Top Banner
UNCORRECTED PROOF 1 Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug 2 loaded into stealth lipid nanocapsules 3 Anne-Laure Q1 Laine a,b Q2 , Anne Clavreul b,c , Audrey Rousseau d , Clément Tétaud b,c , 4 Anne Vessieres e , Emmanuel Garcion a,b , Gerard Jaouen e , Léo Aubert f , Matthieu Guilbert f , 5 Jean-Pierre Benoit a,b , Robert-Alain Toillon f , Catherine Passirani a,b, 6 a LUNAM Université Micro et Nanomédecines Biomimétiques, Angers, France 7 b Inserm U1066, IBS-CHU, Angers, France 8 c Département de Neurochirurgie CHU, Angers, France 9 d Département de Pathologie Cellulaire et Tissulaire CHU, Angers, France 10 e CNRS, UMR 7576, ENSCP, Paris, France 11 f Inserm U908, Université Lille 1, Villeneuve d'Ascq, France 12 Received 12 September 2013; accepted 3 May 2014 13 Abstract 14 In this work, a novel ferrocenyl complex (ansa-FcdiOH) was assessed for brain tumor therapy through stealth lipid nanocapsules (LNCs). 15 Stealth LNCs, prepared according to a one-step process, showed rapid uptake by cancer cells and extended blood circulation time. The 16 ferrocenyl complex was successfully encapsulated into these LNCs measuring 40 nm with a high loading capacity (6.4%). In vitro studies 17 showed a potent anticancer effect of ansa-FcdiOH on 9L cells with a low IC50 value (0.1 μM) associated with an oxidative stress and a dose- 18 dependent alteration of the cell cycle. Repeated intravenous injections of stealth ansa-FcdiOH LNCs in ectopic glioma bearing rats induced a 19 significant tumor growth inhibition, supported by a reduced number of proliferative cells in tumors compared to control group. Additionally, 20 no liver damage manifestation was observed in treated animals. These results indicated that stealth ansa-FcdiOH LNCs might be considered 21 as a potential new approach for cancer chemotherapy. 22 Q3 © 2014 Published by Elsevier Inc. 23 Key words: ansa-FcdiOH; Nanomedicine; Cell cycle; EPR effect 24 25 Q4 Introduction 26 Malignant brain tumor remains one of the main medical 27 challenges for scientists as no life-saving treatment has been 28 found to date despite aggressive therapy modalities (Stupp 29 protocol). 1 Consequently, alternative strategies are needed to 30 tackle this dismal tumor. 31 Over the past few years, Pr. Jaouen and his team have 32 struggled to develop potent bioorganometalic complexes for 33 anticancer application. Their chemistry is based on the coupling 34 between a ferrocene Q5 moiety and a tamoxifen-like skeleton, and 35 the resulting complex belongs to the Q6 ferrocifen family. 2,3 Several 36 molecules from their collection have already showed strong 37 antiproliferative activities on experimental glioma and breast 38 cancer models. 47 Recently, a new ferrocenyl complex, ansa- 39 FcdiOH (Figure 1), has emerged and turned out to be the most 40 effective agent according to in vitro antiproliferative assays. 41 Interestingly, ansa-FcdiOH was tested on 60 cancer cell lines of 42 the National Cancer Institute (NCI) and showed to be strongly 43 active on CNS (central nervous system) cancer cells. In addition, 44 its cytotoxic behavior did not match with any of the 171 reference 45 molecules from the NCI database (COMPARE analysis). 2 So far, 46 its mechanism of action is not fully understood although the role 47 played by its redox properties has been already underlined. 8 48 Therefore, ansa-FcdiOH was selected in this work to further study 49 its anticancer activity in vitro and in vivo on 9L brain tumors. 50 Due to the hydrophobic characteristic of this molecule, the 51 use of a dosage form was required for its in vivo evaluation. Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx xxx nanomedjournal.com The authors state no competing interests. This work was supported by the ANR Blanc Program Mecaferrol. Corresponding author at: Inserm U1066, IBS-CHU Angers, 4 rue Larrey, 49933 Angers Cedex 9. E-mail address: [email protected] (C. Passirani). http://dx.doi.org/10.1016/j.nano.2014.05.002 1549-9634/© 2014 Published by Elsevier Inc. Please cite this article as: Laine A.-L., et al., Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules. Nanomedicine: NBM 2014;xx:1-11, http://dx.doi.org/10.1016/j.nano.2014.05.002 NANO-00941; No of Pages 11
12

Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

May 13, 2023

Download

Documents

Beatrice Caseau
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

1

2

3Q1Q2

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

Q3

23

24

25Q4

26

27

28

29

30

31

32

33

34Q5

Nanomedicine: Nanotechnology, Biology, and Medicinexx (2014) xxx–xxx

nanomedjournal.com

NANO-00941; No of Pages 11

RO

OF

Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drugloaded into stealth lipid nanocapsules

Anne-Laure Lainea,b, Anne Clavreulb,c, Audrey Rousseaud, Clément Tétaudb,c,Anne Vessierese, Emmanuel Garciona,b, Gerard Jaouene, Léo Aubert f, Matthieu Guilbert f,

Jean-Pierre Benoita,b, Robert-Alain Toillonf, Catherine Passirania,b,⁎aLUNAM Université – Micro et Nanomédecines Biomimétiques, Angers, France

bInserm U1066, IBS-CHU, Angers, FrancecDépartement de Neurochirurgie – CHU, Angers, France

dDépartement de Pathologie Cellulaire et Tissulaire – CHU, Angers, FranceeCNRS, UMR 7576, ENSCP, Paris, France

fInserm U908, Université Lille 1, Villeneuve d'Ascq, France

Received 12 September 2013; accepted 3 May 2014

CTED P

Abstract

In this work, a novel ferrocenyl complex (ansa-FcdiOH) was assessed for brain tumor therapy through stealth lipid nanocapsules (LNCs).Stealth LNCs, prepared according to a one-step process, showed rapid uptake by cancer cells and extended blood circulation time. Theferrocenyl complex was successfully encapsulated into these LNCs measuring 40 nm with a high loading capacity (6.4%). In vitro studiesshowed a potent anticancer effect of ansa-FcdiOH on 9L cells with a low IC50 value (0.1 μM) associated with an oxidative stress and a dose-dependent alteration of the cell cycle. Repeated intravenous injections of stealth ansa-FcdiOH LNCs in ectopic glioma bearing rats induced asignificant tumor growth inhibition, supported by a reduced number of proliferative cells in tumors compared to control group. Additionally,no liver damage manifestation was observed in treated animals. These results indicated that stealth ansa-FcdiOH LNCs might be consideredas a potential new approach for cancer chemotherapy.

E© 2014 Published by Elsevier Inc.

RKey words: ansa-FcdiOH; Nanomedicine; Cell cycle; EPR effect

35 Q6

36

37

38

39

40

41

42

43

44

45

UNCO

RIntroduction

Malignant brain tumor remains one of the main medicalchallenges for scientists as no life-saving treatment has beenfound to date despite aggressive therapy modalities (Stuppprotocol).1 Consequently, alternative strategies are needed totackle this dismal tumor.

Over the past few years, Pr. Jaouen and his team havestruggled to develop potent bioorganometalic complexes foranticancer application. Their chemistry is based on the couplingbetween a ferrocene moiety and a tamoxifen-like skeleton, and

46

47

48

49

50

51

The authors state no competing interests.This work was supported by the ANR Blanc Program Mecaferrol.⁎Corresponding author at: Inserm U1066, IBS-CHU Angers, 4 rue

Larrey, 49933 Angers Cedex 9.E-mail address: [email protected] (C. Passirani).

http://dx.doi.org/10.1016/j.nano.2014.05.0021549-9634/© 2014 Published by Elsevier Inc.

Please cite this article as: Laine A.-L., et al., Inhibition of ectopic glioma tumorNanomedicine: NBM 2014;xx:1-11, http://dx.doi.org/10.1016/j.nano.2014.05.0

the resulting complex belongs to the ferrocifen family.2,3 Severalmolecules from their collection have already showed strongantiproliferative activities on experimental glioma and breastcancer models.4–7 Recently, a new ferrocenyl complex, ansa-FcdiOH (Figure 1), has emerged and turned out to be the mosteffective agent according to in vitro antiproliferative assays.Interestingly, ansa-FcdiOH was tested on 60 cancer cell lines ofthe National Cancer Institute (NCI) and showed to be stronglyactive on CNS (central nervous system) cancer cells. In addition,its cytotoxic behavior did not match with any of the 171 referencemolecules from the NCI database (COMPARE analysis).2 So far,its mechanism of action is not fully understood although the roleplayed by its redox properties has been already underlined.8

Therefore, ansa-FcdiOH was selected in this work to further studyits anticancer activity in vitro and in vivo on 9L brain tumors.

Due to the hydrophobic characteristic of this molecule, theuse of a dosage form was required for its in vivo evaluation.

growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules.02

Page 2: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

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

2 A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

EC

Currently, nanocarriers represent the most promising and mostinvestigated delivery systems for various unmet medicalneeds9,10 including cancer therapy.11 The discovery of theenhanced permeability and retention (EPR) phenomenon hasprompted galenists to develop long circulating nanocarriers (alsocalled stealth nanocarriers) to promote the EPR effect andultimately to achieve an enhanced drug accumulation intotumors.12 A widely used technique to obtain such nanocarriersconsists in coating nanoparticles with a polyethylene glycol(PEG) based molecule.13,14

Our laboratory have developed and patented a nanodeliverysystem based on a phase inversion temperature method andcalled lipid nanocapsules (LNCs).15 Standard PEGylation ofLNCs is achieved via the post-insertion (PI) technique16 ofDSPE-mPEG 2000 (1,2-Distearoyl-sn-glycero-3-phosphoetha-nolamine-N-[methoxy-(polyethyleneglycol)-2000]) involving aprior LNC formulation step and a subsequent incubation step asperformed on liposomes.17 The main drawback with this methodconcerns the additional 2 to 4 h incubation step extending thecomplete formulation process time. An alternative PEGylationmethod was approached by Hoarau et al consisting in incorporat-ing the DSPE-PEG in the initial LNC mixture,18 but had not beensubjected to further evaluation so far. Because this latter techniqueprovides a simple while efficient PEGylation process, we designedfor this study “one-step” (OS) stealth drug-loaded LNCs. In anattempt to determine their pharmacological behavior, 9L celluptake, blood circulation profile and accelerated blood clearance(ABC) were assessed through fluorescent stealth OS LNCs.

The anticancer activity of ansa-FcdiOH was investigatedthereafter on 9L rat glioma model, firstly in vitro with cell cycleanalyses and, secondly, in vivo through stealth ansa-FcdiOH-LNC injections in ectopic glioma bearing rats. To furtherdecipher the ferrocenyl drug mechanism of action, cancer cellproliferation and intratumoral immune reaction were evaluatedon tumor sections.

R 137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

UNCO

RMethod

Stealth ansa-FcdiOH-LNC formulation

Stealth LNCswere prepared according to the “one-step process”based on a phase inversion temperature method15,18 Briefly, thepreparation process consisted in mixing all the excipients [SolutolHS15 (15.9% w/w), Lipoid (1.4% w/w), Labrafac (19.4% w/w),NaCl (3.0% w/w), water (55.8% w/w), the drug as powder(2.6% w/w) and DSPE-mPEG 2000 (1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy-(polyethyleneglycol)-2000],1.9% w/w)] under magnetic stirring. Three cycles of progressiveheating and cooling between 90 °C and 50 °C were thenperformed. At the last cooling step, the ansa-FcdiOH LNCsuspension was diluted with water (71.4% w/w). The resultingsuspension was passed through a 0.2 μM filter to remove thenon-entrapped drug. The final ansa-FcdiOH concentration was8 mg/mL (19 mM).

Fluorescent stealth LNCs were obtained by labeling the LNCswith the fluorescent DiI (1,10-dioctadecyl-3,3,30,30-tetramethylindocarbocyanine perchlorate) probe at 4 mmol/LLabrafac or with

TED P

RO

OF

DiD (1,1'-Dioctadecyl-3,3,3',3'-tetramethylindodicarbocyanineperchlorate) at 2.5 mmol/LLabrafac.

LNC characterization

The determination of the drug loading was achieved afterfiltering the LNC step through a 0.2 μMfilter and dissolving 30 μLof LNCs into 5 mL of methanol (Fisher chemical). As the drugsolubility in water is very low, it was assumed that the drug fractionunentrapped during the LNC formulation is in a precipitated state.Then, the filtering step allowed the free drug to be separated fromthe drug loaded into LNC. The drug loading in the filtered LNCsuspension is subsequently measured by spectrophotometry (UV2600, Schimadzu, Champs surMarne, France) against a calibrationcurve generated from the drug solubilized in methanol.

The average hydrodynamic diameter and the zeta potential ofnanocapsules were determined at 25 °C, in triplicate, using aMalvern Zetasizer (Nano Series DTS 1060, Malvern InstrumentsS.A., Worcestershire, UK). For the measurement, the LNCs werediluted in MilliQ water (50 μL LNC in 2.95 mL water).

Osmolarity measurement was performed in triplicate on aVapro Osmometer (ELITECH group, Signes, France).

Tumor cell line and culture

Rat 9L gliosarcoma cells were obtained from the EuropeanCollection of Cell Culture (Sigma, Saint-Quentin Fallavier,France). The cells were cultured at 37 °C in a humidifiedatmosphere containing 5% CO2 in Eagle's minimal essentialmedium (EMEM) (Lonza, Verviers, Belgium) supplied with 1%non essential amino acids (Lonza), 10% fetal calf serum (FCS)(Lonza) and 1% antibiotic and antimycotic solution (Sigma).

In vitro cell uptake

9L cells were seeded in 6-well plates for 72 h. Cells were thentreated with DiD-LNCs, OS stealth DiD-LNCs and PI stealth DID,diluted at 1/500 in a serum-free culturemedium, for 2 h at 37 °C or4 °C. At the end of the incubation period, cells were fixed in 2%formaldehyde and analyzed using a FACScan flow cytometer withCellQuest Software (BD Biosciences). All experiments wereperformed in triplicate and presented as mean ± SD.

In vitro cell viability

A suspension of 9L cells (1.9 × 104 cells/mL) was put in eachwell of 24-well plates for 48 h. On day 2, the culture medium wasremoved and cells were treated with increasing concentrations(0.01-10 μmol/L) of various formulations. After 72 h of incuba-tion at 37 °C, the medium containing samples was replaced byfresh medium. Cell survival percentage was estimated by theMTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) survival assay according to thesupplier's instructions (Promega, Lyon, France).

In vitro reactive oxygen species (ROS) generation

In order to assess the ROS involvement in the ansa-FcdiOHmechanism of action, cells were co-treated with ansa-FcdiOH atthe IC50 concentration and an antioxidant agent (ascorbic acid(Sigma) at 200 μM). The test was performed through the MTS

Page 3: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

T157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

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

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229230231

232

233

234

235

236

237

238

239

240

241

242

243

244

Figure 1. The ansa-FcdiOH molecule.

3A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

UNCO

RREC

assay in the same conditions (seeding, treatment schedule,measurement) as described above. As controls, cells were treatedwith ascorbic acid alone. This experiment was performed intriplicate and presented as mean ± SD.

Cell cycle analysis

Cell cycle analysis was performed as previously described.4

Briefly, cells were treated with free ansa-FcdiOH at increasingconcentration (0.05-0.5 μM) for 24, 48 and 72 h of treatment. Atthe end of the experiment, cells were trypsinized, washed twicewith PBS and fixed (ice cold EtOH 70% v/v, 1 h, −20 °C). Cellswere subsequently incubated in PBS containing RNAse A(50 μg/mL, 15 min, 20 °C) and then in PBS containingpropidium iodide (20 μg/mL, 4 h, 20 °C, in the dark). Cellcycle was analyzed with Cyan LX9 cytometer (BeckmanCoulter, France) and data were processed by a multicyclesoftware (Phoenix Flow Systems, San Diego, USA).

Determination of apoptotic cells

One hundred thousand cells were seeded in 35-mm dishes andtreated for 24 h with ansa-FcdiOH. Cells were then fixed in coldmethanol (−20 °C, 10 min) and stained with 1 μg/mL Hoechst33258 (30 min, 20 °C, in the dark). Apoptotic cells exhibitingcondensed and fragmented nuclei were counted under a NikonEclipse fluorescence microscope. At least, 200 cells in randomlyselected fields were examined.

Animals

In vivo efficacy experiments were carried out on 10 to11-week-old Syngeneic Fischer F344 female rats (Charles RiverLaboratories France, L'Arbresle, France), weighing 160-180 g.Animal care was performed in strict accordance with FrenchMinistry of Agriculture regulations. The animal procedures wereapproved by the Animal Experimentation Ethic Committee ofPays de la Loire, protocol number CEEA.2012.32.

For accelerated blood clearance experiments, 10 to 11-week-old female Sprague rats (SCAHU, Service Commund'Animalerie Hospitalo-Universitaire, Angers, France) wereused weighing 260-300 g.

ED P

RO

OF

ABC phenomenon evaluation

Female Sprague rats were randomly divided into 2 groups.The ABC phenomenon was investigated after a second injectionof LNCs. The initial injection used stealth LNCs andconventional LNCs for comparison and was administered viathe tail vein (400 μL/rats). For the second injection, stealth DiI-labeled LNCs and DiI-LNCs were injected intravenously(400 μL/rats) at day 7.

Blood samples were collected by cardiac puncture atdesignated time intervals (5 min, 30 min, 1 h, 2 h, 4 h and6 h, 8 rats per group) in a blood collection heparin tube(Microtube, 1.3 mL, Sarstedt AG & Co., Nümbrecht, Germany).DiI fluorescence contained in plasma was measured at theemission wavelength of 544 nm with an excitation wavelengthof 590 nm by a Fluoroscan (Ascent FL, Thermo FisherScientific, Cergy-Pontoise, France). One hundred percent offluorescence was considered as the value counted at 5 min post-injection. The blood circulation profiles after the secondinjection were compared to the original profiles obtained througha distinct experiment with one single injection.

In vivo antitumor efficacy

In vivo anticancer activity was evaluated against 9L tumor-bearing fisher rats. Animals were manipulated under isoflur-ane–oxygen anaesthesia. After shaving and disinfection, ratswere intradermically implanted with 1.5 × 106 9L cells on theright flank. At day 7 post tumor implantation, animals werehomogeneously divided into three groups: physiologicalsaline solution (0.9% NaCl), unloaded stealth LNCs andstealth ansa-FcdiOH-LNCs (ansa-FcdiOH dose: 20 mg/kg),with 9 rats per group. The treatments, administered via thelateral tail vein, were started at day 7 and had beenadministered daily for 2 weeks with a mid-term break of2 days (10 injections in total). Tumour growth was followedby regularly measuring the length and width of tumours with acalliper. The tumour volume (V) was estimated by themathematical ellipsoid formula:

V ¼ π=6ð Þ � widthð Þ2 � lengthð Þ

At day 21 post-tumor inoculation, rats were sacrificed in aCO2 chamber, and tumors and livers were excised andimmediately fixed in 4% formaldehyde solution to be embeddedin paraffin.

Tumor and liver slide histology

The embedded tissues were sectioned (4 μM) along thelongitudinal axis of the tumor. The liver and tumor sections werestained with hematoxylin and eosin (H&E) for histologicalanalysis and were observed by an anatomopathologist.

Tumor immunostaining

Tumor tissues were subjected to immunohistochemicalstaining against OX6 and CD3 to analyze macrophage and Tlymphocyte infiltration, respectively. Cell proliferation was alsoassessed through PCNA immunofluorescence. Briefly, tumor

Page 4: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

ECTED P

RO

OF

245

246

247

248

249

250

251

252

253

254

255

256

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

Figure 2. PEG coating density influence on 9L cell uptake. The percentage of positive cells and the Geo mean are indicated on the figure. DiD-LNCs: no PEGcoating, stealth DiD-LNCs (OS): low PEG coating, stealth DiD-LNCs (PI): high PEG coating.

Table 1t1:1

Physico-chemical characteristics of unloaded stealth LNCs and stealth ansa-FcdiOH-LNCs.t1:2

t1:3 Hydrodynamic diameter (nm) PdI Zeta potential (mV) Osmolarity (mOsm) Drug payload (% w/w)

t1:4 Stealth LNCs 60.3 ± 0.8 0.08 −18.8 ± 0.7 327 ± 4 –t1:5 Stealth ansa-FcdiOH-LNCs 40.1 ± 0.1 0.03 −14.1 ± 1.1 370 ± 3 6.4

4 A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

UNCO

RR

slides were deparaffinized and subsequently subjected to antigenretrieval using a citrate buffer pH 6 for 40 min at 90 °C. Inorder to block nonspecific binding, sections were incubated inPBS containing 4% BSA and 10% normal goat serum.Incubations with primary antibodies (anti-rat OX6 (1/50),anti-rat CD3 (1/50) and anti-rat PCNA (1/1000)) wereperformed overnight at 4 °C. Anti-OX6 and anti-CD3 primaryantibodies were detected using a biotinylated secondaryantibody followed by the avidin–biotin complex methodaccording to the supplier's directions (Vector). Sections wererevealed with 3,3-diaminobenzidine (Sigma) counterstainedwith hematoxylin and permanently mounted. Anti-PCNAprimary antibody was detected using a biotinylated secondaryantibody amplified with streptavidin–FITC (Dako). Nucleiwere counterstained with DAPI (Sigma). Sections of three ratsof each group (control and stealth ansa-FcdiOH-LNCs) wereanalyzed under a microscope (Axioscope 2 optical, Zein, LePecq, Germany). PCNA+ cell counting was performed in threetumor sections per rat using the Metaview computerized image-analysis system (Roper scientific, Evry, France). Six fields persections taken at ×200 magnification were randomly chosen in

the tumor. Results were expressed as the mean number ofPCNA+ cells/mm2 for each group ± SEM.

Statistical analysis

For in vitro experiments, the results were expressed as themean ± SD and Student t test was performed between treatedand control groups.

Statistical significance between control and treated groups forin vivo experiments was evaluated using Mann and Withney testand was considered as significant with P b 0.05. Results wereexpressed as a mean ± SEM.

Results

Formulation and characterization of the stealthansa-FcdiOH-loaded LNCs

Stealth ansa-FcdiOH loaded LNCs were successfully pre-pared at 8 mg/mL (6.4% w/w) (Table 1). The direct incorpora-tion of DSPE-PEG into the LNC mixture slightly increased thecarrier size in comparison to conventional LNCs (50 nm) in

Page 5: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

TED P

RO

OF

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

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

Figure 3. Antiproliferative activity of ansa-FcdiOH. (A) 9L cell survival after 72 h exposure to various treatments. (B) Co-treatment with ascorbic acid (AA).*P b 0.05, **P b 0.01.

5A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

UNCO

RRECconsistence with previous studies.18 On the contrary, the drug

encapsulation markedly decreased the diameter compared tostealth LNCs, reaching a size of 40 nm. The zeta potentialmeasurement revealed similar negatively charged surfaces forboth stealth LNCs, indicative of the DSPE-PEG sheathing. Asthe formulations were intended for intravenous delivery, theosmolarity was settled at approximately 300 mOsm.

In vitro cell uptake

The glioma cell uptake of stealth DiD-labeled LNCs obtainedvia the OS process were measured and compared to uncoatedLNCs at an early time point (2 h) and at 4 °C and 37 °C(Figure 2). Additionally, in order to further assess the PEGcorona influence on the cell internalization, LNCs with higherDSPE-PEG coating density were analyzed as a control with thetwo other formulations. These LNCs were prepared through thePI technique, as it allowed a higher DSPE-PEG coating than withthe OS one, as previously described.16 Accordingly, the DSPE-PEG content for PI DiD-LNCs (9% w/w) was almost twice asmuch as that of OS DiD-LNCs (4.8% w/w).

High DSPE-PEG coating density hampered the LNC celluptake (Geo mean = 33.5) in comparison to low PEG density,whose uptake was similar to conventional LNCs (Geo mean =66.5 and 63.2, respectively). Four degrees celsius condition,which usually permits to turn off cell metabolic activitiesincluding active cell internalization, allowed the assessment of

passive uptake. For each LNC batch, a weaker internalizationwas observed at 4 °C in comparison to 37 °C, revealing a majorcontribution from the active uptake. In addition, DSPE-PEGcoating tended to inhibit the passive LNC internalization with thestrongest effect observed for PI LNCs.

In vitro antiproliferative assay

After 72 h cell exposure, ansa-FcdiOH was able to reach anIC50 value of 0.1 μM (Figure 3, A). Similar profiles and IC50values were obtained for free and encapsulated drug whichproved that ansa-FcdiOH was still pharmacologically activedespite its entrapment in LNCs. Moreover, the co-treatment withan antioxidant agent partially inhibited the activity of ansa-FcdiOH (Figure 3, B).

Cell cycle analysis

Two major dose-dependent effects on the cell cycledistribution could be distinguished (Figure 4). At low dose(50 nM), ansa-FcdiOH treatment induced an increase of 9L cellsin S phase fraction (Figure 4, A). This effect was detectable from24 h treatment, subsequently leading to a prolonged S phaseblockade up to 72 h with a slight increase of sub G0/G1population at 0.1 μM. For higher concentrations (0.5 μM), cellswere rapidly stacked in G0/G1 phase and cell cycles were nolonger analyzed because of a massive increase of sub G0/G1population at 48 and 72 h (Figure 4, B). Such sub G0/G1 cell

Page 6: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

UNCO

RRECTED P

RO

OF

Figure 4. Cell cycle distribution and apoptosis counting. (A) Concentration dependent effects. (B) Time dependent effects. (C) Percentage of Hoechst-stainedapoptotic cells after 24 h exposure to increasing ansa-FcdiOH dose.

6 A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

Page 7: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

TED P

RO

OF

332

333

334

335

336

337

338

339

340

341

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

370

371

372

373

374

375

376

377

378

379

380

Figure 5. In vivo study. (A) ABC phenomenon evaluation (1st inj.: first injection, 2nd inj.: second injection). (B) In vivo antitumor efficacy. Treatment days areindicated by red arrows. The pictures represent an overview of the tumor mass at D14 for each group of animal. n = number of animals per group. *P b 0.05.

7A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

UNCO

RREC

population revealed a degradation of DNA within the treatedcells which could occur by necrosis or apoptosis. Therefore,Hoechst staining determination of apoptosis was performed after24 h treatment. As shown in Figure 4, C, apoptotic nucleiincreased in a dose dependent manner under ansa-FcdiOHtreatment indicating that sub G0/G1 fraction observed by flowcytometry was due to apoptosis induction.

Accelerated blood clearance (ABC) phenomenon investigation

ABC phenomenon was investigated at the second stealth DiILNC injection administered 7 days after pretreatment withunloaded stealth LNCs. The blood circulation profiles of thestealth LNCs are displayed in Figure 5, A, and compared toconventional LNCs (without DSPE-mPEG coating) as shortcirculating LNC control. A slight decrease in stealth LNC bloodcirculation time was observed at the second injection. However,the whole plasmatic profile remained extended compared touncoated LNCs.

In vivo tumor growth inhibition study

Interestingly, stealth ansa-FcdiOH-LNC treatment sloweddown the tumor growth from day 11 compared to the two controlgroups (Figure 5, B). From day 14, the tumor volume curveplateaued meaning that ansa-FcdiOH inhibited the tumor growth.

To determine whether ansa-FcdiOH affects 9L tumor growthat the level of cell proliferation, a PCNA immunofluorescentanalysis was performed (Figure 6, A). The number of positivelyresponding cells was estimated at 1553 ± 105 PCNA+ cells/mm2

for the drug treated group, which was significantly lowercomparatively to that of the saline group with 2025 ± 123PCNA+ cells/mm2.

Anti-tumor immune response analysis

In an attempt to evaluate whether the tumor growth inhibitioncould stem from an intratumoral immune response, macrophageand T lymphocyte infiltrations were immunohistologicallystained (Figure 6, B). Both tumor sections of stealth ansa-FcdiOH-LNCs and saline groups displayed similar macrophageand lymphocyte infiltration.

Liver histology

Considering the aggressive protocol involving repeatedinjections of stealth ansa-FcdiOH LNCs, it seems necessary totake an interest in the rat livers. Specimen sections of the controlgroups and ansa-FcdiOH treated groups (Figure 6, B) showednormal histological appearance of the organ meaning that nodamage manifestation was detected within ansa-FcdiOH groups.

Discussion

Despite intensive research efforts, the diagnosis of amalignant brain cancer remains associated with a grim prognosis.Current available drug therapies are insufficiently effective totreat this aggressive disease, mainly attributed to tumor-cellresistance, and ultimately leading to ineluctable recurrence.

Page 8: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

UNCO

RRECTED P

RO

OF

Figure 6. Immunohistological analyses. (A) PCNA-stained proliferating cells in tissue sections. Green: PCNA+ cells, blue: nuclei stained by DAPI. **P b 0.01(scale bar = 100 μM). (B) Effect of stealth ansa-FcdiOH-LNCs treatment on liver (b) and on tumor (d, f) compared to saline treatment (a and c, e respectively)(scale bar = 50 μM).

8 A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

Page 9: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

T

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

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

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

495

496

9A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

UNCO

RREC

Consequently, innovative treatments are urgently needed toimprove the outcome for patients.

The drug of interest in this work is a ferrocenyl molecule(ansa-FcdiOH) which can be described as a dyphenol metallo-complex. Polyphenol are currently under considerable investi-gation due to their cytotoxic effect on cancer cells. Among them,curcumin is subject to great interest in particular in glioblastomatreatment and has already shown promising outcome.19,20

Besides a potential polyphenolic activity, ansa-FcdiOH canalso take advantage of its metallic part which may confer aunique and alternative activity over organic compound.

Considering the poor water soluble property of this ferrocenylcomplex, a drug delivery system was required for furtherevaluation. As LNCs have already proved to be a suitable tumor-delivery nanosystem for other ferrocenyl compounds,4,5 weobviously selected this carrier for the development of injectableansa-FcdiOH dosage form. As above-mentioned, the in vivodrug delivery performance of nanoparticles is commonly boostedthrough the addition of PEG onto their surface.

While conventional LNC PEGylation is achieved through apost-insertion technique, we designed for this study alternativestealth ansa-FcdiOH-LNCs via a one-step process intended tofacilitate a future transposition in clinic. In a study conducted inparallel, the influence of the quantity of PEGylated phospho-lipids (DSPE-mPEG) added into the LNC formulation wasassessed.21 It has been shown that the formation of PEGylatedLNCs through a unique step was possible up to a DSPE-PEGconcentration of 6% w/w resulting in a gradual increase in LNCdiameter. In the present study, the DSPE-mPEG concentrationwas set at 4.8% (w/w), which was shown to provide longcirculation property while maintaining a satisfactory size. Thedrug loading capacity of these OS-LNCs was therefore evaluatedthrough the encapsulation of ansa-FcdiOH, achieving samemaximal payload as that of the uncoated ones (data not shown).Accordingly, stealth ansa-FcdiOH-LNCs were successfullyprepared by the one-step phase inversion temperature methodwith a satisfactory size of 40 nm, high drug loading and stealthfeatures, making them suitable for intravenous delivery.

It could be noticed that drug loaded stealth LNCs showedsmaller hydrodynamic size than unloaded stealth LNCs. Thisphenomenon may be induced by the ansa-FcdiOH entrapmentinto the LNC shell structure as previously reported with theencapsulation of another polyphenol molecule, the quercetin.22

As a matter of fact, polyphenols are able to interact withmembrane bilayers and can bind to the lipid–water interface.23

Based on these considerations, the size alteration is assumed toresult from the drug arrangement between the HS-PEG chains atthe LNC interface.

Proper PEGylated phospholipid anchorage onto the LNCsurface was evidenced by the high negative surface chargestemming from the PEG dipoles4 as well as from the negativelycharged phosphate groups of DSPE-mPEG. Additionally,the stealth ansa-FcdiOH-LNCs displayed a narrow sizedistribution (PdI = 0.05), which is a critical parameter controllingthe biodistribution.

As nanocarrier stability is an important factor to consider, astability study at 37 °C was performed on stealth ansa-FcdiOH-LNCs in comparison to uncoated ansa-FcdiOH LNCs. Surpris-

ED P

RO

OF

ingly, stealth ansa-FcdiOH-LNCs exhibited enhanced physicalstability over regular ansa-FcdiOH-LNCs. The latter displayedindeed a dramatic increase in size probably originating from theinterface destabilization by the drug (data not shown). This resultemphasized the capacity of PEGylation to ensure the physicalLNC stability. Additionally, no precipitation phenomenon wasobserved either visually or via particle size measurement duringthe stability assay, meaning that no burst release of the drugseemed to occur. Furthermore, we recently assessed the releasekinetics from the LNCs through FRET (Förster resonance energytransfer) monitoring. This fluorescence technique based on theco-loading of a donor/acceptor probe pair permitted to easilymonitor the carrier stability of encapsulation. Although themolecules entrapped were fluorescent probes, stealth LNCsshowed great stability of encapsulation. More interestingly, longPEG chain grafting seemed to prevent from drug leakingobserved with uncoated LNCs.21

It was previously demonstrated that conventional LNCs wererapidly internalized by glioma cells through endocyticpathway.24 However, the influence of DSPE-PEG coating onLNC uptake had not been evaluated so far. Although PEGcoating create a hydrophilic corona able to prevent LNCs fromthe opsonization phenomenon, it might also hinder nanoparticleinternalization by cells. Accordingly, we assessed the LNCuptake by 9L glioma cells depending on the level of PEG coatingdensity. Highly PEG coated-LNCs obtained through the PItechnique showed reduced cell uptake compared to LNCs with alower OS PEG coating. Interestingly, the later displayed similarinternalization to uncoated LNCs. This study highlighted the factthat hydrophilic PEG corona could reduce cell interactions,likely owing to steric repulsions and lack of affinity. Theseobservations further supported our choice of a simple OS processfor the design of stealth LNCs, achieving a low while adequatePEG coating over the PI technique. Additionally, these OSstealth LNCs exhibited the capacity to deliver their loading at theintracellular level mostly through active uptake.

In vitro antiproliferative assay demonstrated strong ansa-FcdiOH effect against 9L glioma cells achieving an IC50 of0.1 μM and the encapsulation in stealth LNCs did not alter itspotency. In comparison to FcdiOH, an analogous drug to ansa-FcdiOH which has already shown promising activity on braintumor,5 ansa-FcdiOH displayed a lower IC50 value on 9L cellline demonstrating its hopeful potent activity. Remarkably, ansa-FcdiOH showed also a lower IC50 value against glioma cell linesin comparison to curcumin with 25 μM19 or to gold compoundsreaching 3 μM.25 The partial inhibition of the drug activity whenassociated with an antioxidant supported a ROS-mediated celldeath. This is commonly encountered with metallodrugs such asruthenium compounds26 and gold-based compounds25 andpolyphenol drugs.27,28

In order to get a deeper insight into the ansa-FcdiOHmechanismof action, cell cycle analyses were performed. As a result, ansa-FcdiOH altered the 9L cell cycle in a dose dependent manner withthe arrest in S-phase at low dose (50 nM) and in G0/G1 phase athigher dose associated with apoptosis induction. In a previouswork, the impact of LNCs in the drug effect on cell cycleprogression was investigated. Cell cycle analyses were conductedafter incubation of MDA-MB-231 breast cancer cells with

Page 10: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560Q7

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

10 A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

UNCO

RREC

FcOHTam, a compound belonging to the same ferrocenyl family asansa-FcdiOH.4 Interestingly, it has been shown that the drugdisplayed similar activity on the cell cycle regardless of theformulation (free drug, drug loaded-LNCs, drug loaded stealthLNCs). Additionally, the unloaded vehicles did not affect thecell cycle.

Based on clinical settings, we planned for the in vivo efficacystudy a treatment protocol composed of multiple doses. Repeatedinjections indeed aimed to increase the drug concentration intumor tissue as demonstrated in the literature with multipleinjections of chitosan-based nanoparticles.29 However, it hasbeen reported that multiple injections of PEGylated nanocarriersinduced an immune reaction with M immunoglobulin productionagainst PEG which, as consequence, led to an accelerated bloodclearance phenomenon as soon as the second injection ofPEGylated nanocarriers was given.30 Additionally, the seconddose was shown to preferentially end up in Kupffer cells of theliver.31 This consideration is a matter of concern, as, besidesaffecting the passive targeting ability of the cargo, it also cancause severe liver damage in the case of toxic anticancer drugs.Consequently, ABC phenomenon was investigated at the secondstealth DiI LNC injection administered 7 days after pretreatmentwith unloaded stealth LNCs. As reported in the literature, this7 day interval corresponds to the maximum magnitude of ABCeffect.30 Comfortingly, no dramatic decrease in LNC bloodcirculation time was observed in comparison to PEG liposomeswhich lose their entire stealth properties at the second dose.30

Additionally, the extended circulation time for stealth LNCs overconventional LNCs was confirmed. This ABC effect seems to behighly dependent on the type of nanocarrier used and, to date, noparameter can help in predicting the potential induction ofantibodies upon injections of PEGylated nanosystems.

In vivo antitumor studies showed that repeated injection ofstealth ansa-FcdiOH-LNCs could significantly inhibit 9L tumorgrowth compared to controls. This observed anticancer activitywas further evidenced by a significant reduced number ofintratumoral proliferating cells upon ansa-FcdiOH treatment.This could be due to the cell blockade in G0/G1 phase but also inS-phase since prolonged S-phase arrest is associated withdecrease of S phase related cell cycle protein like PCNA.32,33

It must be pointed out that 9L tumor has been shown to behighly immunogenic34 which might mislead about the drug-induced tumor treatment. However, macrophage and T lympho-cyte infiltration staining suggested that ansa-FcdiOH activity didnot seem to be related to a distinct activation of an intratumoralimmune response.

Hepatotoxicity is one of the major issues encountered withpatients undergoing chemotherapy.35 Interestingly, the repeatedinjections of stealth ansa-FcdiOH-LNCs did not induce apparentliver damage. This is intriguing since metal-based drugsespecially cisplatin36 often induced hepatic adverse effects.This non-toxicity to liver may also be ascribed to the drugconfinement into the LNCs protecting healthy tissue which is oneof the crucial advantages of nanocarriers permitting to overcomedose-limiting toxicity encountered with several drugs.37

Taken all together, these results have proved that stealth ansa-FcdiOH-LNCs may represent an alternative approach to treatcancer and may comply with the current need in oncology.

Moreover, nanoparticles seem to be a promising tool capable ofovercoming many of the major obstacles encountered in braintumor treatment.38 Accordingly, further studies have beenplanned in order to assess the performance of these stealthansa-FcdiOH-LNCs in an orthotopic glioma model.

Acknowledgments

The authors would like to thank Pascal Pigeon (ENSCP) foransa-FcdiOH synthesis.

OFAppendix A. Supplementary data

Supplementary data to this article can be found online athttp://dx.doi.org/10.1016/j.nano.2014.05.002.

TED P

RO

References

1. Stupp R, Hegi ME, MasonWP, van den Bent MJ, Taphoorn MJB, JanzerRC, et al. Effects of radiotherapy with concomitant and adjuvanttemozolomide versus radiotherapy alone on survival in glioblastoma in arandomised phase III study: 5-year analysis of the EORTC-NCIC trial.Lancet Oncol 2009;10:459-66.

2. Vessières A. Metal carbonyl tracers and the ferrocifen family: Two facetsof bioorganometallic chemistry. J Organomet Chem 2013;734:3-16.

3. Görmen M, Pigeon P, Top S, Hillard EA, Huché M, Hartinger CG, et al.Synthesis, cytotoxicity, and COMPARE analysis of ferrocene and [3]ferrocenophane tetrasubstituted olefin derivatives against human cancercells. ChemMedChem 2010;5:2039-50.

4. Lainé A-L, Adriaenssens E, Vessières A, Jaouen G, Corbet C, DesruellesE, et al. The in vivo performance of ferrocenyl tamoxifen lipidnanocapsules in xenografted triple negative breast cancer. Biomaterials2013;34:6949-56.

5. Laine A-L, Huynh NT, Clavreul A, Balzeau J, Béjaud J, Vessieres A, et al.Brain tumour targeting strategies via coated ferrociphenol lipid nanocap-sules. Eur J Pharm Biopharm 2012;81:690-3.

6. Huynh NT, Passirani C, Allard-Vannier E, Lemaire L, Roux J, GarcionE, et al. Administration-dependent efficacy of ferrociphenol lipidnanocapsules for the treatment of intracranial 9L rat gliosarcoma. Int JPharm 2012;423:55-62.

7. RogerM,Clavreul A, HuynhNT, Passirani C, Schiller P,Vessières A, et al.Ferrociphenol lipid nanocapsule delivery by mesenchymal stromal cells inbrain tumor therapy. Int J Pharm 2012;423:63-8.

8. Plazuk D, Vessières A, Hillard EA, Buriez O, Labbé E, Pigeon P, et al. A[3]ferrocenophane polyphenol showing a remarkable antiproliferativeactivity on breast and prostate cancer cell lines. J Med Chem2009;52:4964-7.

9. Büyüktimkin B, Wang Q, Kiptoo P, Stewart JM, Berkland C, SiahaanTJ. Vaccine-like controlled-release delivery of an immunomodulatingpeptide to treat experimental autoimmune encephalomyelitis. MolPharm 2012;9:979-85.

10. Gu Z, Aimetti AA, Wang Q, Dang TT, Zhang Y, Veiseh O, et al.Injectable nano-network for glucose-mediated insulin delivery. ACSNano 2013;7:4194-201.

11. Jia F, Liu X, Li L, Mallapragada S, Narasimhan B, Wang Q.Multifunctional nanoparticles for targeted delivery of immune activatingand cancer therapeutic agents. J Control Release 2013;172:1020-34.

12. Prabhakar U, Maeda H, Jain RK, Sevick-Muraca EM, Zamboni W,Farokhzad OC, et al. Challenges and key considerations of the enhancedpermeability and retention effect for nanomedicine drug delivery inoncology. Cancer Res 2013;73:2412-7.

Page 11: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

T

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638Q8639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

685

686

687

688

689

690

691

693

11A.-L. Laine et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2014) xxx–xxx

EC

13. Jokerst JV, Lobovkina T, Zare RN, Gambhir SS. NanoparticlePEGylation for imaging and therapy. Nanomedicine (Lond)2011;6:715-28.

14. Huynh NT, Roger E, Lautram N, Benoît J-P, Passirani C. The rise andrise of stealth nanocarriers for cancer therapy: passive versus activetargeting. Nanomedicine (Lond) 2010;5:1415-33.

15. Huynh NT, Passirani C, Saulnier P, Benoit JP. Lipid nanocapsules: anew platform for nanomedicine. Int J Pharm 2009;379:201-9.

16. Perrier T, Saulnier P, Fouchet F, Lautram N, Benoît J-P. Post-insertioninto lipid nanocapsules (LNCs): From experimental aspects tomechanisms. Int J Pharm 2010;396:204-9.

17. Uster PS, Allen TM, Daniel BE, Mendez CJ, Newman MS, Zhu GZ.Insertion of poly(ethylene glycol) derivatized phospholipid into pre-formed liposomes results in prolonged in vivo circulation time. FEBSLett 1996;386:243-6.

18. Hoarau D, Delmas P, David S, Roux E, Leroux J-C. Novel long-circulating lipid nanocapsules. Pharm Res 2004;21:1783-9.

19. Purkayastha S, Berliner A, Fernando SS, Ranasinghe B, Ray I, Tariq H,et al. Curcumin blocks brain tumor formation. Brain Res 2009, http://dx.doi.org/10.1016/j.brainres.2009.01.066.

20. Zanotto-Filho A, Coradini K, Braganhol E, Schröder R, de Oliveira CM,Simões-Pires A, et al. Curcumin-loaded lipid-core nanocapsules as astrategy to improve pharmacological efficacy of curcumin in gliomatreatment. Eur J Pharm Biopharm 2012, http://dx.doi.org/10.1016/j.ejpb.2012.10.019.

21. Lainé A-L, Gravier J, Henry M, Sancey L, Bejaud J, Pancani, E, et al.Conventional versus stealth lipid nanoparticles: formulation and in vivofate prediction through FRET monitoring. Submitted [date unknown],[no volume].

22. Barras A, Mezzetti A, Richard A, Lazzaroni S, Roux S, Melnyk P, et al.Formulation and characterization of polyphenol-loaded lipid nanocap-sules. Int J Pharm 2009;379:270-7.

23. Oteiza PI, Erlejman AG, Verstraeten SV, Keen CL, Fraga CG.Flavonoid-membrane interactions: a protective role of flavonoids atthe membrane surface? Clin Dev Immunol 2005;12:19-25.

24. Paillard A, Hindré F, Vignes-Colombeix C, Benoit J-P, Garcion E. Theimportance of endo-lysosomal escape with lipid nanocapsules for drugsubcellular bioavailability. Biomaterials 2010;31:7542-54.

25. Illán-Cabeza NA, García-García AR, Martínez-Martos JM, Ramírez-Expósito MJ, Peña-Ruiz T, Moreno-Carretero MN. A potentialantitumor agent, (6-amino-1-methyl-5-nitrosouracilato-N3)-triphenyl-

UNCO

R

692

ED P

RO

OF

phosphine-gold(I): Structural studies and in vivo biological effectsagainst experimental glioma. Eur J Med Chem 2013;64:260-72.

26. Vidimar V, Meng X, Klajner M, Licona C, Fetzer L, Harlepp S, et al.Induction of caspase 8 and reactive oxygen species by ruthenium-derived anticancer compounds with improved water solubility andcytotoxicity. Biochem Pharmacol 2012;84:1428-36.

27. Sharma V, Joseph C, Ghosh S, Agarwal A, Mishra MK, Sen E.Kaempferol induces apoptosis in glioblastoma cells through oxidativestress. Mol Cancer Ther 2007;6:2544-53.

28. Thayyullathil F, Chathoth S, Hago A, Patel M, Galadari S. Rapidreactive oxygen species (ROS) generation induced by curcumin leads tocaspase-dependent and -independent apoptosis in L929 cells. Free RadicBiol Med 2008;45:1403-12.

29. Kim K, Kim JH, Park H, Kim Y-S, Park K, Nam H, et al. Tumor-homingmultifunctional nanoparticles for cancer theragnosis: simultaneousdiagnosis, drug delivery, and therapeutic monitoring. J Control Release2010;146:219-27.

30. Ishida T, Kiwada H. Accelerated blood clearance (ABC) phenomenonupon repeated injection of PEGylated liposomes. Int J Pharm2008;354:56-62.

31. Knop K, Hoogenboom R, Fischer D, Schubert US. Poly(ethylene glycol)in drug delivery: pros and cons as well as potential alternatives. AngewChem Int Ed Engl 2010;49:6288-308.

32. Tinnemans MMFJ, Lenders M-HJH, ten Velde GPM, Blijham GH,Ramaekers FCS, Schutte B. S-phase arrest of nutrient deprived lungcancer cells. Cytometry 1995;19:326-33.

33. Borel F, Lacroix FB, Margolis RL. Prolonged arrest of mammalian cellsat the G1/S boundary results in permanent S phase stasis. J Cell Sci2002;115:2829-38.

34. Barth RF, Kaur B. Rat brain tumor models in experimental neuro-oncology: the C6, 9L, T9, RG2, F98, BT4C, RT-2 and CNS-1 gliomas.J Neurooncol 2009;94:299-312.

35. Floyd J, Mirza I, Sachs B, Perry MC. Hepatotoxicity of chemotherapy.Semin Oncol 2006;33:50-67.

36. Lu Y, Cederbaum AI. Cisplatin-induced hepatotoxicity is enhanced byelevated expression of cytochrome P450 2E1. Toxicol Sci2006;89:515-23.

37. Lainé A-L, Passirani C. Novel metal-based anticancer drugs: a newchallenge in drug delivery. Curr Opin Pharmacol 2012;12:420-6.

38. Li M, Deng H, Peng H, Wang Q. Functional nanoparticles in targetingglioma diagnosis and therapies. J Nanosci Nanotechnol 2014;14:415-32.

R

Page 12: Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules

UNCO

RRECTED P

RO

OF

1 Graphical Abstract

2 Nanomedicine: Nanotechnology, Biology, and Medicine xxx (2014) xxx

4

5 Inhibition of ectopic glioma tumor growth by a potent ferrocenyl6 drug loaded into stealth lipid nanocapsules

7

8 Anne-Laure Laine a,b, Anne Clavreul b,c, Audrey Rousseau d,9 Clément Tétaud b,c, Anne Vessieres e, Emmanuel Garcion a,b,10 Gerard Jaouen e, Léo Aubert f, Matthieu Guilbert f, Jean-Pierre Benoit a,b,11 Robert-Alain Toillon f, Catherine Passirani a,b,⁎

1213

aLUNAM Université – Micro et Nanomédecines Biomimétiques, Angers, France14

bInserm U1066, IBS-CHU, Angers, France15

cDépartement de Neurochirurgie – CHU, Angers, France16

dDépartement de Pathologie Cellulaire et Tissulaire – CHU, Angers, France17

eCNRS, UMR 7576, ENSCP, Paris, France18

fInserm U908, Université Lille 1, Villeneuve d'Ascq, France

1920 Daily intravenous injections of stealth LNCs loaded with a novel ferrocenyl21 compound (ansa-FcdiOH) induced a significant tumor growth inhibition22 compared to controls.

23

24

Nanomedicine: Nanotechnology, Biology, and Medicinexx (2014) xxx

nanomedjournal.com