Y. Nagasaki, Redox nanoparticle therapeutics tocancer—increase in therapeutic effect of doxorubicin, suppressing its adverse effect,J. Control. Release 172 (2013) 137–143.

[26] R. Shrestha, M. Elsabahy, H. Luehmann, S. Samarajeewa, S. Florez-Malaver, N.S. Lee,M.J.Welch, Y. Liu, K.L.Wooley, Hierarchically assembled theranostic nanostructures forsiRNA delivery and imaging applications, J. Am. Chem. Soc. 134 (2012) 17362–17365.

[27] K.A. Whitehead, R. Langer, D.G. Anderson, Knocking down barriers: advances insiRNA delivery, Nat. Rev. Drug Discov. 8 (2009) 129–138.

[28] F. Alexis, E. Pridgen, L.K. Molnar, O.C. Farokhzad, Factors affecting the clearance andbiodistribution of polymeric nanoparticles, Mol. Pharm. 5 (2008) 505–515.

[29] P. Rai, S. Mallidi, X. Zheng, R. Rahmanzadeh, Y. Mir, S. Elrington, A. Khurshid, T.Hasan, Development and applications of photo-triggered theranostic agents, Adv.Drug Deliv. Rev. 62 (2010) 1094–1124.

[30] S.M. Janib, A.S. Moses, J.A. MacKay, Imaging and drug delivery using theranosticnanoparticles, Adv. Drug Deliv. Rev. 62 (2010) 1052–1063.

[31] M.F. Kircher, H. Hricak, S.M. Larson, Molecular imaging for personalized cancer care,Mol. Oncol. 6 (2012) 182–195.

[32] J.K. Willmann, N. van Bruggen, L.M. Dinkelborg, S.S. Gambhir, Molecular imaging indrug development, Nat. Rev. Drug Discov. 7 (2008) 591–607.

[33] Y. Huang, S. He, W. Cao, K. Cai, X.-J. Liang, Biomedical nanomaterials for imaging-guided cancer therapy, Nanoscale 4 (2012) 6135–6149.

[34] G. Kramer-Marek, J. Gore, M. Korc, Molecular imaging in pancreatic cancer—aroadmap for therapeutic decisions, Cancer Lett. 341 (2013) 132–138.

[35] M. de Smet, S. Langereis, S. van den Bosch, K. Bitter, N.M. Hijnen, E. Heijman, H.Grüll, SPECT/CT imaging of temperature-sensitive liposomes for MR-image guideddrug delivery with high intensity focused ultrasound, J. Control. Release, 169 82–90

[36] A.E. John, J.C. Luckett, A.L. Tatler, R.O. Awais, A. Desai, A. Habgood, S. Ludbrook, A.D.Blanchard, A.C. Perkins, R.G. Jenkins, J.F. Marshall, Preclinical SPECT/CT imaging ofαvβ6 integrins for molecular stratification of idiopathic pulmonary fibrosis, J.Nucl. Med. 54 (2013) 2146–2152.

[37] D.L. Vaux, S.J. Korsmeyer, Cell death in development, Cell 96 (1999) 245–254.[38] M. Brentnall, L. Rodriguez-Menocal, R. De Guevara, E. Cepero, L. Boise, Caspase-9,

caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis, BMC CellBiol. 14 (2013) 32.

[39] S.J. Riedl, Y. Shi, Molecular mechanisms of caspase regulation during apoptosis, Nat.Rev. Mol. Cell Biol. 5 (2004) 897–907.

[40] S. Lee, K.Y. Choi, H. Chung, J.H. Ryu, A. Lee, H. Koo, I.-C. Youn, J.H. Park, I.-S. Kim, S.Y.Kim, X. Chen, S.Y. Jeong, I.C. Kwon, K. Kim, K. Choi, Real time, high resolution videoimaging of apoptosis in single cells with a polymeric nanoprobe, Bioconjug. Chem.22 (2011) 125–131.

[41] S. Lee, J.H. Ryu, K. Park, A. Lee, S.-Y. Lee, I.-C. Youn, C.-H. Ahn, S.M. Yoon, S.-J. Myung,D.H. Moon, X. Chen, K. Choi, I.C. Kwon, K. Kim, Polymeric nanoparticle-basedactivatable near-infrared nanosensor for protease determination in vivo, Nano Lett.9 (2009) 4412–4416.

[42] J.H. Ryu, J.H. Na, H.K. Ko, D.G. You, S. Park, E. Jun, H.J. Yeom, D.H. Seo, J.H. Park, S.Y.Jeong, I.-S. Kim, B.-S. Kim, I.C. Kwon, K. Choi, K. Kim, Non-invasive optical imagingof cathepsin B with activatable fluorogenic nanoprobes in various metastaticmodels, Biomaterials 35 (2014) 2302–2311.

[43] J.H. Ryu, S.A. Kim, H. Koo, J.Y. Yhee, A. Lee, J.H. Na, I. Youn, K. Choi, I.C. Kwon, B.-S.Kim, K. Kim, Cathepsin B-sensitive nanoprobe for in vivo tumor diagnosis, J. Mater.Chem. 21 (2011).

[44] J. Ryu, A. Lee, J. Na, S. Lee, H. Ahn, J. Park, C.-H. Ahn, B.-S. Kim, I. Kwon, K. Choi, I.Youn, K. Kim, Optimization of matrix metalloproteinase fluorogenic probes for oste-oarthritis imaging, Amino Acids 41 (2011) 1113–1122.

[45] X. Huang,M. Swierczewska, K.Y. Choi, L. Zhu, A. Bhirde, J. Park, K. Kim, J. Xie, G. Niu, K.C.Lee, S. Lee, X. Chen,Multiplex imaging of an intracellular proteolytic cascade by using abroad-spectrum nanoquencher, Angew. Chem. Int. Ed. 51 (2012) 1625–1630.

[46] S. Yang, Y. Chen, R. Ahmadie, E.A. Ho, Advancements in the field of intravaginalsiRNA delivery, J. Control. Release 167 (2013) 29–39.

Please cite this article as: J.H. Ryu, et al., Theranostic nanoparticles for futu10.1016/j.jconrel.2014.04.027

ED P

RO

OF

[47] S. Zhang, B. Zhao, H. Jiang, B. Wang, B. Ma, Cationic lipids and polymers mediatedvectors for delivery of siRNA, J. Control. Release 123 (2007) 1–10.

[48] H.Y. Yoon, H.R. Kim, G. Saravanakumar, R. Heo, S.Y. Chae, W. Um, K. Kim, I.C. Kwon, J.Y. Lee, D.S. Lee, J.C. Park, J.H. Park, Bioreducible hyaluronic acid conjugates as siRNAcarrier for tumor targeting, J. Control. Release 172 (2013) 653–661.

[49] R. Kanasty, J.R. Dorkin, A. Vegas, D. Anderson, Deliverymaterials for siRNA therapeu-tics, Nat. Mater. 12 (2013) 967–977.

[50] M.E. Davis, J.E. Zuckerman, C.H.J. Choi, D. Seligson, A. Tolcher, C.A. Alabi, Y. Yen, J.D.Heidel, A. Ribas, Evidence of RNAi in humans from systemically administered siRNAvia targeted nanoparticles, Nature 464 (2010) 1067–1070.

[51] M. Feldmann, R.N.Maini, TNF defined as a therapeutic target for rheumatoid arthritisand other autoimmune diseases, Nat. Med. 9 (2003) 1245–1250.

[52] Y. Komano, N. Yagi, I. Onoue, K. Kaneko, N. Miyasaka, T. Nanki, Arthritic joint-targeting small interfering RNA-encapsulated liposome: implication for treat-ment strategy for rheumatoid arthritis, J. Pharmacol. Exp. Ther. 340 (2012)109–113.

[53] J.-L. Davignon, M. Hayder, M. Baron, J.-F. Boyer, A. Constantin, F. Apparailly, R.Poupot, A. Cantagrel, Targeting monocytes/macrophages in the treatment of rheu-matoid arthritis, Rheumatology 52 (2013) 590–598.

[54] A. Alghasham, Z. Rasheed, Therapeutic targets for rheumatoid arthritis: Progress andpromises, Autoimmunity, 0 1–18.

[55] S.J. Lee, M.S. Huh, S.Y. Lee, S. Min, S. Lee, H. Koo, J.-U. Chu, K.E. Lee, H. Jeon, Y. Choi, K.Choi, Y. Byun, S.Y. Jeong, K. Park, K. Kim, I.C. Kwon, Tumor-homing poly-siRNA/glycol chitosan self-cross-linked nanoparticles for systemic siRNA delivery in cancertreatment, Angew. Chem. Int. Ed. 51 (2012) 7203–7207.

[56] S.J. Lee, J.Y. Yhee, S.H. Kim, I.C. Kwon, K. Kim, Biocompatible gelatin nanoparticles fortumor-targeted delivery of polymerized siRNA in tumor-bearing mice, J. Control.Release 172 (2013) 358–366.

[57] S. Son, S. Song, S.J. Lee, S. Min, S.A. Kim, J.Y. Yhee, M.S. Huh, I. Chan Kwon, S.Y. Jeong,Y. Byun, S.H. Kim, K. Kim, Self-crosslinked human serum albumin nanocarriers forsystemic delivery of polymerized siRNA to tumors, Biomaterials 34 (2013)9475–9485.

[58] I. Tchetverikov, L.R. Lard, J. DeGroot, N. Verzijl, J.M. TeKoppele, F.C. Breedveld, T.W.J.Huizinga, R. Hanemaaijer, Matrix metalloproteinases-3, -8, -9 as markers of diseaseactivity and joint damage progression in early rheumatoid arthritis, Ann. Rheum.Dis. 62 (2003) 1094–1099.

[59] M.D. Posthumus, P.C. Limburg, J. Westra, H.A. Cats, R.E. Stewart, M.A. van Leeuwen,M.H. van Rijswijk, Serum levels of matrix metalloproteinase-3 in relation to thedevelopment of radiological damage in patients with early rheumatoid arthritis,Rheumatology 38 (1999) 1081–1087.

[60] A.I. Catrina, J. Lampa, S. Ernestam, E. Af Klint, J. Bratt, L. Klareskog, A.K. Ulfgren, Anti‐tumour necrosis factor (TNF)‐α therapy (etanercept) down‐regulates serummatrixmetalloproteinase (MMP)‐3 and MMP‐1 in rheumatoid arthritis, Rheumatology 41(2002) 484–489.

[61] A. Lee, Y. Qiao, G. Grigoriev, J. Chen, K.-H. Park-Min, S.H. Park, L.B. Ivashkiv, G.D.Kalliolias, Tumor necrosis factor α induces sustained signaling and a prolongedand unremitting inflammatory response in rheumatoid arthritis synovialfibroblasts,Arthritis Rheum. 65 (2013) 928–938.

[62] M. van Essen, A. Sundin, E.P. Krenning, D.J. Kwekkeboom, Neuroendocrine tumours:the role of imaging for diagnosis and therapy, Nat. Rev. Endocrinol. (2013) (advanceonline publication).

[63] M. de Jong, W.A.P. Breeman, D.J. Kwekkeboom, R. Valkema, E.P. Krenning, Tumorimaging and therapy using radiolabeled somatostatin analogues, Acc. Chem. Res.42 (2009) 873–880.

[64] K.Y. Choi, E.J. Jeon, H.Y. Yoon, B.S. Lee, J.H. Na, K.H. Min, S.Y. Kim, S.-J. Myung, S. Lee,X. Chen, I.C. Kwon, K. Choi, S.Y. Jeong, K. Kim, J.H. Park, Theranostic nanoparticlesbased on PEGylated hyaluronic acid for the diagnosis, therapy and monitoring ofcolon cancer, Biomaterials 33 (2012) 6186–6193.

[65] K.Y. Choi, H.Y. Yoon, J.-H. Kim, S.M. Bae, R.-W. Park, Y.M. Kang, I.-S. Kim, I.C. Kwon, K.Choi, S.Y. Jeong, K. Kim, J.H. Park, Smart nanocarrier based on PEGylated hyaluronicacid for cancer therapy, ACS Nano 5 (2011) 8591–8599.

[66] J.-H. Park, H.-J. Cho, H.Y. Yoon, I.-S. Yoon, S.-H. Ko, J.-S. Shim, J.-H. Cho, J.H. Park,K. Kim, I.C. Kwon, D.-D. Kim, Hyaluronic acid derivative-coated nanohybrid lipo-somes for cancer imaging and drug delivery, J. Control. Release 174 (2014)98–108.

[67] S. Svenson, Theranostics: are we there yet? Mol. Pharm. 10 (2013) 848–856.[68] K. Kim, J.H. Kim, H. Park, Y.-S. Kim, K. Park, H. Nam, S. Lee, J.H. Park, R.-W. Park, I.-S.

Kim, K. Choi, S.Y. Kim, K. Park, I.C. Kwon, Tumor-homing multifunctional nanoparti-cles for cancer theragnosis: simultaneous diagnosis, drug delivery, and therapeuticmonitoring, J. Control. Release 146 (2010) 219–227.

[69] J. Shi, Z. Xiao, N. Kamaly, O.C. Farokhzad, Self-assembled targeted nanoparticles:evolution of technologies and bench to bedside translation, Acc. Chem. Res. 44(2011) 1123–1134.

re personalized medicine, J. Control. Release (2014), http://dx.doi.org/