ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 Acta Biomaterialia xxx (2015) xxx–xxx 1

Contents lists available at ScienceDirect

Acta Biomaterialia journal homepage: www.elsevier.com/locate/actabiomat 5 6 3 4

Q1

7

Q2

8 9 10 11

Q3

Chang-Fang Wang a,⇑, Ermei M. Mäkilä a,b, Martti H. Kaasalainen b, Marja V. Hagström c, Jarno J. Salonen b, Jouni T. Hirvonen a, Hélder A. Santos a,⇑ a b c

12 13

Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Finland Laboratory of Industrial Physics, Department of Physics and Astronomy, University of Turku, FI-20014, Finland Centre for Drug Research, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland

a r t i c l e

1 2 5 8 16 17 18 19 20 21 22 23 24 25 26 27

Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and combination therapy

i n f o

Article history: Received 23 September 2014 Received in revised form 12 January 2015 Accepted 16 January 2015 Available online xxxx

Q4

Keywords: Intracellular uptake Sustained release Combination therapy Porous silicon nanoparticle Methotrexate

a b s t r a c t Dual-drug delivery of antiangiogenic drug and chemotherapeutic drug can enhance the therapeutic effect for cancer therapy. Conjugation of methotrexate (MTX) to PSi nanoparticles (MTX–PSi) with positively charged surfaces can improve the cellular uptake of MTX and inhibit the proliferation of cancer cells. Herein, MTX–PSi conjugates sustained the release of MTX up to 96 h, and the released fragments including MTX were confirmed by mass spectrometry. The intracellular distribution of the MTX–PSi nanoparticles was confirmed by transmission electronic microscopy. Compared to pure MTX, the MTX–PSi achieved similar inhibition of cell proliferation in folate receptor (FR) over-expressing U87 MG cancer cells, and a higher effect in low FR-expressing EA.hy926 cells. Nuclear fragmentation analysis demonstrated programed cell apoptosis of the MTX–PSi in the high/low FR-expressing cancer cells, whereas PSi alone at the same dose had a minor effect on cell apoptosis. Finally, the porous structure of MTX– PSi enabled a successful concomitant loading of another anti-angiogenic hydrophobic drug, sorafenib, and considerably enhanced the dissolution rate of sorafenib. Overall, the MTX–PSi nanoparticle system can be used as a platform for combination chemotherapy by enhancing the dissolution rate of the hydrophobic drug and sustaining the release of the conjugated chemotherapeutic drug. Ó 2015 Published by Elsevier Ltd. on behalf of Acta Materialia Inc.

29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45

46 47

1. Introduction

48

Nanomedicines have been investigated for cancer therapy for several decades, with several products in clinical trials or in the market [1]. Safe and efficient delivery of poorly-water soluble and/or low permeable chemotherapeutic drugs is still one of the main tasks to be overcome by cancer medicines. Porous silicon (PSi) nanomaterials possess high biocompatibility and have a number of unique properties that render them as potential drug delivery carriers in biomedical applications [2–12], such as increasing the dissolution rate of poorly water-soluble drugs [13], high drug loading capacity [14], and tunable surface structure for tailoring the biological activities by different surface chemistries [8,15–17]. For example, the carboxylic acid- and amine-terminated

49 50 51 52 53 54 55 56 57 58 59

⇑ Corresponding authors at: Division of Pharmaceutical Chemistry and mTechnology, Faculty of Pharmacy, P.O. Box 56 (Viikinkaari 5E), University of Helsinki, FI-00014, Finland. Tel.: +358 9 19159661; fax: +358 9 19159144. E-mail addresses: chang-fang.wang@helsinki.fi (C.-F. Wang), helder.santos@ helsinki.fi (H.A. Santos).

surfaces of PSi can be used for further chemical functionalization [15,18,19]. Chemical conjugation of anticancer drugs to nanoparticles has been one of the approaches to overcome the drug solubility/permeability obstacles for the delivery of chemotherapeutics [20,21]. Methotrexate (MTX) is a folic acid analog anticancer drug [22]. MTX is able to inhibit the activity of dihydrofolate reductase (DHFR) enzyme in the cytosol of the cells, which results in the suppression of purine and pyrimidine precursor synthesis and consequently DNA biosynthesis, leading to cell apoptosis [23]. MTX has specific affinity to the folate receptor (FR) and good cellular uptake by certain cells due to the FR-mediated cellular uptake pathway [24]. Over-expression of the FR has been found in many human malignancies, especially when associated with aggressively growing tumors [25]. The cancer cells highly expressing the FR include ovarian, endometrial, colorectal, breast, lung, renal cell carcinoma, brain metastases derived from epithelial cancer, and neuroendocrine carcinoma cells [25,26]. However, MTX has low cellular uptake in receptor deficient cells [20]. As with many other chemotherapeutics, drug resistance has been an obstacle faced by patients when treated with MTX [22].

http://dx.doi.org/10.1016/j.actbio.2015.01.021 1742-7061/Ó 2015 Published by Elsevier Ltd. on behalf of Acta Materialia Inc.

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 2 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

C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

One mechanism of the MTX resistance is to decrease the receptor expression and thus reduce the receptor-mediated cell uptake [27]. Cationic nanoparticles can be non-specifically up-taken in the cells through endocytosis by interacting with the negatively charged cell membrane. By chemically conjugating MTX to the cationic carriers, the cellular uptake can be increased [28–30]. Conjugation of MTX to chitosan [28], dendrimeric polyamidoamine [29,31,32], poly-L-lysine [20,30,33], targeting peptide cyclo(1,12) PenPRGGSVLVTGC [34], gelatin [35], human serum albumin [36], magnetic nanoparticles [37], quantum dots [38], multi-walled carbon nanotubes [39], and porous silica [40] have been reported to enhance the cell uptake and/or to overcome drug resistance. The conjugation was achieved by a covalent linkage formed between the carboxylic acid group of the MTX and the amine groups of the conjugated carriers. The 3-aminopropyltriethoxysilane (APTES) surface functionalized thermally carbonized PSi particles have amine-terminated groups on the surface of the PSi [19]. The conjugation of MTX to these biodegradable PSi nanoparticles with cationic charge can aid in improving the MTX cell uptake and inhibit the proliferation of cancer cells. The porous structure of MTX-conjugated PSi nanoparticles can be further used to load another hydrophobic drug for combination therapy. For example, the anti-angiogenic sorafenib is a multi-kinase inhibitor by targeting RAf kinase, platelet-derived growth factor, vascular endothelial growth factor (VEGF) receptor 2&3 kinases and c-Kit receptor [41]. Clinical studies have reported a high interindividual variability in the pharmacokinetics, clinical efficacy and adverse effects of sorafenib treatment [42]. Side effects and variation in pharmacokinetics caused by low solubility increase the risks associated with the clinical applications of sorafenib [43,44]. By enhancing the aqueous solubility of sorafenib, controllable plasma concentrations and more precise control on the effects of sorafenib can be attained, with reduced clinical risks to patients. In this work, the chemotherapeutical anticancer drug MTX was chemically conjugated to the amine-terminated thermally carbonized PSi to form MTX–PSi nanoparticles for enhancing the cell uptake and sustaining the release of MTX. Simultaneously, the hydrophobic anti-angiogenic sorafenib was loaded to the MTX– PSi to enhance the dissolution rate of this drug for combination therapy. The propose of the dual-drug delivery system developed in this work was to: (1) enhance the dissolution rate of the poor water soluble anti-angiogenic drug, sorafenib, which can inhibit the kinase of the neovascular cells on the cell membrane to slow down the tumor growth; and (2) to enhance the cellular uptake and sustain the release of the chemotherapeutical drug MTX to kill tumor cells. The conjugation of MTX, sorafenib loading to MTX–PSi nanoparticles, the dissolution profiles of MTX and sorafenib, as well as the anti-proliferation and cell apoptosis of MTX–PSi were evaluated in this study.

131

2. Materials and methods

132

2.1. Materials and cell culturing

133

MTX was purchased from TCI (Chuo-ku, Japan). Sorafenib was obtained from LC Laboratories (Woburn, USA). All other chemicals and solvents purchased were of analytical grade from Sigma– Aldrich (USA) and used as received. Hypoxanthine–aminopterin– thymidine (50 HAT) was purchased from GibcoÒ (Carlsbad, USA). Dulbecco’s phosphate buffered saline (10 PBS) and Hank’s balanced salt solution (10 HBSS), Dulbecco’s Modified Eagle’s Medium (DMEM), fetal bovine serum (FBS), trypsin (2.5%), sodium pyruvate, nonessential amino acids (100 NEAA), L-glutamine (100), penicillin–streptomycin (100) were all purchased from

134 135 136 137 138 139 140 141 142

HyClone (Waltham, USA). CellTiter-GloÒ luminescent cell viability assay kit was purchased from Promega (Madison, USA). MTX was dissolved in dimethylsulfoxide (DMSO, 2 mM) as stock solution for further tests. Endothelial EA.hy926 (ATCC, USA) cells were incubated in DMEM with high glucose and 2% HAT, while brain cancer cells U87 MG (ATCC, USA) were cultured with DMEM with low glucose. Both cell lines were cultured in 75 cm2 flasks for further experiments at 37 °C with humidified atmosphere containing 5% CO2, and the DMEM was supplemented by 10% FBS, 1% sodium pyruvate, 1% NEAA, 1% L-glutamine, and 1% penicillin–streptomycin (100 IU/mL).

143

2.2. Preparation of PSi nanoparticles

155

Amine-functionalized thermally carbonized PSi nanoparticles were prepared by adapting the procedure as previously reported elsewhere [19]. Briefly, multilayer PSi films were produced by electrochemically etching monocrystalline p+-type Sih1 0 0i wafers of 0.01–0.02 X cm resistivity in a 1:1 (v/v) aqueous hydrofluoric acid (HF, 40%) – ethanol electrolyte as described previously [4]. The free standing films were thermally carbonized with acetylene at 500 °C and 820 °C [13], after which the obtained films were immersed into HF to generate surface silanol termination for APTES attachment, following the previously described process using a 10 v-% APTES-toluene solution [19]. The size reduction of the PSi multilayer films to nanoparticles was performed by wet-milling using a 5 v-% APTES-toluene solution as the milling liquid. After milling, the excess silane was removed by replacing the liquid and re-dispersing the nanoparticles to fresh toluene and ethanol at least 3 times using centrifugation. The final size selection of the nanoparticles was done by centrifugation.

156

2.3. Conjugation of MTX to PSi nanoparticles

173

MTX was conjugated to the amine-terminated PSi nanoparticles using N-hydroxysuccinimide (NHS) and 1-Ethyl-3-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) coupling reaction. Briefly, 2.2 mg (5 lmol) of MTX was dissolved in 1 mL DMSO with 10 mM of 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES, pH 5.5), 1.7 mg (15 lmol) of NHS and 13 lL (60 lmol of EDC) were added to the solution. The mixture was stirred for 30 min at room temperature to activate the carboxylic acid group of MTX. After that, 1 mg of the amine-terminated PSi nanoparticles in ethanol was added to the reaction mixture, and the pH was adjusted to 7.5 by 1 M NaOH. After 45 min reaction, the PSi nanoparticles were collected from the reaction mixture by centrifugation (Sorvall RC 5B plus, thermo Fisher Scientific, USA) at 10,000 rcf for 3 min and washed with 1 mL of DMSO, water and ethanol 3 times to obtain the MTX conjugated PSi nanoparticles (MTX–PSi), which were then re-suspended in ethanol for further use.

174

2.4. Physicochemical characterization of PSi nanoparticles

191

The physical parameters of the PSi nanoparticles were determined by nitrogen sorption at 196 °C using TriStar 3000 (Micromeritics Inc., USA). The specific surface area of the PSi nanoparticles was calculated using the Brunauer–Emmett–Teller theory. The total pore volume was obtained as the total adsorbed amount at a relative pressure p/p0 = 0.97. The average pore diameter was calculated from the obtained surface area and pore volume by assuming the pores as cylindrical. The qualitative analysis of the MTX–PSi nanoparticles were performed by Fourier transform infrared spectroscopy (FTIR) with a Vertex 70 FTIR spectrometer (Bruker Optics, USA) using a

192

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

144 145 146 147 148 149 150 151 152 153 154

157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172

175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190

193 194 195 196 197 198 199 200 201 202

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

226

horizontal attenuated total reflectance (ATR) accessory (MIRacle, PIKE Technologies, USA). The transmittance spectra were recorded between 4000 and 650 cm1 with a 4 cm1 resolution. The amount of MTX covalently conjugated to the PSi nanoparticles was determined by elemental analysis using a Vario Micro cube CHN analyzer (Elementar Analysensysteme, GmbH, Germany). The percentages of carbon (C), hydrogen (H) and nitrogen (N) were recorded. The conjugation efficiency was calculated based on the percentage of each element and the chemical structure of each sample. The particle hydrodynamic diameter (Z-average) and the zeta (f)-potential measurements of the nanoparticles were carried out by dynamic light scattering (DLS) using a Zetasizer Nano ZS (Malvern Instruments, UK) equipped with a He–Ne laser beam (633 nm, fixed scattering angle of 173°) at 25 °C. The reported data are presented as the average of three independent measurements, i.e., the data are an average of three Z-average size values. The standard deviations were calculated based on the three Z-average size values. The morphology of the nanoparticles was observed by transmission electron microscope (TEM) (Jeol, JEM-1400, Japan) with voltage of 80 kV and magnification between 250 and 10,000. The samples were prepared by adding one drop of nanoparticle suspension to a copper grid coated with carbon and dried overnight before imaging.

227

2.5. Cell uptake of nanoparticles

228

243

Endothelial EA.hy926 cells, which have low expression of FR, and brain tumor U87 MG cells, which have high expression of FR, were used to evaluate the cell uptake efficiency of MTX–PSi by TEM. About 2 mL/well of 5  105 cells/well of EA.hy926 cells or 6  105 cells/well of U87 MG were seeded in 6-well plates containing 18  18 mm cover slip (Menzel-Gläser, Braunschweig, Germany) in each well. After reaching 80% confluency, the medium was replaced by a new medium containing 100 lg/mL of each nanoparticle. After 2 h incubation, the nanoparticle suspensions were removed and the cells were washed 3 times with HBSS (pH 7.4). 1 mL/well of 2.5% glutaraldehyde was added to the cells and incubated at 37 °C for 20 min for cell fixation, followed by washing 3 times with HBSS. Ultrathin sections of both control and exposed cells to the nanoparticles were prepared as described elsewhere [45]. The TEM images were obtained with the same parameters as described above.

244

2.6. Drug loading and release

245

The poorly water-soluble sorafenib was loaded to the MTX–PSi nanoparticles for combination therapy. The PSi and MTX–PSi nanoparticles were immersed in 15 mg/mL of sorafenib dissolved in acetone solution with a ratio of 1 mg of PSi nanoparticles to 1 mL of drug in solution, and stirred for 2 h at room temperature. The drug-loaded nanoparticles were collected and washed once with Milli-Q water. The loading degrees were determined by immersing 200 lg of the dual drug-loaded nanoparticles to 1 mL of acetonitrile/water mixture (42:58%, v/v) under vigorous stirring for 1 h. The released MTX fragment from the MTX–PSi nanoparticles was confirmed by a Waters ACQUITY UPLCÒ (Waters, Massachusetts, MO, USA) coupled to a WaterÒ Xevo™ quadrupole time-of flight mass spectrometer (Q-TOF MS, Waters, Massachusetts, MO,). The release profiles of MTX from the MTX–PSi nanoparticles loaded with/without sorafenib were performed at 37 ± 1 °C in sink conditions using a shaking method with a shaking speed rate of 100 rpm. The release media used were 10 mM of 2-(N-morpholino)ethanesulfonic acid (MES, pH 5.5), which mimics the pH of the endosome inside the cells when the PSi–MTX are uptaken by the cells, and 10 mM HEPES (pH 7.4), which is the physiological

203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225

229 230 231 232 233 234 235 236 237 238 239 240 241 242

246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264

3

pH condition, with/without 10% FBS. 10% FBS was added to the media for the drug release studies in order to confirm two effects. Firstly, these studies evaluate whether the protein influences the MTX release from the PSi cargo. Secondly, based on our previous study [17], sorafenib can be dissolved very fast from the PSi nanoparticles in a buffer containing 10% FBS, but is not released from the nanoparticle in a buffer without FBS. In this study, we compared the MTX release from the PSi nanoparticles with/without loaded sorafenib to confirm whether sorafenib loading in MTX– PSi nanoparticles affected the release of MTX. The dissolution profiles of sorafenib-loaded in both PSi and MTX–PSi, or free sorafenib, were performed at pH 7.4 with 10% FBS, because the purpose of this study was to prove that the solubility of sorafenib was enhanced in physiological conditions. 200 lL of samples was withdrawn from each dissolution test at predetermined time points. The collected samples were centrifuged at 13,400 rcf for 3 min and the supernatant was analyzed by HPLC as described below. The amounts of MTX and sorafenib released from the PSi nanoparticles were determined by HPLC (Agilent 1260, Agilent Technologies, USA). The chromatographic separation was achieved using a Zorbax C18 (4.6  100 mm, 5 lm) column. For MTX, the mobile phase used was composed of 90% of aqueous phase (0.1 M citric acid and 0.2 M Na2HPO4 mixed at a ratio of 2:1, pH 6.0) and 10% of acetonitrile with flow rate of 1 mL/min, while a UV detector was set at the wavelength of 302 nm. For sorafenib, the mobile phase was composed of 0.2% trifluoroacetic acid (pH 2) and acetonitrile at a ratio of 42:58% (v/v) with 1.0 mL/min flow rate and UV detector set at the wavelength of 254 nm.

265

2.7. Anti-proliferation activity

293

The in vitro anti-proliferation effect of the MTX–PSi nanoparticles against the cancer cells was evaluated by cell proliferation experiments. Briefly, EA.hy926 and U87 MG cells were seeded in 96-well plates at the density of 1  104 cells/well and 1.5  104 cells/well, respectively, and allowed to attach overnight. Then, the cell culture medium was replaced by 100 lL of medium containing different concentrations of MTX and MTX–PSi nanoparticles. After 24 and 48 h incubation, the amount of living cells was determined by the CellTiter GloÒ luminescence cell viability assay kit. Each experiment was performed at least in triplicate.

294

2.8. MTX–PSi induced cell apoptosis

304

MTX induces cell death by inhibiting the activity of DHFR, and thus, inhibiting the DNA biosynthesis. In order to confirm the bioactivity of the released MTX fragments from the MTX–PSi, 7amino-actinomycin D (7-AAD, BioLegnadÒ) labeling DNA fragmentation programed cell death experiment was performed by flow cytometry analysis. Briefly, 2  105 cells/mL/well of each cell line was seeded in 12-well plates separately. After reaching 80% confluency, the medium was replaced by a new medium containing certain concentrations of both PSi and MTX–PSi nanoparticles for 24 h incubation. After removing the nanoparticle solutions and washing the cells 3 times with HBSS (pH 7.4) to remove non-adherent nanoparticles, the cells were harvested by trypsin-ethylenediaminetetraacetic acid and kept on ice. Cells were then re-suspended in 100 lL BioLegand’s Cell Staining Buffer and 5 lL of 7-AAD (50 lg/mL) was added to each sample and incubated for 15 min at room temperature in the dark. After that, 400 lL of HBSS buffer was added and the flow cytometry experiments were performed with a LSR II flow cytometer (BD Biosciences, USA) using a FACSDiva software and a laser excitation wavelength of 488 nm. 10,000 events were collected for each sample and the data were analyzed by FACSDiva software.

305

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

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

295 296 297 298 299 300 301 302 303

306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 4

C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

326

2.9. Statistical analysis

327

Results of the assays were expressed as mean ± s.d. of at least three independent experiments. Student’s t-test was used to evaluate the significant differences and set at probabilities of ⁄⁄p < 0.01 and ⁄⁄⁄p < 0.001 using Origin 8.6 (OriginLab Corp., USA).

328 329 330

331

3. Results and discussion

332

3.1. Characterization of MTX–PSi nanoparticles

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

Nitrogen sorption results indicated that the amine-terminated PSi nanoparticles retained a porous structure according to the shape of the isotherm. The calculated specific surface area was 301 ± 16 m2/g with a pore volume of 0.60 ± 0.01 cm3/g, and the average pore diameter of 8.0 ± 0.4 nm. The zeta (f)-potential of the PSi nanoparticles obtained from electrophoretic mobility measurements was +40.6 mV. The conjugation of MTX to PSi was based on the NHS/EDC coupling reaction between the carboxylic acid group of MTX and the free amine groups of the PSi nanoparticles to form an amide bond (Scheme 1). According to ATR–FTIR (Fig. 1a), the exhibition of two characteristic bands of MTX–PSi corresponded to m(NC@O) (amide I) and d(CNAH) (amide II) vibrations at 1650 and 1540 cm1, respectively, which indicated the formation of an amide bond between MTX and the nanoparticle. The strong peak showed at 1610 cm1 represented the typical NAH bending vibrations of primary amine in MTX. The peak 1450 cm1 was assigned to CH2 bending of MTX. All these indicated that MTX was conjugated to PSi nanoparticles. From the MS measurements of the MTX release solutions, the found compound was MTX attached with fragment 3-aminopropylesilicic acid moiety (Scheme 1D and Fig. S1). There was no free MTX remaining in the MTX–PSi conjugation. Further quantification of the conjugation ratio was accessed by elemental analysis (Table S1) and calculated based on the increasing mass percentage of each element resulting from the conjugation related to the molecular Q5 formula of MTX and the surface chemistry of PSi. About 8.4 ± 1.6% (molar ratio) of the amine group in PSi was reacted with MTX, and thus, the mass loading of MTX to PSi was 5 lg/1 mg of nanoparticles.

The nanoparticle size and f-potential of both PSi and MTX–PSi were determined by DLS (Table 1 and Fig. 1B). The nanoparticles used in this work exhibited a nanosize distribution and free dispersibility in aqueous solutions with low polydispersity indices (PDI). The f-potential of MTX–PSi was slightly lower than that of bare PSi nanoparticles. The pKa-values of MTX are 4.8 and 5.5 [46], which present a slightly negative charge compared to the reacted amine groups on the surface of PSi at neutral conditions. Fig. 1C and D shows the morphologies of PSi and MTX–PSi nanoparticles obtained by TEM with an overview and closer observation (inside images). This result was consistent with our previous work on the PSi nanoparticle surface modification with peptides [17]. The porous structure of the MTX–PSi nanoparticles can be accessed for further drug loading.

363

3.2. Cell uptake of MTX–PSi nanoparticles

377

FR is widely over-expressed by a number of cancer cells. MTX has high affinity to FR, which can form FR-mediated cell uptake [24]. It has been demonstrated that MTX conjugated to dendrimers through amide linkage showed more than 4000-times higher affinity to FR than free MTX, and also multi-valent conjugate to one single dendrimer molecule showed a much higher affinity to FR than the free MTX to FR [31]. Thus, we hypothesize that MTX conjugated to PSi can increase the cellular uptake of the PSi nanoparticles in the high FR-expressing cells. On the other hand, the nanoparticle’s positive surface charge is beneficial for the non-specific cell internalization of the nanoparticles [47,48]. Cells with low FR-expression have very low cell uptake of MTX [49]. By conjugation of MTX to positively charged nanoparticles, the cell uptake of MTX could be increased [20]. Furthermore, the charge-based non-specific cell internalization can help to overcome the drug resistance of MTX. In order to evaluate the aforementioned capacity with our systems, PSi and MTX–PSi nanoparticles were incubated with the two cancer cells to determine the cell uptake efficacy. One cell line used was the hybrid endothelial EA.hy926 with a low expression of FR. The other cell line used was the human glioblastoma U87 MG, which highly over-expresses the FR. Fig. 2 shows qualitative TEM images of the PSi and MTX–PSi cell uptake by the EA.hy926 and U87 MG cells. MTX–PSi nanoparticles were uptaken by low FR

378

Scheme 1. (A) MTX-conjugated to PSi by NHS/EDC coupling reaction to form MTX–PSi. (B) Molecular structure of MTX. (C) Released fragment from MTX–PSi. (D) Dehydrate form containing MTX determined by MS.

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

364 365 366 367 368 369 370 371 372 373 374 375 376

379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 5

C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

Fig. 1. (A) ATR–FTIR of PSi and MTX–PSi. (B) Particle size distribution of PSi and MTX–PSi based on intensity values determined by DLS in Milli-Q water. (C and D) TEM pictures of PSi and MTX–PSi nanoparticles showing their characteristic morphology. Scale bars are 500 nm for the overview image and 200 nm for the insert images.

Table 1 Physiochemical characterization of the Z-average size, f-potential (in Milli-Q water) and drug loading degree of PSi and MTX–PSi. Data represent the mean ± s.d. of at least three independent measurements. The reported data are presented as the average of three independent measurements, i.e., the data are an average of three Z-average size values. The standard deviations were calculated based on the three Z-average size values. Sample

Z-average size (nm)

PDI

f-potential (mV)

Loading degree of sorafenib (w-%)

PSi MTX–PSi

177.0 ± 1.8 200.0 ± 1.7

0.106 ± 0.007 0.118 ± 0.05

+40.6 ± 1.5 +38.5 ± 1.9

6.1 ± 0.7 6.0 ± 0.6

Fig. 2. Intracellular uptake and distribution of the PSi and MTX–PSi nanoparticles in EA.hy926 and U87 MG cells. TEM images of ultrathin sections of EA.hy926 and U87 MG cells were used as controls or incubated with PSi and MTX–PSi nanoparticles at 37 °C (Inset scale bars 200 nm).

402 403 404 405 406 407

expressed EA.hy926 cells, although there were less MTX–PSi nanoparticles located intracellularly than of the bare PSi nanoparticles. Amine-terminals on the surface of PSi enhance cellular uptake due to the positive charge interaction with the negative cell membrane and thus induce cellular endotheliosis [20]. The presence of MTX on the surface of PSi normalized the non-specific cell uptake of

MTX–PSi nanoparticles, and thus, led to a lower cell uptake. For the FR over-expressed U87 MG cells, there were more MTX–PSi nanoparticles located and distributed in the cytoplasm of the cells than with the bare PSi nanoparticles. This confirmed our hypothesis that MTX conjugation can increase the FR-mediated cell uptake by FR over-expressed cancer cells.

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

408 409 410 411 412 413

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 6

C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

414

3.3. Drug loading and release

415

MTX is a folic acid analog cancer drug which acts as an inhibitor of DHFR to stop the tetrahydrofolate synthesis and causes cell death [22]. The conjugation of MTX to PSi nanoparticles can increase the affinity to FR. However, DHFR inhibition using MTXdendrimer conjugates has been shown to be comparatively less active to DHFR than free MTX. This was presumably because only one MTX molecule on each dendrimer could bind to DHFR molecules [31]. From another report of MTX conjugated to carbon nanotubes (CNT), in vivo anticancer activities of the ester-linked MTX–CNT conjugates were significantly higher than the amidelinked counterpart [39]. This result suggested that the cleavability of linkers between MTX and the carrier was crucially determining the therapeutic performance of the conjugated MTX. Thus, our hypothesis is that the release of MTX from the PSi nanoparticles would be essential for increasing the efficacy of MTX in the developed cancer drug formulation. The aminosilane surface moiety can be hydrolyzed and detached from the PSi surface in an aqueous solution [19]. Thus the MTX fragment can be released from the PSi nanoparticles through hydrolysis. The released compound from the MTX–PSi was first examined by MS analysis. The main detected molecule by MS was MTX with a fragment of 3-aminopropylesilicic acid (Scheme 1D, Mw 555.2 g/mol; the compound was detected as the protonated molecule ([M+H]+, m/z 556.2)). The results confirmed that MTX with a fragment of (3-aminopropyl)trihydroxylsilane (Scheme 1C) was released from MTX–PSi, and then dehydrolyzed to the molecule presented in Scheme 1D. Next, the drug release profiles against time were analyzed by HPLC. Fig. 3A–C shows a sustained release of MTX from the MTX–PSi nanoparticles. MTX was released faster at pH 7.4 than at pH 5.5, due to the surface silane moiety that was hydrolyzed and detached from PSi faster at higher pH-values [19]. The MTX release profiles from MTX–PSi in buffer without FBS (Fig. 3B) and

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

from MTX–PSi loaded with sorafenib in buffer with FBS (Fig. 3C) were similar to the release profiles of MTX–PSi in buffer with 10% FBS at both the pH-values (Fig. 3A). This was consistent with the results that the release of MTX was due to the cleavage of the aminosilane moiety from the PSi surface. This cleavage was not affected by serum proteins or by the loaded sorafenib in the PSi nanoparticles. During the first 12 h, there was approximately a linear sustained release of MTX in all the release conditions. Over 40% (pH 7.4) and 12% (pH 5.5) of MTX were released from the MTX–PSi nanoparticles during the first 12 h, followed by a sustained release up to 96 h, where the concentration of MTX reached 0.65 and 0.92 lg/mL at pH 5.5 and 7.4, respectively (Fig. 3B). In addition to the MTX conjugation to the PSi surface, another anti-angiogenic hydrophobic drug, sorafenib, was loaded into the MTX–PSi nanoparticles to explore potential combination therapy applications. After drug loading, 200 lg of each sorafenib loaded nanoparticle was resuspended in 1 mL of 48% mixture of ACN/ water solution and the supernatant was analyzed by HPLC. The concentrations were 12.9 and 12.7 lg/mL of sorafenib for PSi and MTX–PSi nanoparticles, respectively. The drug loading studies of sorafenib in PSi and MTX–PSi showed that the MTX conjugation to PSi did not affect the loading capacity of PSi nanoparticles (Table 1), leading to similar loading degrees of sorafenib both in the presence or absence of MTX. The dissolution rate of sorafenib in aqueous solution was, however, considerably enhanced by loading it into the both PSi nanoparticles (Fig. 4A and B). The solubility of sorafenib in 10% FBS aqueous buffer was 8.8 lg/mL. The dissolution studies were done in sink-conditions [17]. The concentration of sorafenib was 0.43 and 0.42 lg/mL after 8 h for PSi and MTX–PSi nanoparticles, respectively while only 0.03 lg/ mL of pure sorafenib was dissolved in the same condition at 8 h time point. These results were consistent with our previous work Q6 of peptides modification of PSi nanoparticles for drug delivery [17].

Fig. 3. Release profiles of MTX from MTX–PSi nanoparticles. (A) MTX release from MTX–PSi in buffer containing 10% FBS. (B) MTX release from MTX–PSi in buffer without FBS. (C) MTX release from sorafenib-loaded MTX–PSi nanoparticles in buffer containing 10% FBS.

Fig. 4. Dissolution profiles of sorafenib. (A) Sorafenib released from PSi and MTX–PSi nanoparticles in buffer (pH 7.4) containing 10% FBS, and (B) free sorafenib dissolution profile in buffer containing (pH 7.4) 10% FBS.

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

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

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

7

Fig. 5. Cancer cell growth inhibition by treatment with pure MTX, PSi and MTX–PSi nanoparticles. The nanoparticles were suspended in full cell media. Cells were exposed to pure MTX, PSi and MTX–PSi nanoparticles for 24 (A: EA.hy926, C: U87 MG) and 48 h (B: EA.hy926, D: U87 MG) at 37 °C. Pure MTX was dissolved into DMSO (2 mg/mL). The cell viability of 0.1% of DMSO for EA.hy926 and U87 MG cells at 24 and 48 h is presented in Fig. S2.

Fig. 6. MTX–PSi nanoparticles inducing programed cell apoptosis. EA.hy926 and U87 MG cells were incubated with PSi and MTX–PSi nanoparticles for 24 h. After washing and harvesting, the cells were stained with 7-AAD for 15 min, followed by flow cytometry analysis to determine the percentage of DNA fragmentation in the samples. The MTX concentrations used were 0.1 and 0.5 lg/mL, which corresponded to the concentrations of 20 and 100 lg/mL for bare PSi and MTX–PSi nanoparticles.

481

3.4. Inhibition of cancer cell proliferation

482

In order to study the anti-proliferation efficacy of the MTX–PSi nanoparticles, EA.hy926 and U87 MG cells were exposed to free MTX or MTX–PSi with concentrations from 0.01 to 2 lg/mL for predetermined time, and the cells were quantified by an ATP-based cell proliferation assay (Fig. 5). Bare PSi nanoparticles at the same concentrations as the MTX–PSi nanoparticles were used as a control. At both 24 and 48 h incubation of MTX with EA.hy926, a minor effect on the cell proliferation was observed. Contrarily, MTX–PSi significantly inhibited the growth of EA.hy926 cells in a concentration-dependent manner, while the PSi nanoparticles had a modest effect on the cell growth (Fig. 5A and B). For the low folic acid receptor expressing EA.hy926 cells, free MTX had no effect on the cells up to concentrations of 2 lg/mL. However, after conjugation to the PSi nanoparticles, the half maximal inhibitory concentration (IC50) of MTX in EA.hy926 cells was 0.5 lg/mL at 24 h and 1.0 lg/mL at 48 h, respectively. This higher inhibition efficiency of MTX–PSi on the cell proliferation was probably due to the cationic surface of the nanoparticles enhancing the cellular uptake of MTX–PSi. After internalization, MTX was released from the MTX–PSi nanoparticles to the cytosol, inducing a higher cell death. Since free MTX had low cell uptake in EA.hy926 cells, its effect on cell proliferation was minimal.

483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503

In the case of U87 MG cells, which highly express the FR, MTX can be internalized into the cells by FR-mediated cell uptake. The results showed that MTX successfully inhibited the U87 MG cell proliferation at both 24 and 48 h. MTX–PSi had a lower efficiency on the proliferation inhibition of U87 MG at concentrations lower than 0.5 lg/mL; however, at higher concentrations, MTX– PSi had comparable efficacy on the cell proliferation as free MTX, whereas the cell viability was close to the background levels (Fig. 5C and D). The IC50 of MTX in U87 cells was ca. 0.05 lg/mL at both 24 and 48 h incubation times. The IC50 of MTX–PSi was 0.5 lg/mL (24 h) and 0.25 lg/mL (48 h). The IC50 was increased when MTX was conjugated to the PSi nanoparticles. This might be due to the sustained release of MTX from MTX–PSi nanoparticles; there was a delay of MTX to inhibit the activity of DHFR enzyme.

504

3.5. MTX-induced cell apoptosis

519

During the interphase of cell proliferation, the enzyme DHFR catalyzes the dihydrofolate to tetrahydrofolate for thymidylate synthesis intended for DNA duplication [50]. MTX can inhibit the activity of DHFR in the cytosol, resulting in inhibition of thymidylate synthesis and consequently stopping the DNA biosynthesis and leading to cell apoptosis [27]. To ensure that the cell death

520

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

505 506 507 508 509 510 511 512 513 514 515 516 517 518

521 522 523 524 525

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 8

C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx

541

determined in cell proliferation experiments was due to the MTX-induced cell apoptosis, DNA fragmentation was accessed by 7-AAD stained and flow cytometry analysis. The percentage of the cells with DNA segmentation was measured for both EA.hy926 and U87 MG cells treated with PSi and MTX–PSi for 24 h (Fig. 6). In both cells, MTX–PSi caused a concentration-dependent DNA fragmentation. When treated with 0.1 lg/mL of MTX contained MTX–PSi nanoparticles, there was 25.7% and 11.2% of nuclear fragmentation for EA.hy926 and U87 MG cells, respectively. At a higher concentration (0.5 lg/mL), 80.8% and 72.8% for the EA.hy926 and U87 MG cells induced nuclear segmentation, respectively. Importantly, for both cell lines, the bare PSi nanoparticles did not cause a significant nuclear segmentation. These results demonstrated that MTX–PSi nanoparticles were up-taken by the cells and the biological activity of the released MTX was maintained.

542

4. Conclusion

543

568

PSi nanoparticles are versatile biomaterials with a nano-porous structure and large surface areas that can be used for drug loading and enhanced dissolution rate of poorly-water soluble drugs. MTX is a folate analog anticancer drug, which acts by inhibiting DHFR enzymatic activity in cytosol to induce cell death. In this work, MTX was successfully conjugated to amine-terminated PSi nanoparticles to obtain a non-specific cell uptake and to overcome potential drug resistance. The MTX conjugation did not affect the drug loading capacity of the PSi nanosystem. The hydrophobic anti-angiogenic drug, sorafenib, was successfully loaded to the MTX–PSi nanoparticles. The dissolution rate of this poorly-water soluble drug was dramatically enhanced due to the porous structure of the PSi nanosystem. Furthermore, MTX had a prolonged release from the MTX–PSi nanoparticles as a result of the covalent attachment to the surface of the PSi. The MTX release was due to the cleavage of 3-aminopropylsilane moiety from the surface of PSi nanoparticles. The cell viability showed that the MTX–PSi nanoparticles successfully inhibited the cell proliferation of both FR-over-expressed and low FR-expressing cells due to the induced MTX programed cell apoptosis. With this combination, fast released sorafenib can be expected to inhibit the neovascularization of the tumor in the tumor environment, while the improved cellular uptake of MTX–PSi nanoparticles and sustained release of chemotherapeutical MTX can inhibit the tumor cell proliferation for prolonged dosing times for cancer therapy.

569

Conflict of interest

526 527 528 529 530 531 532 533 534 535 536 537 538 539 540

544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567

570

571 572 573 574 575 576 577 578 579 580 581 582 583

The authors declare no competing financial interest. Acknowledgements C.-F. Wang acknowledges financial support from the Chinese Q7 Scholarship Council (Grant No. 2009627022). H.A. Santos acknowlQ8 edges the Academy of Finland (Decision nos. 252215 and 256394), the University of Helsinki Funds, Biocentrum Helsinki, and the European Research Council under the European Union’s Seventh Framework Programme (FP/2007–2013, Grant No. 310892) for financial support. We thank Dr. Anu Airaksinen and Dr. Mirkka Sarparanta (Department of Chemistry, University of Helsinki) for their kind help with the elemental analysis, and Dr. Pirjo Laakkonen (Research Programs Unit, Translational Cancer Biology and Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki,) for kindly provided the U87MG cells.

Appendix A. Supplementary data

584

Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.actbio.2015.01. 021.

585

References

588

[1] van der Meel R, Vehmeijer LJ, Kok RJ, Storm G, van Gaal EV. Ligand-targeted particulate nanomedicines undergoing clinical evaluation: current status. Adv Drug Deliv Rev 2013;65:1284–98. [2] Shahbazi MA, Hamidi M, Makila EM, Zhang H, Almeida PV, Kaasalainen M, et al. The mechanisms of surface chemistry effects of mesoporous silicon nanoparticles on immunotoxicity and biocompatibility. Biomaterials 2013;34:7776–89. [3] Bimbo LM, Denisova OV, Makila E, Kaasalainen M, De Brabander JK, Hirvonen J, et al. Inhibition of influenza a virus infection in vitro by saliphenylhalamideloaded porous silicon nanoparticles. ACS Nano 2013;7:6884–93. [4] Bimbo LM, Makila E, Raula J, Laaksonen T, Laaksonen P, Strommer K, et al. Functional hydrophobin-coating of thermally hydrocarbonized porous silicon microparticles. Biomaterials 2011;32:9089–99. [5] Liu D, Bimbo LM, Makila E, Villanova F, Kaasalainen M, Herranz-Blanco B, et al. Co-delivery of a hydrophobic small molecule and a hydrophilic peptide by porous silicon nanoparticles. J Control Release 2013;170:268–78. [6] Sarparanta MP, Bimbo LM, Makila EM, Salonen JJ, Laaksonen PH, Helariutta AM, et al. The mucoadhesive and gastroretentive properties of hydrophobincoated porous silicon nanoparticle oral drug delivery systems. Biomaterials 2012;33:3353–62. [7] Santos HA, Riikonen J, Salonen J, Makila E, Heikkila T, Laaksonen T, et al. In vitro cytotoxicity of porous silicon microparticles: effect of the particle concentration, surface chemistry and size. Acta Biomater 2010;6:2721–31. [8] Mann AP, Tanaka T, Somasunderam A, Liu X, Gorenstein DG, Ferrari M. Eselectin-targeted porous silicon particle for nanoparticle delivery to the bone marrow. Adv Mater 2011;23:H278–82. [9] Godin B, Gu J, Serda RE, Ferrati S, Liu X, Chiappini C, et al. Multistage mesoporous silicon-based nanocarriers: biocompatibility with immune cells and controlled degradation in physiological fluids. Control Release Newsl. 2008;25:9–11. [10] Tanaka T, Mangala LS, Vivas-Mejia PE, Nieves-Alicea R, Mann AP, Mora E, et al. Sustained small interfering RNA delivery by mesoporous silicon particles. Cancer Res 2010;70:3687–96. [11] von Maltzahn G, Park JH, Lin KY, Singh N, Schwoppe C, Mesters R, et al. Nanoparticles that communicate in vivo to amplify tumour targeting. Nat Mater 2011;10:545–52. [12] Park JH, Gu L, von Maltzahn G, Ruoslahti E, Bhatia SN, Sailor MJ. Biodegradable luminescent porous silicon nanoparticles for in vivo applications. Nat Mater 2009;8:331–6. [13] Salonen J, Laitinen L, Kaukonen AM, Tuura J, Bjorkqvist M, Heikkila T, et al. Mesoporous silicon microparticles for oral drug delivery: loading and release of five model drugs. J Control Release 2005;108:362–74. [14] Santos HA, Bimbo LM, Lehto VP, Airaksinen AJ, Salonen J, Hirvonen J. Multifunctional porous silicon for therapeutic drug delivery and imaging. Curr Drug Discov Technol 2011;8:228–49. [15] Jarvis KL, Barnes TJ, Prestidge CA. Surface chemical modification to control molecular interactions with porous silicon. J Colloid Interface Sci 2011;363:327–33. [16] Kinnari PJ, Hyvonen ML, Makila EM, Kaasalainen MH, Rivinoja A, Salonen JJ, et al. Tumour homing peptide-functionalized porous silicon nanovectors for cancer therapy. Biomaterials 2013;34:9134–41. [17] Wang CF, Makila EM, Kaasalainen MH, Liu D, Sarparanta MP, Airaksinen AJ, et al. Copper-free azide-alkyne cycloaddition of targeting peptides to porous silicon nanoparticles for intracellular drug uptake. Biomaterials 2014;35:1257–66. [18] Wu EC, Andrew JS, Cheng L, Freeman WR, Pearson L, Sailor MJ. Real-time monitoring of sustained drug release using the optical properties of porous silicon photonic crystal particles. Biomaterials 2011;32:1957–66. [19] Makila E, Bimbo LM, Kaasalainen M, Herranz B, Airaksinen AJ, Heinonen M, et al. Amine modification of thermally carbonized porous silicon with silane coupling chemistry. Langmuir 2012;28:14045–54. [20] Ryser HJ, Shen WC. Conjugation of methotrexate to poly(L-lysine) increases drug transport and overcomes drug resistance in cultured cells. Proc Natl Acad Sci USA 1978;75:3867–70. [21] Jiang T, Li YM, Lv Y, Cheng YJ, He F, Zhuo RX. Amphiphilic polycarbonate conjugates of doxorubicin with pH-sensitive hydrazone linker for controlled release. Colloids Surf B Biointerfaces 2013;10:542–8. [22] Gonen N, Assaraf YG. Antifolates in cancer therapy: structure, activity and mechanisms of drug resistance. Drug Resist Updat 2012;15:183–210. [23] Neradil J, Pavlasova G, Veselska R. New mechanisms for an old drug; DHFRand non-DHFR-mediated effects of methotrexate in cancer cells. Klin Onkol 2012;25:2S87–92. [24] Barton AK, Antoieta C. Receptor-mediated folate accumulation is regulated by the cellular folate content.pdf. Proc Natl Acad Sci USA 1986;83:5983–7. [25] Zwicke GL, Mansoori GA, Jeffery CJ. Utilizing the folate receptor for active targeting of cancer nanotherapeutics. Nano Rev 2012;3:18496.

589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

586 587

ACTBIO 3560

No. of Pages 9, Model 5G

28 January 2015 C.-F. Wang et al. / Acta Biomaterialia xxx (2015) xxx–xxx 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705

[26] Parker N, Turk MJ, Westrick E, Lewis JD, Low PS, Leamon CP. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem 2005;338:284–93. [27] Bertino JR, Göker E, Gorlick R, Li WW, Banerjee D. Resistance mechanisms to methotrexate in tumors. Stem Cells 1996;14:5–9. [28] Chen J, Huang L, Lai H, Lu C, Fang M, Zhang Q, et al. Methotrexate-loaded PEGylated chitosan nanoparticles: synthesis, characterization, and in vitro and in vivo antitumoral activity. Mol Pharm 2014;11:2213–23. [29] Gurdag S, Khandare J, Stapels S, Matherly LH, Kannan RM. Activity of dendrimer–methotrexate conjugates on methotrexate-sensitive and resistant cell lines. Bioconjug Chem 2006;17:275–83. [30] Kaminskas LM, Kelly BD, McLeod VM, Boyd BJ, Krippner GY, Williams ED, et al. Pharmacokinetics and tumor disposition of PEGylated, methotrexate conjugated poly-L-lysine dendrimers. Mol Pharm 2009;6:1190–204. [31] Thomas TP, Huang B, Choi SK, Silpe JE, Kotlyar A, Desai AM, et al. Polyvalent dendrimer–methotrexate as a folate receptor-targeted cancer therapeutic. Mol Pharm 2012;9:2669–76. [32] Huang B, Otis J, Joice M, Kotlyar A, Thomas TP. PSMA-targeted stably-linked ‘dendrimer-glutamate urea-methotrexate’ as a prostate cancer therapeutic. Biomacromolecules 2014;15:915–23. [33] Kaminskas LM, Kelly BD, McLeod VM, Sberna G, Boyd BJ, Owen DJ, et al. Capping methotrexate alpha-carboxyl groups enhances systemic exposure and retains the cytotoxicity of drug conjugated PEGylated polylysine dendrimers. Mol Pharm 2011;8:338–49. [34] Majumdar S, Anderson ME, Xu CR, Yakovleva TV, Gu LC, Malefyt TR, et al. Methotrexate (MTX)-cIBR conjugate for targeting MTX to leukocytes: conjugate stability and in vivo efficacy in suppressing rheumatoid arthritis. J Pharm Sci 2012;101:3275–91. [35] Pica K, Tchao R, Ofner 3rd CM. Gelatin-methotrexate conjugate microspheres as a potential drug delivery system. J Pharm Sci 2006;95:1896–908. [36] Taheri A, Dinarvand R, Ahadi F, Khorramizadeh MR, Atyabi F. The in vivo antitumor activity of LHRH targeted methotrexate-human serum albumin nanoparticles in 4T1 tumor-bearing Balb/c mice. Int J Pharm 2012;431:183–9. [37] Kohler N, Sun C, Fichtenholtz A, Gunn J, Fang C, Zhang M. Methotrexateimmobilized poly(ethylene glycol) magnetic nanoparticles for MR imaging and drug delivery. Small 2006;2:785–92. [38] Choi Y, Kim M, Cho Y, Yun E, Song R. Synthesis, characterization and target protein binding of drug-conjugated quantum dots in vitro and in living cells. Nanotechnology 2013;24:075101. [39] Das M, Datir SR, Singh RP, Jain S. Augmented anticancer activity of a targeted, intracellularly activatable, theranostic nanomedicine based on fluorescent and

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

9

radiolabeled, methotrexate-folic acid-multiwalled carbon nanotube conjugate. Mol Pharm 2013;10:2543–57. Rosenholm JM, Peuhu E, Bate-Eya LT, Eriksson JE, Sahlgren C, Linden M. Cancer-cell-specific induction of apoptosis using mesoporous silica nanoparticles as drug-delivery vectors. Small 2010;6:1234–41. Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, et al. BAY 439006 exhibits broad spectrum oral antitumor activity and targets the RAF/ MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 2004;64:7099–109. Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY, et al. Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol J Am Soc Clin Oncol 2010;28:1856–62. Moore M, Hirte HW, Siu L, Oza A, Hotte SJ, Petrenciuc O, et al. Phase I study to determine the safety and pharmacokinetics of the novel Raf kinase and VEGFR inhibitor BAY 43-9006, administered for 28 days on/7 days off in patients with advanced, refractory solid tumors. Ann Oncol 2005;16:1688–94. Strumberg D, Clark JW, Awada A, Moore MJ, Richly H, Hendlisz A, et al. Safety, pharmacokinetics, and preliminary antitumor activity of sorafenib: a review of four phase I trials in patients with advanced refractory solid tumors. Oncologist 2007;12:426–37. Bimbo LM, Makila E, Laaksonen T, Lehto VP, Salonen J, Hirvonen J, et al. Drug permeation across intestinal epithelial cells using porous silicon nanoparticles. Biomaterials 2011;32:2625–33. Cannon WR, Garrison BJ, Benkovic SJ. Consideration of the pH-dependent inhibition of dihydrofolate reductase by methotrexate. J Mol Biol 1997;271:656–68. Gratton SE, Ropp PA, Pohlhaus PD, Luft JC, Madden VJ, Napier ME, et al. The effect of particle design on cellular internalization pathways. Proc Natl Acad Sci USA 2008;105:11613–8. Krasnici S, Werner A, Eichhorn ME, Schmitt-Sody M, Pahernik SA, Sauer B, et al. Effect of the surface charge of liposomes on their uptake by angiogenic tumor vessels. Int J Cancer 2003;105:561–7. Kane MA, Portillo RM, Elwood PC, Antony AC, Kolhouse JF. The influence of extracellular folate concentration on methotrexate uptake by human KB cells. Partial characterization of a membrane-associated methotrexate binding protein. J Biol Chem 1986;261:44–9. Rajagopalan PT, Zhang Z, McCourt L, Dwyer M, Benkovic SJ, Hammes GG. Interaction of dihydrofolate reductase with methotrexate: ensemble and single-molecule kinetics. Proc Natl Acad Sci USA 2002;99:13481–6.

Please cite this article in press as: Wang C-F et al. Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and

Q1 combination therapy. Acta Biomater (2015), http://dx.doi.org/10.1016/j.actbio.2015.01.021

706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746

Dual-drug delivery by porous silicon nanoparticles for improved cellular uptake, sustained release, and combination therapy.

Dual-drug delivery of antiangiogenic and chemotherapeutic drugs can enhance the therapeutic effect for cancer therapy. Conjugation of methotrexate (MT...
2MB Sizes 0 Downloads 8 Views