Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the...

10
UNCORRECTED PROOF 1 Simultaneous active intracellular delivery of doxorubicin and 2 C6-ceramide shifts the additive/antagonistic drug interaction of 3 non-encapsulated combination Q2 Nuno A. Fonseca a,b , Lígia C. Gomes-da-Silva a , Vera Moura a,c , Sérgio Simões a,b , João Nuno Moreira a,b, 5 a CNCCentre for Neurosciences and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal 6 b FFUCFaculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal 7 c TREAT U, SA, Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal abstract 8 article info 9 Article history: 10 Received 26 May 2014 11 Accepted 25 September 2014 12 Available online xxxx 13 Keywords: 14 Combinatorial ratiometric drug design 15 C6-ceramide 16 Nanoparticles 17 Nucleolin 18 F3-targeted delivery 19 Drug resistance remains the Achilles tendon undermining the success of chemotherapy. It has been recognized 20 that success requires the identication of compounds that, when combined, lead to synergistic tumor inhibition 21 while simultaneously minimizing systemic toxicity. However, in vivo application of such protocols is dependent 22 on the ability to deliver the appropriate drug ratio at the tumor level. In this respect, nanotechnology-based de- 23 livery platforms, like liposomes, offer an elegant solution for the in vivo translation of such strategy. 24 In this work, we propose the active intracellular delivery of combinations of doxorubicin and the pro-apoptotic 25 sphingolipid, C6-ceramide, using our previously described cytosolic triggered release-enabling liposomes, 26 targeting nucleolin with the F3 peptide. 27 Combination of doxorubicin (DXR):C6-ceramide (C6-Cer) at 1:2 molar ratio interacted synergistically against 28 drug resistant/triple negative MDA-MB-231 breast cancer cells, as well as drug sensitive MDA-MB-435S melano- 29 ma cells. Cell viability studies indicated that F3-targeted liposomes encapsulating DXR:C6-Cer 1:2 molar ratio (p 30 [F3]DC12) performed similarly as targeted liposomal DXR (p[F3]SL), encapsulating twice the amount of DXR, at 31 the IC 50 , for an incubation time of 24 h. Importantly, F3-targeted liposomes encapsulating DXR:C6-Cer 1:2 molar 32 ratio (p[F3]DC12) enabled a cell death above 90% at 24 h of treatment against both DXR-resistant and sensitive 33 cells, unattainable by the F3-targeted liposomal doxorubicin. Furthermore, a F3-targeted formulation encapsulat- 34 ing a mildly additive/antagonistic DXR:C6-Cer 1:1 molar ratio (p[F3]DC11) enabled an effect above 90% for an in- 35 cubation period as short as 4 h, suggesting that the delivery route at the cell level may shift the nature of drug 36 interaction. Such activity, including the one for p[F3]DC12, induced a marked cell and nucleus swelling at similar 37 extent, consistent with necrotic cell death. 38 Overall, these results demonstrated that F3-targeted intracellular delivery of different DXR/C6-Cer ratios, with 39 diversed drug interactions, enabled a highly relevant increased efcacy against chemotherapy resistant cells. 40 © 2014 Elsevier B.V. All rights reserved. 41 42 43 44 45 1. Introduction 46 As global cancer burden continues to increase, 1.6 million new 47 cancer cases were expected in 2013, from which approximately 48 0.58 million deaths were estimated only in US [1,2]. Chemotherapy 49 still represents the cornerstone treatment of disseminated malignan- 50 cies. Nonetheless, drug resistance remains as the Achilles tendon 51 undermining the success of chemotherapy, mainly owing to molecular 52 and cellular heterogeneity within the tumor microenvironment [3,4]. 53 One of the attempts to solve this problem is the combination of drugs 54 near to their maximum tolerated dose (MTD), often exposing patients 55 to unacceptable toxicity. Alternatively, it has been recognized that suc- 56 cess requires the identication of compounds that, when combined, 57 lead to synergistic tumor inhibition while simultaneously minimizing 58 systemic toxicity [5,6]. 59 One of the most important pathways deregulated in cancer is the 60 PI3K/Akt signaling cascade [7]. In several murine models of cancer, inhi- 61 bition of PI3K/Akt signaling retards tumorigenesis by restoring the apo- 62 ptotic sensitivity of cancer cells to chemotherapeutic agents [8]. In this 63 respect, ceramides are one of the most promising drugs, which have 64 been described to inhibit the PI3K/Akt pathway both in HL-60 leukemia 65 cells [9] and in PC-3 prostate cancer cells [10]. However, the intracellu- 66 lar activity of ceramides is challenged by glucosylceramide synthase 67 (GCS), an enzyme that converts pro-apoptotic ceramides into inactive Journal of Controlled Release xxx (2014) xxxxxx Corresponding author at: Center for Neurosciences and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal. E-mail addresses: [email protected] (N.A. Fonseca), [email protected] (L.C. Gomes-da-Silva), [email protected] (V. Moura), [email protected] (S. Simões), [email protected] (J.N. Moreira). COREL-07389; No of Pages 10 http://dx.doi.org/10.1016/j.jconrel.2014.09.024 0168-3659/© 2014 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Journal of Controlled Release journal homepage: www.elsevier.com/locate/jconrel Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antag- onistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.org/10.1016/j.jconrel.2014.09.024

Transcript of Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the...

Page 1: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

1

2

3

4Q2

567

8

9101112

131415161718

40

4142

43

44

45

46

47

48

49

50

51

52

Journal of Controlled Release xxx (2014) xxx–xxx

COREL-07389; No of Pages 10

Contents lists available at ScienceDirect

Journal of Controlled Release

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

Simultaneous active intracellular delivery of doxorubicin andC6-ceramide shifts the additive/antagonistic drug interaction ofnon-encapsulated combination

OO

F

Nuno A. Fonseca a,b, Lígia C. Gomes-da-Silva a, Vera Moura a,c, Sérgio Simões a,b, João Nuno Moreira a,b,⁎a CNC—Centre for Neurosciences and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugalb FFUC—Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugalc TREAT U, SA, Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal

⁎ Corresponding author at: Center for NeurosciencesCoimbra, Largo Marquês de Pombal, 3004-517 Coimbra, P

E-mail addresses: [email protected] (N.A. F(L.C. Gomes-da-Silva), [email protected] (V. Moura),[email protected] (J.N. Moreira).

http://dx.doi.org/10.1016/j.jconrel.2014.09.0240168-3659/© 2014 Elsevier B.V. All rights reserved.

Please cite this article as: N.A. Fonseca, et al.,onistic drug interaction of non..., J. Control. R

Ra b s t r a c t

a r t i c l e i n f o

19

20

21

22

23

24

25

26

27

28

Article history:Received 26 May 2014Accepted 25 September 2014Available online xxxx

Keywords:Combinatorial ratiometric drug designC6-ceramideNanoparticlesNucleolinF3-targeted delivery

29

30

31

32

33

34

35

36

37

38

39

RRECTED P

Drug resistance remains the Achilles tendon undermining the success of chemotherapy. It has been recognizedthat success requires the identification of compounds that, when combined, lead to synergistic tumor inhibitionwhile simultaneously minimizing systemic toxicity. However, in vivo application of such protocols is dependenton the ability to deliver the appropriate drug ratio at the tumor level. In this respect, nanotechnology-based de-livery platforms, like liposomes, offer an elegant solution for the in vivo translation of such strategy.In this work, we propose the active intracellular delivery of combinations of doxorubicin and the pro-apoptoticsphingolipid, C6-ceramide, using our previously described cytosolic triggered release-enabling liposomes,targeting nucleolin with the F3 peptide.Combination of doxorubicin (DXR):C6-ceramide (C6-Cer) at 1:2 molar ratio interacted synergistically againstdrug resistant/triple negativeMDA-MB-231 breast cancer cells, as well as drug sensitiveMDA-MB-435Smelano-ma cells. Cell viability studies indicated that F3-targeted liposomes encapsulating DXR:C6-Cer 1:2 molar ratio (p[F3]DC12) performed similarly as targeted liposomal DXR (p[F3]SL), encapsulating twice the amount of DXR, atthe IC50, for an incubation time of 24 h. Importantly, F3-targeted liposomes encapsulating DXR:C6-Cer 1:2 molarratio (p[F3]DC12) enabled a cell death above 90% at 24 h of treatment against both DXR-resistant and sensitivecells, unattainable by the F3-targeted liposomal doxorubicin. Furthermore, a F3-targeted formulation encapsulat-ing amildly additive/antagonistic DXR:C6-Cer 1:1molar ratio (p[F3]DC11) enabled an effect above 90% for an in-cubation period as short as 4 h, suggesting that the delivery route at the cell level may shift the nature of druginteraction. Such activity, including the one for p[F3]DC12, induced amarked cell and nucleus swelling at similarextent, consistent with necrotic cell death.Overall, these results demonstrated that F3-targeted intracellular delivery of different DXR/C6-Cer ratios, withdiversed drug interactions, enabled a highly relevant increased efficacy against chemotherapy resistant cells.

© 2014 Elsevier B.V. All rights reserved.

O

53

54

55

56

57

58

59

60

61

UNC1. Introduction

As global cancer burden continues to increase, 1.6 million newcancer cases were expected in 2013, from which approximately0.58 million deaths were estimated only in US [1,2]. Chemotherapystill represents the cornerstone treatment of disseminated malignan-cies. Nonetheless, drug resistance remains as the Achilles tendonundermining the success of chemotherapy, mainly owing to molecularand cellular heterogeneity within the tumor microenvironment [3,4].

62

63

64

65

66

67

and Cell Biology, University ofortugal.onseca), [email protected]@ci.uc.pt (S. Simões),

Simultaneous active intracelluelease (2014), http://dx.doi.o

One of the attempts to solve this problem is the combination of drugsnear to their maximum tolerated dose (MTD), often exposing patientsto unacceptable toxicity. Alternatively, it has been recognized that suc-cess requires the identification of compounds that, when combined,lead to synergistic tumor inhibition while simultaneously minimizingsystemic toxicity [5,6].

One of the most important pathways deregulated in cancer is thePI3K/Akt signaling cascade [7]. In severalmurinemodels of cancer, inhi-bition of PI3K/Akt signaling retards tumorigenesis by restoring the apo-ptotic sensitivity of cancer cells to chemotherapeutic agents [8]. In thisrespect, ceramides are one of the most promising drugs, which havebeen described to inhibit the PI3K/Akt pathway both in HL-60 leukemiacells [9] and in PC-3 prostate cancer cells [10]. However, the intracellu-lar activity of ceramides is challenged by glucosylceramide synthase(GCS), an enzyme that converts pro-apoptotic ceramides into inactive

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 2: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

T

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

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

180

181

182

183

184

185

186

187

188

189

2 N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RREC

glucosylceramides [11–13]. In addition, GCS has been related withdoxorubicin (DXR) resistance, by modulation and depletion ofdoxorubicin-induced ceramide levels [12,14]. This intrinsic relationsupports the addition of exogenous ceramides concomitantly withdoxorubicin, to counteract GCS-mediated pro-apoptotic sphingolipiddepletion, which along with ceramide-mediated PI3K/Akt modulationcould enable increased efficacy at lower chemotherapeutic doses [15].Additionally, it has been demonstrated that ceramides impair angiogen-esis, the formation of new blood vessels from pre-existing ones, an im-portant component in the tumor microenvironment dynamics whichsupports tumor growth [4,16,17]. Therefore, impairing cellular and bio-logical functions of tumors using synergistic combinations of drugs likeceramides and doxorubicin, could translate into an increased gain in ef-ficacy upon acting at different levels of the tumor microenvironment.

However, in vivo application of such protocols is highly dependenton the ability to synchronize the pharmacokinetics of each individualdrug present in the combination, thus enabling the tumor accumulationof a synergistic drug cocktail [18], namely at the intracellular level oftargeted tumor cells. Nanotechnology-based delivery platforms, suchas liposomes, upon the extracellular release of a encapsulated drugcombination, have shown increased efficacy inmurinemodels of cancer[18,19] aswell as in afirst-in-man study against acutemyeloid leukemia[20,21].

Considering the state-of-the-art, the present work aims at selectinga synergistic drug combination between C6-ceramide and doxorubicinand assessing the impact of its intracellular delivery using the F3peptide-targeted pH-sensitive lipid-based nanoparticle, developed byour group [22]. The F3 peptide enables specific recognition of nucleolin,a protein highly expressed in cancer cells and endothelial cells of tumorangiogenic blood vessels [23–26]. Synergy will be established using themedian-effect analysis proposed by Chou and Talalay [27–29], againstcancer cell models, including a drug resistant breast cancer triple nega-tive model and a drug sensitive melanoma model.

2. Materials and methods

2.1. Materials

Doxorubicin hydrochloride (DXR) was from IdisPharma (UK).Calcein, 4-(2-hydroxyethyl)piperazine-1-ethanesulfonic acid (HEPES),2-(N-morpholino)ethanesulfonic acid (MES), disodium ethylenedi-aminetetraacetate dehydrate (EDTA), Trizma®Base, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), sodiumchloride, 3β-hydroxy-5-cholestene-3-hemisuccinate (CHEMS) andcholesterol (CHOL) were purchased from Sigma-Aldrich (USA). Thelipids 2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2k), 1,2-distearoyl-sn-glycero-3-phosphoethanol-amine-N-[maleimide(polyethylene glycol)-2000] (DSPE-PEG2k-maleimide), and N-hexanoyl-D-erythro-sphingosine (C6-Ceramide)were obtained from Avanti Polar Lipids (USA). F3 (KDEPQRRSARLSAKPAPPKPEPKPKKAPAKK) and the non-specific (NS) peptideswere cus-tom synthetized by Genecust (Luxemburg).

2.2. Cells

MDA-MB-231 triple negative breast cancer cell line and MDA-MB-435Smelanoma cell line [30] (acquired from ATCC, USA) were culturedin RPMI 1640 (Sigma-Aldrich, USA) supplemented with 10% (v/v) ofheat-inactivated fetal bovine serum (FBS) (Invitrogen, USA), 100 U/mlpenicillin, 100 μg/ml streptomycin (Lonza, Switzerland) and main-tained at 37 °C in a 5% CO2 atmosphere. As the cells we have workedwith grow adherent to the plastic of the cell culture flask, all the exper-iments have been performed 24 h after cell seeding, to enable cell at-tachment before the corresponding incubationwith the tested samples.

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

ED P

RO

OF

2.3. In vitro screening for synergy between DXR and C6-ceramide

Eight thousandMDA-MB-231 breast cancer orMDA-MB-435Smela-noma cells/well were incubated with serial dilutions of doxorubicin orC6-Ceramide, alone or in combination, at fixed molar ratios, for 24 h at37 °C in an atmosphere of 5% CO2. Following incubation, cell cultureme-dium was exchanged for fresh one and the experiment was furtherprolonged up to 72 h. Cell viability was then evaluated using the MTTassay as previously described [31].

2.4. Preparation of liposomes

pH-sensitive liposomes, with or without ceramide, were composedof DOPE:CHEMS:DSPC:CHOL:DSPE-PEG2k:C6-ceramide at 4:2:1:1:0.8:2(18 mol.% of C6-ceramide) or 4:2:2:2:0.8:0 molar ratio, respectively.

Dried lipid filmswere hydrated at 60 °Cwith ammonium sulfate (pH8.5) and the resulting liposomes were extruded through 80 nm poresize polycarbonate membranes using a LiposoFast Basic mini extruder(Avestin, Canada). The buffer was exchanged in a Sephadex G-50 gelcolumn (Sigma-Aldrich, USA) equilibrated with Trizma®Base sucrose(10%, w/v, buffered at pH 9.0). Remote encapsulation of DXR (9 or18 mol.% of total lipid, for the DXR:C6-ceramide 1:2 or 1:1 molar ratio,respectively) was carried out through ammonium sulfate gradientmethod, upon incubation with liposomes for 1.5 h at 60 °C [32]. Non-encapsulated DXR was removed using a Sephadex G-50 gel columnequilibrated with 25 mM HEPES, 140 mM NaCl buffer (HBS, pH 7.4).

To further prepare targeted liposomes, DSPE–PEG2k–F3 conjugatewas produced. Briefly, thiolated derivative of F3 peptide was generatedby reaction at room temperature with 2-iminothiolane (Sigma-Aldrich,USA) in 25mMHEPES, 140mMNaCl, 1 mMEDTAbuffer (pH8.0) for 1 hin an inert N2 atmosphere. Thiolated derivatives were then incubatedovernight at room temperature with DSPE–PEG2k–maleimide micellesin 25 mM HEPES, 25 mM MES, 140 mM NaCl, and 1 mM EDTA (pH7.0). The resulting micelles of DSPE–PEG2k–peptide conjugates werepost-inserted onto the liposomal membrane at 2 mol.% relative tototal lipid (TL), upon incubation with pre-formed liposomes, for 1 h at50 °C.

To prepare calcein-loaded liposomes, both lipid films were insteadhydrated with a 40 mM isosmotic calcein solution in 25 mM HEPES,140 mMNaCl buffer (pH 7.4), and extruded as described above. Follow-ing removal of calcein excess, through a Sephadex-G50 column equili-brated with 25 mM HEPES, 140 mM NaCl buffer (pH 7.4), liposomeswere immediately submitted to the post-insertion procedure as previ-ously described.

2.5. Liposome characterization

Liposome size and polydispersion index (PDI) were measured bylight scattering with a N5 particle size analyzer (Beckman Coulter,USA). Final total lipid concentrations were determined upon quantifica-tion of cholesterol using Infinity® Cholesterol kit (ThermoScientific,USA). Encapsulated doxorubicin was assayed at 492 nm from a standardcurve, after liposomal solubilization with 90% absolute ethanol, and theloading efficiency (%) was calculated from the equation [(DXR/TL)final /(DXR/TL)initial] × 100.

To assess drug retention, an aliquot of F3-targeted and non-targetedliposomes, encapsulating either DXR or a combination of DXR andC6-Cer, was incubated in HEPES buffer saline pH 7.4 (HBS), 90%RPMI 1640 culturemedium supplementedwith 10% FBS (Invitrogen)or 90% of non-inactivated bovine serum, at 37 °C. At different time-points (0, 4 and 24 h), DXR fluorescence dequenching was measuredin a Spectramax fluorimeter (λex = 485 nm; λem = 590 nm) (Molec-ular Devices, USA). Drug retention of DXR (% of control) was calculat-ed using the following formula: 100 − [(TestRFUn − MeanRFU0) /(MeanRFUctr − MeanRFU0)] × 100, where TestRFUn and MeanRFU0

stand for the fluorescence of tested sample at different time points

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 3: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

T

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

228228

229

230

231

233233

234

235

236

238238

239

240

241

242

244244

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

283

284

285

286

287

288

289

290

291

292

293

3N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RREC

and time 0 h, respectively and MeanRFUctr is the fluorescence corre-sponding to 100% of release, following incubation with 0.25% (v/v) ofTriton X-100.

2.6. C6-ceramide effect on liposomal pH sensitivity

Two hundred thousand MDA-MB-231 breast cancer and MDA-MB-435S melanoma cells were incubated with 50 μM (total lipid)of liposomal calcein for 1 h at 37 °C and immediately analyzed througha FACScalibur flow cytometer (BD Biosciences, USA). Free calcein(10 μM)was included as control. A total of 20,000 eventswere analyzedwith Cell Quest Pro software (BD Biosciences, USA).

2.7. Evaluation of cytotoxicity of liposomal drug combinations

Different concentrations of F3 peptide-targeted or non-targetedliposomes, containing either single DXR (p[F3]SL or pSL, respective-ly) or combined with C6-ceramide at 1:1 (p[F3]DC11 and pDC11, re-spectively) or 1:2 molar ratio (p[F3]DC12 or pDC12, respectively),were incubated with 8000 MDA-MB-231 breast cancer or MDA-MB-435S melanoma cells/well, for 1, 4 or 24 h, at 37 °C in an atmo-sphere of 5% of CO2. F3 peptide-targeted and non-targeted liposomalC6-ceramide were included as controls (p[F3]SL(C6) and pSL(C6),respectively). Afterwards, cell culture medium was exchanged forfresh one and the experiment was prolonged for a total of 96 h. Cellviability was then evaluated using the MTT assay as previouslydescribed [31].

In order to assess the mechanism of cell death induced by the intra-cellular delivery of DXR:C6-Cer combination, cell death was evaluatedby flow cytometry as previously described, upon incubation of 35,000adherent cells/well with each formulation at 2 μM DXR for 4 h at37 °C [33]. Assessment of cell morphology following DAPI (Applichem,Germany) staining was also performed. Images were captured using a5× objective mounted in an Axiovert 200M microscope equipped withan AxioCamHR (Carl Zeiss, Germany). Image analysis was carried outusing FIJI software (National Institutes of Health, USA).

2.8. Median effect analysis of doxorubicin and C6-ceramide combinations

The nature of the interaction between doxorubicin and C6-ceramide(synergism, additivity or antagonism)was evaluated using themedian-effect model developed by Chou and Talalay [27–29]. The model relieson themedian-effect Eq. (1) to describe any dose–response relationship

f af u

¼ DD50

� �m

ð1Þ

where fa and fu represent the fraction affected and unaffected, respec-tively (i.e. the response), D the dose responsible for a given fa, D50 themedian-effect dose and m the sigmoidicity of a dose–response curve.The acquired data from dose-response studies, using single drugs ortheir combination, was fitted to the median-effect plot Eq. (2)

log1

1− f að Þ� �

¼ m log Dð Þ−m log D50ð Þ ð2Þ

enabling one to estimate m, the slope of the plot, and D50, the dose re-sponsible for 50% of the effect.

These parameters establish a dose–response model for each drug orcombination tested, enabling the determination of the CombinationIndex (CI) using the Eq. (3)

CI ¼ D1 þ D2

Dxð Þ1 þ Dxð Þ2ð3Þ

where (Dx)1 and (Dx)2 are the doses of each single drug responsible fora certain effect fa, and D1 and D2 are the doses of each drug in a given

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

ED P

RO

OF

mixture enabling the same effect fa [28]. As a measure of the nature ofinteraction, a CI value b1,≈1 or N1 is indicative of synergism, additivityor antagonism, respectively [27,28]. Further calculations enable one toestimate the Dose Reduction Index () by the Eq. (4)

DRIn ¼ Dxð ÞnDn

ð4Þ

for the drug n, understood as the drug fold decrease in a combination ascompared to the same drug given alone, for a determined effect level[27,28].

3. Results

3.1. Cytotoxicity of individual drugs against cancer cell lines of diverse his-tological origin

Assessment of single cytotoxicity of the drugs in study is a require-ment for the establishment of their nature of interaction using themedian-effect analysis [28]. Fulfilling that requisite, triple negativebreast cancer MDA-MB-231 and melanoma MDA-MB-435S [30] celllines were incubated with serial dilutions of either DXR or C6-ceramide for 24 h at 37 °C, in an atmosphere of 5% CO2. The formerwas clearlymore resistant to DXR than the latter (IC50= 1.66 μM versusIC50=0.40 μM; Fig. 1, A versus C), in contrast to the higher sensitivity toC6-ceramide (IC50 = 6.15 μM versus IC50 = 11.5 μM; Fig. 1, B versus D,respectively). Expectedly, the described inactive analogue, C6-dihydroceramide, presented a significantly lower activity than C6-ceramide (corresponding to a 10-fold higher IC50), rendering it uncon-sidered for subsequent studies (data not shown).

3.2. Establishment of synergistic combinations of doxorubicin and C6-ceramide

In order to assess the nature of interaction between the two drugs,different doxorubicin (DXR):C6-ceramide (C6-Cer) ratios, from 1:40 to5:1, were tested, using the MDA-MB-231 breast cancer and MDA-MB-435S melanoma cell lines. Data generated from the in vitro screening,were analyzed using the median-effect method described by Chou andTalalay [27,28]. With this method, the combination index (CI), a mea-surement of the nature of interaction between the two drugs, hasbeen determined. A combination index of b1, ≈1 or N1, correspondsto a synergistic, additive or antagonistic interaction, respectively.

The results indicated that the combination of doxorubicin and C6-ceramide exhibited synergistic activity (Fig. 2). Interestingly, the na-ture of the interaction varied according the fraction affected (fa)(percentage of cell death), i.e. the same drug ratio may present si-multaneously an antagonistic/additive or synergistic interaction de-pending on the levels of fa, i.e. cell death (Fig. 2). Surprisingly, theDXR:C6-ceramide molar ratio 1:2 revealed to interact synergisticallyat all fa levels, an effect that was independent of the cell line tested(Fig. 2A and B). Such result translated into a dose-reduction index(DRI) between 4- and 6-fold for doxorubicin. Additionally, other testeddrug ratios also presented relevant DRI values (Fig. 2C and D). However,the nature of the interaction of those did not hold among all fa range.Based on these results, the synergistic DXR:C6-ceramide molar ratio1:2 was selected for further co-encapsulation into liposomes, alongwith the 1:1 molar ratio (mildly additive/antagonistic), as a controlfor the synergy effect.

3.3. Effect of bilayer-incorporated ceramide on intracellular triggereddelivery of pH-sensitive liposomes

The work by Moura et al. (2012), using F3-targeted pH-sensitiveliposomes contaning DXR, has demonstrated in a murine model ofMDA-MB-435 tumors, the therapeutic advantage of endowing a

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 4: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

RO

OF

294

295

296

297

298

299

Q6 Fig. 1. Cytotoxicity of free doxorubicin (DXR) and C6-ceramide (C6-Cer) against breast andmelanoma cancer cell lines.DXR or C6-ceramide (in serially diluted concentrations)were incubatedfor 24 h at 37 °C with either MDA-MB-231 (A and B, respectively) orMDA-MB-435 (C and D, respectively) breast cancer ormelanoma cell lines respectively, and the experiment was fur-ther prolonged for 72 h after which cell death was assessed by the MTT assay. Dose–response curves represent the mean± SEM of 3–4 independent experiments for each concentrationtested. Doted-line: 50% of cell death.

4 N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

nanoparticle with the capacity to enable intracellular triggered re-lease of the encapsulated payload. Therefore, before proceedingwith the loading of the established drug ratio into the pH-sensitive

UNCO

RRECT

Fig. 2. In vitro screening for synergy of different doxorubicin (DXR):C6-ceramide (C6-Cer) ratios. Ffrom 1:40 to 5:1, the nature of the interaction between the two drugs was assessed against MDwell as the Dose Reduction Index (DRI) of doxorubicin, at IC50 (fa 0.5) or IC70 (fa 0.7), for bothbetween drugs was used, where CI values b1, N1 or≈1 indicate synergy, antagonism and adddependent experiments performed in triplicate. Dose reduction index (DRI) as a measure of a dnation, both producing the similar effect “fa”. Mean DRI ± SEM was calculated from data of 3–

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

D Pliposomes, the influence of the presence of ceramide in pH-sensitivity

was assessed. Liposomes were thus loaded with calcein at a self-quenching concentration as previously described [22].

E

ollowing incubation with different doxorubicin (DXR):C6-ceramide (C6-Cer) molar ratios,A-MB-231 breast cancer or MDA-MB-435S melanoma cell lines (A and B, respectively) ascell lines (C and D respectively). Combination Index (CI) as a measure of the interactionitivity, respectively. Mean Combination Index ± SEM was calculated from data of 3–6 in-ose fold-reduction is obtained by the ratio of each drug alone versus the drug in a combi-6 experiments, each performed in triplicate.

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 5: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

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

332

333

334

335

336

337

338

339

340

341

5N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

As such, it was possible to assess the impact on the efficiency of thepayload release following the probe dequenching. F3 peptide-targeted li-posomes containing calcein, with or without C6-ceramide (p[F3]SL(C6)andp[F3]SL, respectively), presented a similar extent of intracellular pay-load release (Fig. 3), with greater efficiency than the corresponding non-targeted counterparts (pSL(C6) and pSL). Not less significant, cell incu-bated with an excess of free calcein presented 4-fold higher signal thanthe non-targeted formulation, but significantly lower than the targetedones (Fig. 3C and D). The first indicated that calcein was clearly retainedinside the liposomes,while the second points to thewidespread signal oftargeted formulations as result of extensive intracellular delivery.

Overall, these results demonstrated that bilayer-incorporated C6-ceramidedid not affect the ability of pH-sensitive F3-targeted liposomesto promote intracellular triggered delivery of the payload located in itsaqueous core.

3.4. Characterization of pH-sensitive F3-peptide targeted liposomesco-encapsulating doxorubicin and C6-ceramide

As shown previously, the DXR:C6-cer 1:2 molar ratio presented asynergistic interaction between the two drugs, contrasting to the 1:1molar ratio, which was mildly additive or antagonist depending on the

UNCO

RRECT

Fig. 3. Effect of membrane-incorporated ceramide on liposomal pH-sensitivity. Two hundred thouwith excess of free calcein (10 μM) or with pH-sensitive liposomes containing calcein either atargeted, pSL(C6), or F3-targeted, p[F3]SL(C6)), at 50 μM of lipid for 1 h at 37 °C and immediplots of event distribution for each tested condition. Calcein geometric mean fluorescence, nor231 and MDA-MB-435S cell lines (C and D, respectively). Data represent the mean ± SEM of 3

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

OO

F

cell line (Fig. 2). Therefore, pH-sensitive F3-peptide targeted and non-targeted liposomes containing the indicated DXR:C6-Cer ratio, wereprepared by incorporating C6-ceramide in the liposomal bilayer at thementioned fixed molar ratios and further characterized from a physicalstandpoint.

The presence of C6-ceramide in the bilayer of liposomes hadmin-imal impact in parameters like loading efficiency, mean size andpolydispersion index, when compared to the targeted (p[F3]SL)and non-targeted (pSL) counterparts without ceramide (Fig. 4A, B andC). Interestingly, non-targeted formulations were more heterogeneousthat the F3-targeted liposomes in terms of mean size (Fig. 4C). Furtherevidence of improved stability arouse from drug release studies per-formed at 37 °C. None of the nanosystems tested, either F3-targetedor non-targeted, presented significant drug release in 4 h, regardlessthe incubation medium. At 24 h of incubation, either in cell culture me-dium (Fig. 4E) or serum (Fig. 4F), all formulations, either targeted ornon-targeted, presented a similar extent of drug retention. The experi-ment performed in serum, evidenced a decrease on the ability to retainthe encapsulated DXR, with values that varied between 55 and 84%, forboth targeted and non-targeted liposomes. Thiswas not statistically dif-ferent from the counterparts without C6-ceramide (Fig. 4F). Strikingly,the stability study performed in HBS (Fig. 4D) clearly demonstrated

ED P

R

sand of MDA-MB-231 breast cancer and MDA-MB-435S melanoma cells were incubatedlone (non-targeted, pSL, or F3-targeted, p[F3]SL) or co-encapsulated with ceramide (non-ately analyzed through a FACScalibur flow cytometer. (A) and (B) are representative dotmalized against the respective signal of the untreated control, is presented for MDA-MB-experiments (one-way ANOVA p b 0.001; ns p N 0.05 Tukey's multiple comparison test).

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 6: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

ECTED P

RO

OF

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

381

382

383

384

385

386

387

Fig. 4. Characterization of liposomes encapsulating doxorubicin (DXR):C6-ceramide (C6-Cer) ratios.Different liposomal formulations incorporating doxorubicin and C6-ceramide at 1:1 or 1:2molar ratios, either targeted (p[F3]DC11 and p[F3]DC12, respectively) or non-targeted (pDC11 and pDC12, respectively) have been characterized, in comparison with the correspondingcontrols containing only DXR, either targeted (p[F3]SL) or non-targeted (pSL), in terms of loading efficiency (A),mean size (B), and polydispersion index (C). (D), (E) and (F) represent thedrug retention capacity of each liposomal formulation when incubated at 37 °C in HBS pH 7.4, RPMI 1640 supplemented with 10% FBS or in 90% non-inactivated serum, respectively. Datarepresent mean ± SEM from 3 to 8 independent experiments (one-way ANOVA ***p b 0.0001, **p b 0.01, *p b 0.05; ns p N 0.05 by Tukey's multiple comparison test).

6 N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RRthat p[F3]DC11 (targeted liposomes encapsulating the DXR:C6-Cer at

1:1 molar ratio) and p[F3]DC12 (targeted liposomes encapsulating theDXR:C6-cer at 1:2molar ratio) presented a similar extent of drug reten-tion, relative the corresponding controls without ceramide (p[F3]SL),and were more stable than the non-targeted counterparts (pDC11 andpDC12, respectively), in accordance with the PDI results (Fig. 4C).

3.5. In vitro cytotoxic of liposomal targeted combinations of doxorubicinand C6-ceramide

In order to evaluate the cytotoxic potential of the targeted drugcombinations, the impact of each formulation on the in vitro viabilityof MDA-MB-231 and MDA-MB-435 cells was assessed. Incubation withp[F3]DC11 or p[F3]DC12 enabled an unique result for an incubationtime as short as 4 h, leading to a 90% decrease on the viability of bothcell lines. This level of decreased viability was not reached following in-cubationwith the counterpart without ceramide (p[F3]SL) in any of thecell lines tested, not even for a 24 h incubation (Fig. 5, and Table 1). In-terestingly, the presence of C6-cer in non-targeted formulations eliciteda decrease in cell viability higher than 90% both in MDA-MB-231 andMBA-MB-435 cells, for 24 h incubations. In any case, these non-targeted formulations presented IC50 and IC90 values 2-fold higherthan the targeted counterparts (Fig. 5 and Table 1).

For short incubation time points (1 h), the previously described ad-vantage arising from the presence of C6-cer was not evident.Within the

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

selected concentrations of DXR, the value of 50% of cell deathwas barelysurpassed. Nevertheless, the IC50 values determined for targeted lipo-somes were 2-fold lower relative to the non-targeted liposomes, an ef-fect that was independent of the incorporation of C6-Cer (Fig. 5 andTable 1).

Overall, these results demonstrated that the co-encapsulation of theDXR:C6-Cer combination within F3-targeted liposomes increased theircytotoxic activity, even with half of the amount of DXR loaded per lipo-some (corresponding to the doxorubicin:C6-ceramide molar ratio of1:2), which ultimately may result in increased targeted cell deathwhile minimizing collateral toxic effects of doxorubicin.

3.6. Effect of the developed synergistic targeted drug combinations on cellmorphology

In order to gain insight into the mechanism of action of the devel-oped nanoparticles, cell morphology was assessed using fluorescencemicroscopy. The collected data indicated that F3-targeted formulationsaltered the distribution of nuclear sizes, inducing nuclear and cell swell-ing to a greater extent than the non-targeted formulations (Fig. 6 andFig. S5, Supplemental Data). The presence of C6-ceramide, in targetedor non-targeted liposomes, did affect neither the nuclear distributionnor the mean nuclear size (Fig. 6B, C, D, E and F). Not less important isthe fact that the liposomes encapsulating the DXR:C6-Cer 1:2 molarratio (p[F3]DC12) presented the same pattern of performance as the

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 7: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

TED P

RO

OF

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

Fig. 5. Cytotoxicity of combinations of doxorubicin and C6-ceramide encapsulated in different liposomal formulations. Cells were incubated for 1, 4 and 24 h with F3-targeted lipo-somal DXR (p[F3]SL) or DXR:C6-Cer combination at a molar ratio of 1:1 or 1:2 (p[F3]DC11 and p[F3]DC12, respectively), at DXR serially diluted concentrations, and the experimentwas further prolonged for total of 96 h, after which cell death was assessed. Results were compared to the respective non-targeted counterparts (pSL, pDC11 and pDC12).Figures represent the dose–response curves for the indicated incubation time points for MDA-MB-231 triple negative breast cancer cell line, and MDA-MB-435S melanoma cell line.The data points represent the mean ± SEM of 3 independent experiments for each concentration tested. Doted-line and full line represent 50 and 90% of cell death, respectively.

t1:1

t1:2Q1

t1:3

t1:4

t1:5

t1:6

t1:7

t1:8

t1:9

t1:10

t1:11

t1:12

t1:13

7N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

ORRECcounterparts encapsulating the double amount of DXR (p[F3]SL or p[F3]

DC11) (Fig. 6F). A similar profile was obtained using the drug sensitivemelanomamodel, MDA-MB-435S (not shown). Overall, these data sug-gested that the F3-targeted formulations were more efficient in induc-ing nuclear and cell swelling, consistent with necrosis-induced celldeath [34].

4. Discussion

Despite all efforts, cancer drug resistance remains a distressing prob-lem in oncology. In order to overcome this barrier, chemotherapeuticsare used in combination, normally at their maximum tolerated dose(MTD), which ultimately is not beneficial to the patient as it representsan increased risk of severe toxicity. Nonetheless, it has been recognized

UNC

Table 1Cytotoxicity of different liposomal DXR:C6-ceramide combinations against MDA-MB-231 and

Drug Cell line

MDA-MB-231

1 h 4 h 24 h

IC50 (μM) IQ90 (μM) IC50 (μM) IC90 (μM) IC50 (

Liposomal doxorubicin p[F3]SL 29.53 – 3.73 – 0.89pSL N50 – 7.16 – 3.65

Liposomal combination p[F3]DC11

25.87 – 6.35 40.79 0.85

pDC11 N50 – 16.21 – 1.47p[F3]DC12

23.60 – 8.85 – 0.90

pDC12 40.12 – 18.13 – 3.28

Data represent the IC50 and the IC90 of the mean dose–response curves calculated through linrespectively.

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

that success demands the identification of drugs that, when combined,lead to synergistic tumor inhibition while minimizing systemic toxicity[5,6]. It has been acknowledge that the nature of the interaction be-tween those drugs varies according to themeasured effect on cell viabil-ity (fraction affected, fa), as well as with the pre-established drugmolarratio, in line with similar studies using drug combinations likeirinotecan/floxuridine or cytarabin/daunorubicin [18,19]. Nonetheless,such scenario is far from ideal as synergistic effects would be limitedto a percentage of tumor cell death above a certain threshold, whichcould be problematic in an in vivo setting. Therefore, ideally a drug com-bination should present a synergistic interaction at all levels of fa. In thisrespect, we have identified in the present work the DXR:C6-Cer 1:2molar ratio as synergistic, regardless the cell line or the level of fa, en-abling at least a 4–6 fold dose reduction of DXR.

MDA-MB-435 cell lines.

MDA-MB-435S

1 h 4 h 24 h

μM) IQ90 (μM) IC50 (μM) IC90 (μM) IC50 (μM) IC90(μM) IC50 (μM) IC90 (μM)

– 9.51 – 8.14 – 0.65 –

– 47.18 – N50 – 6.68 –

10.51 15.98 – 4.88 20.56 1.31 8.71

14.22 35.27 – 15.93 – 2.77 11.8513.60 11.58 – 10.92 – 0.89 17.47

21.19 32.94 – N50 – 3.70 31.74

ear interpolation of the dose values immediately below or above the 50% and 90% effect,

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 8: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

TD P

RO

OF

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

Fig. 6. Effect of F3-targeted combinations of doxorubicin and C6-ceramide on cell morphology. Thirty-five thousandMDA-MB-231 cells/well were incubated for 4 h at 37 °C with F3-targetedliposomal DXR (p[F3]SL) or DXR:C6-Cer combination at amolar ratio of 1:1 or 1:2 (p[F3]DC11 and p[F3]DC12, respectively), or with the non-targeted counterparts (pSL, pDC11 or pDC12,respectively) at 2 μM DXR. The experiment was prolonged up to 92 h. Figures represent the frequency distributions of nuclear area, determined by DAPI staining analysis, for untreatedcells (A), liposomal doxorubicin (B), liposomes encapsulating the DXR:C6-Cer, either at 1:1 (C) or 1:2 molar (D), respectively. (E) Represents the mean nuclear area analysis upon incu-bation with each of the mentioned samples (Bars represent mean area ± SEM; Kruskal–Wallis non-parametric test p b 0.0001; ***p b 0.001 comparing F3-targeted vs non-targeted for-mulations by Dunn's multicomparison test). Data were collected from a representative experiment.

8 N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RREC

However, to take full advantage of such combination, one has todevelop strategies to ensure a synchronized temporal and spatialdelivery of the individual drugs. Several lipid-based formulationscontaining ceramides have been described. Some examples includeceramides formulated either as single agent in non-targeted ortransferrin-targeted nanoparticles [35,36] or in combination withpaclitaxel, doxorubicin or tamoxifen [37–39]. Others have demon-strated that combination of liposomal ceramide with sorafenib in-creases the effectiveness of the latter [40]. Nonetheless, despiteincreased cytotoxicity of those combinations, lack of an appropriatemathematical analysis may render the selected drug ratios not nec-essarily synergistic. Furthermore, EPR-driven tumor accumulationof the mentioned drug combinations, though presenting higher effi-cacy than free drugs, could be limited by the absence of specific intra-cellular delivery to essential (and more accessible) cell populationswithin the tumor microenvironment [4,41]. Considering the impor-tance of targeting different components of the tumor microenviron-ment, as previously emphasized [4,16,17], we have engineered theprevious F3-peptide pH sensitive liposomes developed in ourgroup, to accommodate the selected DXR:C6-Cer synergistic combi-nation. The F3 peptide is a moiety that specifically targets nucleolin,a cell membrane protein overexpressed in cancer cells and endothe-lial cells of angiogenic blood vessels [22,42].

Using calcein-loaded liposomes, we have shown that incorporationof C6-ceramide in the liposomal bilayer did not impair delivery norpromoted significant instability (Fig. 3, non-targeted liposomes ver-sus free calcein). Such result correlates with the one generated withthe 35 mol.% C6-ceramide-incorporating liposomes by Khazanovet al. (maximum load), well above the 18.5 mol.% we used [37]. Theabsence of impaired delivery led us to engineer a F3-targeted formu-lation containing the synergistic DXR:C6-Cer 1:2 molar ratio (Fig. 2).DXR:C6-Cer 1:1 molar ratio was also loaded into liposomes to use asa control for an additive/antagonist effect (Fig. 2).

Characterization data demonstrated that C6-ceramide had mini-mal impact in measured physical parameters (including drug

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

Eretention) as compared to F3-targeted (p[F3]SL) or non-targeted(pSL) liposomes containing only doxorubicin (Fig. 4). DSPC and cho-lesterol are known to increase drug retention in pH sensitive lipo-somes [43]. Their removal to accommodate the incorporation ofC6-ceramide might have led to increased defects in membrane bilay-er promoted by the sphingolipid [37]. This was likely the reason be-hind the differences observed on drug retention (in HBS) betweendoxorubicin- and combination-encapsulating liposomes. Nonethe-less, the same data demonstrated that the F3 ligand stabilized the bi-layer of C6-ceramide loaded liposomes, to the same extent ofliposomes without C6-ceramide (Fig. 4D). This might be explainedby the additional hydration layer on the liposomal surface, arisingfrom the charged F3 peptide. This effect seems to improve drug re-tention (Fig. 4D) and minimize aggregation (Fig. 4C). Furthermore,the differences observed in the different media, might be related totheir differences in ionic and/or protein composition. Those, inturn, may promote different degrees of lipid headgroup hydration,especially of CHEMS, upon providing several counterions, which ren-der unequal levels of membrane stabilization [44,45]. This fact couldexplain the increased drug retention of pDC11 and pDC12 formula-tions in cell culture medium when compared to HBS buffer. Other-wise, in serum, all tested formulations exhibited lower drugretention than in other media, notwithstanding the fact thatcombination-containing liposomes have marginally lower drug re-tention, relative to the counterparts without ceramide (Fig. 4F).That could be due to protein/lipid interaction known to destabilizethe lipid bilayers allowing doxorubicin to escape and bind to serumproteins [46,47].

The incorporation of pro-apoptotic ceramide in liposome bilayer,by itself, has shown to render any formulation more cytotoxic eitherby passive internalization of the intact liposome or by simple lipidtranslocation onto the cell membrane [37]. The free drug combina-tion studies clearly indicated that the 1:2 DXR:C6-cer molar ratiowas synergistic, whereas the 1:1 molar ratio was mildly additive orantagonistic (Fig. 2). Surprisingly, F3-targeted liposomes

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 9: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

T

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

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

556Q3

557

558

559

560

561

562563564565566567568569570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604605606607608609610611612613614615616617618619620621

9N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

UNCO

RREC

encapsulating the DXR:C6-Cer combination at 1:1 molar ratio (p[F3]DC11) enabled 90% of cell death for an incubation period as short as4 h (Fig. 5 and Table 1). This effect was unattainable even by targetedliposomes containing only doxorubicin or C6-ceramide, after 24 h ofincubation (Fig. S1, Supplemental data). Overall, p[F3]DC11 performedsimilarly to p[F3]DC12 nanoparticle against triple negative MDA-MB-231 breast cancer cells, regardless the incubation time (Fig. 5 andTable 1). This unexpected result suggested that the intracellulardelivery of both drugs, at the selected ratio, may change the interactionnature of the combinations, rendering them synergistic (Fig. S2,Supplemental data). This result is, in some extent, different from theone reported by Tardi et al., where encapsulation (in non-targeted lipo-somes) of antagonist 3:1 ratio of irinotecan:cisplatin rendered a lowerefficacy, despite increased irinotecan loading, compared to the synergis-tic CPX-571 formulation [48]. This thus indicated that the interactionbetween drugs might also be dependent on the mechanism of drug de-livery, particularly on the ability to delivery intracellularly a specificdrug ratio. Nonetheless, it has been demonstrated that the encapsula-tion of the combination at 1:2 molar ratio (p[F3]DC12) enabled in-creased liposomal citotoxicity using half of the drug loading utilized toprepare the standard targeted formulation (p[F3]SL). Ultimately, thiscould prove useful to further increase the safety associated with theuse of liposomal doxorubicin.

Despite the increased cytotoxic effect, FACS analysis indicated simi-lar mechanism of cell death for all formulations, with predominance oflate apoptotic/necrotic population and higher levels of apoptotic cellsover necrotic cells (Fig. S4, Supplemental data). Doxorubicin has beendescribed to induce apoptosis (caspase-dependent cell death) or necro-sis (caspase-independent cell death) after mitotic catastrophe [49,50],in a dose-dependent manner. On the other hand, ceramides have beendescribed to mediate apoptosis through several mechanisms includingp38 MAPK, Akt-mediated induction of p27(kip1) and channel-opening in the mitochondria [9,10,51–53]. In this context, whereasDXR depends on cell cycle to promote cell death even at low doses,the apoptosis-induced effect by C6-ceramide is highly dependent onthe dose and time of exposure [9,49]. Consistently, microscopy analysisof nuclear geometry, revealed an increase in the nuclei size, indicative ofnucleus swelling, and cell swelling upon incubation with F3-targetednanoparticles containing DXR, when compared to the non-targetedcounterparts, which is coherent with cell induced-necrosis (Fig. 6)[34]. Therefore, at the tested concentration, observed results demon-strated mainly the contribution of doxorubicin for cell death, despitethe fact that free ceramide can indeed induce apoptotic cell death, asmeasured by us with annexin V (Fig. S3, Supplemental data) and consis-tent with literature [10].

5. Conclusion

In the present work, it has been demonstrated that the DXR:C6-cercombination developed a synergistic interaction at the specific 1:2molar ratio, enabling a DXR dose reduction of, at least, 4-fold de-pending on the cellular model. Furthermore, we established the de-velopment of a novel triggered-release nanoparticle targeted bythe F3 peptide, capable of retaining and intracellularly deliver thesynergistic DXR:C6-cer combination, thus increasing the cytotoxicpotential. Additionally, our study suggested that the strategy of de-livery may alter the nature of drug interaction, since a F3-targetedformulation encapsulating an additive/mildly antagonistic DXR:C6-Cer, demonstrated singularly a cytotoxic effect above 90%, for an in-cubation period as short as 4 h. It has been further validated that theencapsulation of the synergistic DXR:C6-Cer 1:2 molar ratio enableda cytotoxic effect above 90% after 24 h of incubation, unattainable bythe F3-targeted liposomal doxorubicin encapsulating twice theamount. The characteristics of the developed nanoparticle, alongwith the demonstrated tropism of the F3 peptide towards breast can-cer cells and the tumor microenvironment [22,42], could enable an

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

ED P

RO

OF

increase in the therapeutic efficacy, overcoming drug resistancewhile simultaneously decreasing the severe side effects of doxorubi-cin. Overall, the generated data suggest that additive/antagonistic inter-action of free combinationsmay be potentially shifted by specific activeintracellular delivery.

Acknowledgments

Nuno A. Fonseca was a recipient of the fellowship SFRH/BD/64243/2009 from the Portuguese Foundation for Science and Technology(FCT). The work was supported by the grants InovC/UC (2013) andQREN/FEDER/COMPETE (Ref. 30248/IN0617).

Appendix A. Supplementary data

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.jconrel.2014.09.024.

References

[1] A. Jemal, F. Bray, M.M. Center, J. Ferlay, E. Ward, D. Forman, Global cancer statistics,CA Cancer J. Clin. 61 (2011) 69–90.

[2] R. Siegel, D. Naishadham, A. Jemal, Cancer statistics, 2013, CA Cancer J. Clin. 63(2013) 11–30.

[3] T. Saeki, T. Tsuruo, W. Sato, K. Nishikawsa, Drug resistance in chemotherapy forbreast cancer, Cancer Chemother. Pharmacol. 56 (Suppl. 1) (2005) 84–89.

[4] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144(2011) 646–674.

[5] A. Dicko, L.D. Mayer, P.G. Tardi, Use of nanoscale delivery systems to maintain syn-ergistic drug ratios in vivo, Expert Opin. Drug Deliv. 7 (2010) 1329–1341.

[6] S. Ramaswamy, Rational design of cancer-drug combinations, N. Engl. J. Med. 357(2007) 299–300.

[7] J. Baselga, Targeting the phosphoinositide-3 (PI3) kinase pathway in breast cancer,Oncologist 16 (Suppl. 1) (2011) 12–19.

[8] K.D. Courtney, R.B. Corcoran, J.A. Engelman, The PI3K pathway as drug target inhuman cancer, J. Clin. Oncol. 28 (2010) 1075–1083.

[9] H.J. Kim, J.E. Oh, S.W. Kim, Y.J. Chun, M.Y. Kim, Ceramide induces p38 MAPK-dependent apoptosis and Bax translocation via inhibition of Akt in HL-60 cells, Can-cer Lett. 260 (2008) 88–95.

[10] S.W. Kim, H.J. Kim, Y.J. Chun, M.Y. Kim, Ceramide produces apoptosis through induc-tion of p27(kip1) by protein phosphatase 2A-dependent Akt dephosphorylation inPC-3 prostate cancer cells, J. Toxicol. Environ. Health A 73 (2010) 1465–1476.

[11] Y.Y. Liu, V. Gupta, G.A. Patwardhan, K. Bhinge, Y. Zhao, J. Bao, H. Mehendale, M.C.Cabot, Y.T. Li, S.M. Jazwinski, Glucosylceramide synthase upregulates MDR1 expres-sion in the regulation of cancer drug resistance through cSrc and beta-catenin sig-naling, Mol. Cancer 9 (2010) 145.

[12] Y. Uchida, M. Itoh, Y. Taguchi, S. Yamaoka, H. Umehara, S. Ichikawa, Y. Hirabayashi,W.M. Holleran, T. Okazaki, Ceramide reduction and transcriptional up-regulation ofglucosylceramide synthase through doxorubicin-activated Sp1 in drug-resistant HL-60/ADR cells, Cancer Res. 64 (2004) 6271–6279.

[13] V. Gouaze, Y.Y. Liu, C.S. Prickett, J.Y. Yu, A.E. Giuliano, M.C. Cabot, Glucosylceramidesynthase blockade down-regulates P-glycoprotein and resensitizes multidrug-resistant breast cancer cells to anticancer drugs, Cancer Res. 65 (2005) 3861–3867.

[14] Y.Y. Liu, J.Y. Yu, D. Yin, G.A. Patwardhan, V. Gupta, Y. Hirabayashi, W.M. Holleran, A.E.Giuliano, S.M. Jazwinski, V. Gouaze-Andersson, D.P. Consoli, M.C. Cabot, A role forceramide in driving cancer cell resistance to doxorubicin, FASEB J. 22 (2008)2541–2551.

[15] C. Ji, B. Yang, Y.L. Yang, S.H. He, D.S. Miao, L. He, Z.G. Bi, Exogenous cell-permeable C6ceramide sensitizes multiple cancer cell lines to doxorubicin-induced apoptosis bypromoting AMPK activation and mTORC1 inhibition, Oncogene 29 (2010)6557–6568.

[16] R.R. Bansode, Coupling in vitro and in vivo paradigm reveals a dose dependent inhi-bition of angiogenesis followed by initiation of autophagy by C6-ceramide, Int. J.Biol. Sci. (2011) 629–644.

[17] G. Bocci, A. Fioravanti, P. Orlandi, T. Di Desidero, G. Natale, G. Fanelli, P. Viacava, A.G.Naccarato, G. Francia, R. Danesi, Metronomic ceramide analogs inhibit angiogenesisin pancreatic cancer through up-regulation of caveolin-1 and thrombospondin-1and down-regulation of cyclin D1, Neoplasia 14 (2012) 833–845.

[18] L.D. Mayer, T.O. Harasym, P.G. Tardi, N.L. Harasym, C.R. Shew, S.A. Johnstone, E.C.Ramsay, M.B. Bally, A.S. Janoff, Ratiometric dosing of anticancer drug combinations:controlling drug ratios after systemic administration regulates therapeutic activityin tumor-bearing mice, Mol. Cancer Ther. 5 (2006) 1854–1863.

[19] P. Tardi, S. Johnstone, N. Harasym, S. Xie, T. Harasym, N. Zisman, P. Harvie, D.Bermudes, L. Mayer, In vivomaintenance of synergistic cytarabine:daunorubicin ra-tios greatly enhances therapeutic efficacy, Leuk. Res. 33 (2009) 129–139.

[20] E.J. Feldman, J.E. Lancet, J.E. Kolitz, E.K. Ritchie, G.J. Roboz, A.F. List, S.L. Allen, E.Asatiani, L.D. Mayer, C. Swenson, A.C. Louie, First-in-man study of CPX-351: a lipo-somal carrier containing cytarabine and daunorubicin in a fixed 5:1 molar ratiofor the treatment of relapsed and refractory acute myeloid leukemia, J. Clin. Oncol.29 (2011) 979–985.

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024

Page 10: Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination

T

622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653654655656657658659660661Q5662663664665666667668669670

671672673674675676677678679680681682683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717

718

10 N.A. Fonseca et al. / Journal of Controlled Release xxx (2014) xxx–xxx

C

[21] W.S. Lim, P.G. Tardi, N. Dos Santos, X. Xie, M. Fan, B.D. Liboiron, X. Huang, T.O.Harasym, D. Bermudes, L.D. Mayer, Leukemia-selective uptake and cytotoxicity ofCPX-351, a synergistic fixed-ratio cytarabine:daunorubicin formulation, in bonemarrow xenografts, Leuk. Res. 34 (2010) 1214–1223.

[22] V. Moura, M. Lacerda, P. Figueiredo, M.L. Corvo, M.E. Cruz, R. Soares, M.C. de Lima, S.Simoes, J.N. Moreira, Targeted and intracellular triggered delivery of therapeutics tocancer cells and the tumor microenvironment: impact on the treatment of breastcancer, Breast Cancer Res. Treat. 133 (2012) 61–73.

[23] A.G. Hovanessian, C. Soundaramourty, D.E. Khoury, I. Nondier, J. Svab, B. Krust, Sur-face expressed nucleolin is constantly induced in tumor cells to mediate calcium-dependent ligand internalization, PLoS One 5 (2010) e15787.

[24] H. Shi, Y. Huang, H. Zhou, X. Song, S. Yuan, Y. Fu, Y. Luo, Nucleolin is a receptor thatmediates antiangiogenic and antitumor activity of endostatin, Blood 110 (2007)2899–2906.

[25] S. Christian, J. Pilch, M.E. Akerman, K. Porkka, P. Laakkonen, E. Ruoslahti, Nucleolinexpressed at the cell surface is a marker of endothelial cells in angiogenic blood ves-sels, J. Cell Biol. 163 (2003) 871–878.

[26] M. Srivastava, H.B. Pollard, Molecular dissection of nucleolin's role in growth andcell proliferation: new insights, FASEB J. 13 (1999) 1911–1922.

[27] T.C. Chou, The mass-action law based algorithm for cost-effective approach for can-cer drug discovery and development, Am. J. Cancer Res. 1 (2011) 925–954.

[28] T.C. Chou, Theoretical basis, experimental design, and computerized simulation ofsynergism and antagonism in drug combination studies, Pharmacol. Rev. 58(2006) 621–681.

[29] T.C. Chou, P. Talalay, Quantitative analysis of dose-effect relationships: the combinedeffects of multiple drugs or enzyme inhibitors, Adv. Enzyme Regul. 22 (1984) 27–55.

[30] J.M. Rae, C.J. Creighton, J.M. Meck, B.R. Haddad, M.D. Johnson, MDA-MB-435 cells arederived from M14 melanoma cells—a loss for breast cancer, but a boon for melano-ma research, Breast Cancer Res. Treat. 104 (2007) 13–19.

[31] J.N. Moreira, T. Ishida, R. Gaspar, T.M. Allen, Use of the post-insertion technique toinsert peptide ligands into pre-formed stealth liposomes with retention of bindingactivity and cytotoxicity, Pharm. Res. 19 (2002) 265–269.

[32] G. Haran, R. Cohen, L.K. Bar, Y. Barenholz, Transmembrane ammonium sulfate gradi-ents in liposomes produce efficient and stable entrapment of amphipathic weakbases, Biochim. Biophys. Acta 1151 (1993) 201–215.

[33] A. Santos, A.B. Sarmento-Ribeiro, M.C. de Lima, S. Simoes, J.N. Moreira, Simultaneousevaluation of viability and Bcl-2 in small-cell lung cancer, Cytometry A 73A (2008)1165–1172.

[34] B.S. Cummings, L.P. Wills, R.G. Schnellmann, Measurement of cell death in Mamma-lian cells, Curr. Protoc. Pharmacol. (2012) (Chapter 12, Unit12 18).

[35] T.C. Stover, A. Sharma, G.P. Robertson, M. Kester, Systemic delivery of liposomalshort-chain ceramide limits solid tumor growth in murine models of breast adeno-carcinoma, Clin. Cancer Res. 11 (2005) 3465–3474.

[36] A. Koshkaryev, A. Piroyan, V.P. Torchilin, Increased apoptosis in cancer cells in vitroand in vivo by ceramides in transferrin-modified liposomes, Cancer Biol. Ther. 13(2012) 50–60.

[37] E. Khazanov, A. Priev, J.P. Shillemans, Y. Barenholz, Physicochemical and biologicalcharacterization of ceramide-containing liposomes: paving the way to ceramidetherapeutic application, Langmuir 24 (2008) 6965–6980.

UNCO

RRE

Please cite this article as: N.A. Fonseca, et al., Simultaneous active intracelluonistic drug interaction of non..., J. Control. Release (2014), http://dx.doi.o

ED P

RO

OF

[38] L.E. van Vlerken, Z. Duan, M.V. Seiden, M.M. Amiji, Modulation of intracellular cer-amide using polymeric nanoparticles to overcome multidrug resistance in cancer,Cancer Res. 67 (2007) 4843–4850.

[39] S.A. Morad, J.P. Madigan, J.C. Levin, N. Abdelmageed, R. Karimi, D.W. Rosenberg, M.Kester, S.S. Shanmugavelandy, M.C. Cabot, Tamoxifen magnifies therapeutic impactof ceramide in human colorectal cancer cells independent of p53, Biochem.Pharmacol. 85 (2013) 1057–1065.

[40] M.A. Tran, C.D. Smith,M. Kester, G.P. Robertson, Combining nanoliposomal ceramidewith sorafenib synergistically inhibits melanoma and breast cancer cell survival todecrease tumor development, Clin. Cancer Res. 14 (2008) 3571–3581.

[41] L.C. Gomes-da-Silva, A.O. Santos, L.M. Bimbo, V. Moura, J.S. Ramalho, M.C. Lima, S.Simoes, J.N. Moreira, Towards a siRNA-containing nanoparticle targeted to breastcancer cells and the tumor microenvironment, Int. J. Pharm. 434 (2012) 9–19.

[42] K. Porkka, P. Laakkonen, J.A. Hoffman, M. Bernasconi, E. Ruoslahti, A fragment of theHMGN2 protein homes to the nuclei of tumor cells and tumor endothelial cellsin vivo, Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 7444–7449.

[43] T. Ishida, Y. Okada, T. Kobayashi, H. Kiwada, Development of pH-sensitive liposomesthat efficiently retain encapsulated doxorubicin (DXR) in blood, Int. J. Pharm. 309(2006) 94–100.

[44] X. Li, M. Schick, Theory of tunable pH-sensitive vesicles of anionic and cationic lipidsor anionic and neutral lipids, Biophys. J. 80 (2001) 1703–1711.

[45] I.M. Hafez, P.R. Cullis, Cholesteryl hemisuccinate exhibits pH sensitive polymorphicphase behavior, Biochim. Biophys. Acta 1463 (2000) 107–114.

[46] D. Agudelo, P. Bourassa, J. Bruneau, G. Berube, E. Asselin, H.A. Tajmir-Riahi, Probingthe binding sites of antibiotic drugs doxorubicin and N-(trifluoroacetyl) doxorubicinwith human and bovine serum albumins, PLoS One 7 (2012) e43814.

[47] T. Hernandez-Caselles, J. Villalain, J.C. Gomez-Fernandez, Influence of liposomecharge and composition on their interaction with human blood serum proteins,Mol. Cell. Biochem. 120 (1993) 119–126.

[48] P.G. Tardi, N. Dos Santos, T.O. Harasym, S.A. Johnstone, N. Zisman, A.W. Tsang, D.G.Bermudes, L.D. Mayer, Drug ratio-dependent antitumor activity of irinotecan andcisplatin combinations in vitro and in vivo, Mol. Cancer Ther. 8 (2009) 2266–2275.

[49] Y.W. Eom, M.A. Kim, S.S. Park, M.J. Goo, H.J. Kwon, S. Sohn, W.H. Kim, G. Yoon, K.S.Choi, Two distinct modes of cell death induced by doxorubicin: apoptosis and celldeath throughmitotic catastrophe accompanied by senescence-like phenotype, On-cogene 24 (2005) 4765–4777.

[50] S. Mansilla, W. Priebe, J. Portugal, Mitotic catastrophe results in cell death bycaspase-dependent and caspase-independent mechanisms, Cell Cycle 5 (2006)53–60.

[51] L.J. Siskind, T.D. Mullen, K. Romero Rosales, C.J. Clarke,M.J. Hernandez-Corbacho, A.L.Edinger, L.M. Obeid, The BCL-2 protein BAK is required for long-chain ceramide gen-eration during apoptosis, J. Biol. Chem. 285 (2010) 11818–11826.

[52] L.J. Siskind, R.N. Kolesnick, M. Colombini, Ceramide forms channels inmitochondrialouter membranes at physiologically relevant concentrations, Mitochondrion 6(2006) 118–125.

[53] L.J. Siskind, Mitochondrial ceramide and the induction of apoptosis, J. Bioenerg.Biomembr. 37 (2005) 143–153.

lar delivery of doxorubicin and C6-ceramide shifts the additive/antag-rg/10.1016/j.jconrel.2014.09.024