Nanoemulsion loaded gel for topical co-delivery of ...

67 downloads 57 Views 1MB Size Report
Imiquimod. 14 induced psoriatic BALB/c mice revealed significantly higher anti-psoriatic activity of nanoemulsion gel as compared to free drugs and. 15.
NANO-01531; No of Pages 10

Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx – xxx nanomedjournal.com

Q33Q2

Amanpreet Kaur, Sameer S. Katiyar, Varun Kushwah, Sanyog Jain⁎

6

Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, SAS Nagar, Punjab, India Received 8 July 2016; accepted 9 February 2017

R O O

4 5

F

2

Nanoemulsion loaded gel for topical co-delivery of clobitasol propionate and calcipotriol in psoriasis

1Q1

Abstract

8

16

Current work reports the development and optimization of clobitasol propionate (CP) and calcipotriol (CT) loaded nanoemulsion based gel for topical treatment of psoriasis. Components of nanoemulsion viz., oil and surfactant/co-surfactant were selected depending upon solubility and emulsification potential respectively. The optimized ratio of 5:3:2 of Capmul MCM C8 EP, Cremophor RH 40 and Labrafil 1944 CS was selected. Carbopol 980 was used as gelling agent to achieve final drug concentration of 0.05% w/w and 0.005% w/w respectively for CP and CT. HaCaT cell lines showed higher uptake of drug from nanoemulsion in correlation with the enhancement in penetration of both drugs in stratum corneum (SC) and viable layer from nanoemulsion and gel as compared to free drugs. Imiquimod induced psoriatic BALB/c mice revealed significantly higher anti-psoriatic activity of nanoemulsion gel as compared to free drugs and marketed formulation. The developed formulation showed negligible skin irritation despite increased penetration into the skin. © 2017 Elsevier Inc. All rights reserved.

17

Key words: Nanoemulsion; Psoriasis; Higher permeation; Efficacy

13 14 15

D

12

E

11

25 26 27 28 29 30 31 32

R

R

24

O

23

C

22

N

21

Psoriasis is a chronic autoimmune inflammatory disease, affecting 2%–5% of world population, characterized by macules and plaques at the skin due to hyperproliferation and abnormal keratinocyte differentiation. 1–4 The disease mainly occurs in late childhood or early adulthood (b40 years of age) and persists for lifetime. 5 The disease sententiously affects the quality of life of patients with marked depression and suicidal contemplations leading to high mortality rates. 5,6 Topical therapy is the principal treatment for psoriasis, being employed as monotherapy in mild psoriasis and in adjunct to phototherapy, systemic therapy and biological agents in moderate to severe cases. 7 Topical treatment strategies including corticosteroids like clobitasol propionate (CP) (0.05% w/w) and vitamin D analogues like calcipotriol (CT) (0.005% w/w) are

U

20

E

18 19

T

10

C

9

P

7

well reported in the literature, wherein their combination is even reported as the first choice of treatment for plaque psoriasis requiring once daily application which makes it better than individual therapy of both the drugs. 8–10 CP is a highly potent steroid with rapid and prolonged anti-inflammatory action as it inhibits hyperproliferation in skin by suppressing DNA synthesizing cells. 11–13 The efficacy of CT involves suppression of epidermal proliferation and differentiation due to inhibition of dendritic cells as well as IL-2 and IL-6 production by epidermal T cells in psoriatic lesions. 14–16 CT is often associated with high incidence of skin irritation that can be masked by the use of topical corticosteroids. 16 The psoriatic skin being rough and embedded with plaques paves the major barrier in topical delivery of drugs in psoriasis. 17

Conflict of interest and disclosure: The authors report no financial interest that might pose a potential, perceived, or real conflict. All sources of support for research: The authors are thankful to the Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India and Director NIPER S.A.S. Nagar for providing fellowship and funding for the proposed work. The authors also want to express their thanks to NIPER S.A.S. Nagar for providing necessary benefits. Varun Kushwah is thankful to CSIR, Govt. of India for providing fellowship. The authors also thank Symbiotec Pharmalab, India for providing gift sample of CP and Abitech Corporation, Janesville, USA and Gattefosse, France for providing gift sample of lipids utilized in development of formulation. ⁎Corresponding author. E-mail addresses: [email protected], [email protected] (S. Jain). http://dx.doi.org/10.1016/j.nano.2017.02.009 1549-9634/© 2017 Elsevier Inc. All rights reserved. Please cite this article as: Kaur A., et al., Nanoemulsion loaded gel for topical co-delivery of clobitasol propionate and calcipotriol in psoriasis. Nanomedicine: NBM 2017;xx:1-9, http://dx.doi.org/10.1016/j.nano.2017.02.009

33 34 35 36 37 38 39 40 41 42 43 44 45 46

49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67

Topical delivery systems offer numerous advantageous due to localized action and minimization of systemic side effects. 18 Novel colloidal carriers are potential cargoes for delivering drugs to desired site in the skin. 17 The various carriers majorly used for topical delivery include liposomes, niosomes, transferosomes, ethosomes, SLNs and NLCs. 19–22 However, nanoemulsion as topical delivery vehicles is a potential approach as they are simplest transparent kinetically stable systems with globule size in the range of 20-200 nm. 17 They offer high drug loading and retention into the skin owing to their small globule size and lipidic nature. 23 Nanoemulsion is hypothesized to penetrate the rough plaques of psoriatic skin by extracting and swelling of skin lipids to achieve enhanced penetration through the pores. 24,25 Further, it can be easily loaded into gel form, 26 and it aids in the deliverability of drugs into the skin by hydration mechanism 27 and enhancing retention of drugs in the skin. 26,28,29 In the present work, a nanoemulsion (NE) with the combination of CP and CT was prepared and characterized for release profile, ex vivo skin permeation, ex vivo efficacy studies using HaCaT cell line and in vivo anti-psoriatic efficacy and was assessed by Imiquimod induced psoriasis in mice.

solubility of drug in these components and their emulsification capacity. Capmul MCM C8 EP, Cremophor RH 40 and Labrafil 1944 CS in the weight ratio 5:3:2 were selected as oil, surfactant and co-surfactant. NE was formed by spontaneous emulsification method. Briefly, 5 mg CP and 0.5 mg CT were dissolved in 500 mg oil followed by addition of 300 mg surfactant and 200 mg co-surfactant. The mixture was then subjected to vortexing for 15 min and gently heated at 40 °C for 5 min and then diluted to 10 ml with water to attain final dose of 0.05% w/v CP and 0.005% w/v CT. The resultant NE was allowed to stand for 2 h for equilibration before characterization which is also depicted in supplementary data. Similarly, NE-gel preparation and optimization could be retrieved through electronic supplementary data. Characterization of NE-gel for spreadability, stability and rheological behavior is also supplemented in electronic database.

100

Morphology of NE

116

F

48

101 102 103 104 105 106 107 108 109 110 111 112 113 114 115

The shape and morphology of NE was examined using TEM, (FEI, Tecnai, G2). Sample was stained with 1% w/v aqueous solution of phosphotungstic acid and then viewed under the microscope.

117

121

P

47

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

R O O

2

118 119 120

Methods

Drug release and cell line studies

69

Materials

122

70

CP was provided as a generous gift by Symbiotec Pharmalab, India. CT was purchased from Chem Express, China. Pluronic F-68, Pluronic F-127, TWEEN® 20, TWEEN® 80, trypsin– EDTA, coumarin-6 (C-6), sodium dodecyl sulfate (SDS), triton X-100, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), Dulbecco's modified Eagle's medium (DMEM), and 10% fetal bovine serum were purchased from Sigma, USA. Capmul® MCM EP, Captex® 355, Capmul® MCM-C8, and Captex® 200P were procured as generous gifts from Abitech Corporation, Janesville, USA. Isopropyl myristate (IPM) was procured from Loba Chemie Pvt. Ltd., India. Plurol® Oleique, Caproyl PGMC, Maisine 35-I and Labrafil® 1944 CS were obtained as gift samples from Gattefosse, France. Cremophor RH 40 was purchased from BASF Chemical Company, USA. Carbopol® 980 was purchased from Lubrizol India Pvt. Ltd., India. Penicillin and streptomycin were purchased from PAA Laboratories GmbH, Austria. HaCaT cell line was procured from National Centre for Cell Science (NCCS), Pune, India. ELISA kits, i.e., RayBio® ELISA Kit TNF-alpha and RayBio® ELISA Kit IL-6 were purchased from RayBiotech, Inc. GA, USA. Imiquimod cream 5% (Imiquad™) and betamethasone dipropionte gel 0.05% w/w (Betagel®) was purchased from local chemist. Ultra-pure deionized water was used throughout all the experiments. All other reagents were of analytical grades or higher.

Drug release studies Release of CP–CT from NE and NE-gel was determined by dialysis method using cellophane membrane (molecular weight cut-off 12,000 Da). 30 Release media consisted of phosphate buffer saline (pH 5.8) and methanol in the proportion 70:30 with maintenance of sink conditions. The dialysis bag was filled separately with free CP–CT dispersed in release media and CP– CT loaded NE as well as NE-gel, which were then immersed individually in 30 ml glass vial containing 20 ml release medium. These vials were placed in shaker bath (50 rpm and 37 °C) and 200 μl samples were withdrawn from receptor compartment and replaced with an equal volume of release medium. The samples were then analyzed for cumulative amount of drug release by validated HPLC method. Ex vivo HaCaT cell line studies The ex vivo efficacy studies were carried out on the HaCaT cell lines. 31 HaCaT cell lines were grown in tissue culture flasks (75 cm 2) at 37 °C under 5% CO2 atmosphere. The cell growth medium comprised Dulbecco's modified Eagle's medium (DMEM, Sigma) supplemented with 10% FBS, 100 U/mL penicillin, and 100 μg/mL streptomycin (PAA Laboratories GmbH, Austria). For qualitative uptake and MTT assay, HaCaT cells were seeded at a density of 50,000 cells/well and 10,000 cells/well in 6-well culture plate and 96-well plate (Costars, Corning Inc., NY, USA). For cellular uptake studies, cells were incubated for 3 h with free C6 and C6 loaded nanoformulation (equivalent to 1 μg/mL of free C6). After incubation, extracellular particles were removed by washing with Hanks' Balanced Salt Solution (HBSS) and were observed under a confocal laser microscope (CLSM) (Olympus FV1000). Further for MTT assay, HaCaT cells were treated with fresh media containing 1% Triton-X 100 (positive control), free drugs

136

77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99

E

T

C

76

E

75

R

74

O R

73

C

72

U N

71

D

68

Preparation, optimization and characterization of CP–CT NE and NE-gel Preparation and optimization of drug loaded NE using design of Expert® is described in supplementary data. Selection of oils, surfactants and co-surfactants was accomplished based on

123 124 125 126 127 128 129 130 131 132 133 134 135

137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209

C

168

E

In vivo studies

216

In vivo anti-psoriatic activity The in vivo efficacy studies were carried out using BALB/c mice, which were duly approved by Institutional Animal Ethics Committee, NIPER (IAEC Project Number: IAEC/14/70-R). Animals were housed at 25 ± 2 °C, 45% relative humidity and 12 h light/dark cycle and provided with food and water ad libitum. The animals were divided into 5 groups (4 animals per group) as follows:

217

F

167

Dermal pharmacokinetic study by tape stripping Dermal pharmacokinetics was determined by tape stripping technique. 34,35 The pig skin samples were exposed to free drug, CP–CT loaded NE and NE-gel for 8 h in Franz diffusion cell. After 8 h, the exposed skin was washed thrice with phosphate buffer saline (PBS, pH 5.8) and stratum corneum (SC) was completely removed by detaching 15 strips (19 mm Scotch™ 3M USA) from the exposed surface. The tapes and the remaining skin samples, after removal of SC, were chopped into small pieces and soaked in methanol overnight followed by sonication for 2 h to extract the drugs. The extracted samples were analyzed by validated HPLC method. The amount detected in receptor compartment is the index of transdermal delivery, while the amount in SC, viable layers (epidermis and dermis) is the index of topical delivery.

R

166

O R

162

C

161

U N

160

R O O

Skin permeation The ex vivo skin permeation was studied on pig ear skin 23 using static Franz diffusion cells (PermeGear Inc. USA) having contact surface area of 0.64 cm 2. The assembly was maintained at 37 °C with speed of rotation of magnetic bead in the receptor compartment at 75 rpm. The pig ears were collected from slaughterhouse. For permeation studies, firstly pig ears were processed and dorsal portion of pig ear skin was shaved using Sterling-2 animal hair clipper (Wahl, USA). The subcutaneous fat was removed using blunt forceps and the skin was washed thrice with phosphate buffer saline of pH 5.8. The skin samples of 15 mm diameter were punched and mounted between donor and receptor compartment with the help of clamp. The receptor compartment was filled with 5 ml of phosphate buffer saline of pH 5.8 and methanol (70:30). Then the formulations viz. free CP and CT dissolved in receptor methanol, CP–CT loaded NE and CP–CT loaded NE-gel were applied in the donor compartment. Aliquots of 250 μl were periodically withdrawn from receptor compartment and equal volume of receptor medium was replaced. The samples collected were analyzed by developed and validated HPLC based bioanalytical method.

159

Group I: Healthy animals (sham group) Group II: Negative control (disease induced) Group III: Positive control (marketed formulation) Group IV: Free drug (CP–CT) in gel Group V: CP–CT loaded NE-gel

P

165

158

210

Mechanistic understanding of skin distribution using confocal laser scanning microscopy (CLSM) The skin samples were prepared as per similar procedure followed for ex vivo skin permeation. The excised skin was mounted with a clamp between donor compartment exposed to C6 aqueous dispersion, C6 loaded into optimized NE as well as NE-gel and receptor compartment filled with phosphate buffer saline pH 5.8. The skin samples were collected after 24 h of application and the transverse sections of skin samples (15-20

211 212 213 214 215

218 219 220 221 222 223 224 225 226 227 228 229 230

The hair on the back of animals was shaved using Sterling-2 animal hair clipper (Wahl, USA) a day prior to initiation of the study and psoriasis was induced by once daily application of Imiquimod cream 5%. 36 The cream (0.125 g) was applied in the morning, consecutively for 6 days, onto the shaved back and left ear of each mouse in all the groups except the sham group, which was the control normal mice group (Figure 4, A shows model development). For treatment, 0.05% w/w Betamethasone Dipropionate gel (Betagel®, Micro Labs Pvt. Ltd.) was applied once daily in the evening as positive control (marketed preparation) group and similarly the other two groups, i.e., Group IV and V were treated with once daily dose of free CP– CT in gel and NE-gel respectively for 5 consecutive days in the evening. The negative control group was not provided with any treatment and kept as induced control group. The morning once daily application of Imiquimod was continued during the treatment period also. After 5 days, the animals were sacrificed and blood, skin and spleen were collected from each animal.

231

Measurement of ear thickness and inflammation severity scoring

249

The thickness of the ear (right ear as control and left ear for extent of inflammation) was measured using digital caliper. The severity of redness and inflammation of ear skin and back skin of mice was measured based on the presence of erythema, scaling, thickening and lesions on the skin. The scoring was reported individually for all groups from 0 to 4 as: 0, none; 1, slight; 2, moderate; 3, marked; and 4, very marked.

250

Spleen and skin histology

257

Samples of the spleen and skin were fixed in 10% neutral buffered formalin. The samples were fixed onto the slides after staining and observed under microscope. The skin samples were also visualized under scanning electron microscope and compared for ceding of the lesions.

258

D

Ex vivo studies

157

μm thickness) were taken using Freeze Microtome (MICROM), which were observed under IX 51 microscope combined with Flu view 1000 confocal laser unit (OLYMPUS, Japan). The fluorescent portion images were captured at optical zoom of 10X keeping excitation and emission wavelengths as 459 nm and 505 nm respectively.

E

164

156

T

163

and nanoformulation at a concentration of 20, 50 and 100 μg/mL. After 24 h incubation time, the media was aspirated and cells were washed three times with HBSS. Subsequently, 150 μL of MTT solution (500 μg/mL in PBS) was added to each well and reincubated for 4 h. The developed formazan crystals were then dissolved in 200 μL of DMSO. The optical density (OD) of the resultant solution was then measured at 570 nm using an ELISA plate reader (BioTek, USA). 32,33 IC50 was calculated using GraphPad Prism 7.0 GraphPad Software (San Diego, CA, USA).

155

3

232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248

251 252 253 254 255 256

259 260 261 262

4

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

263

Analysis of TNF-α and IL-6 levels in serum

264

Blood collected from each animal was allowed to clot for 30 min at room temperature and subjected to centrifugation at 2500 rpm for 5 min at 25 °C. Serum was collected in the separate set of plastic vials and stored in freezer at −80 °C and elevated levels of IL-6 and TNF-α were measured. The procedure followed was according to the manual specified with the ELISA kits for TNF-α (RayBio® ELISA Kit TNF-alpha, RayBiotech, Inc. GA, USA) and IL-6 (RayBio® ELISA Kit IL-6, RayBiotech, Inc. GA, USA).

271 272 273 274 275 276 277 278 279 280 281

In vivo skin irritation The in vivo skin irritation study was performed on dorsal shaved (using 1 mm animal hair clipper, Sterling) skin of SD rats (weighing 200-250 g). The rats were grouped as: Group I: Negative control (capsaicin) Group II: Free CP–CT loaded gel Group III: CP–CT loaded NE-gel

Figure 1. TEM image of CP–CT loaded NE.

291

Statistical analysis

292

297

The statistical analysis of all the result was accomplished utilizing GraphPad Prism 7.0 GraphPad Software (San Diego, CA, USA). One way ANOVA followed by Bonferroni multiple comparison procedure was applied throughout the manuscript and P value of b0.01 was considered significant. All results are depicted as value ± SD.

298

Results

299

Preparation, optimization and characterization of CP–CT NE and NE-gel

287 288 289

293 294 295 296

300 301

E

286

R

285

O R

284

C

283

C

290

Samples weighing about 200 mg were applied on the shaved region using a neat cotton swab and kept for 2 h on the shaved region. The trans-epidermal water loss (TEWL) was measured 30 using vapometer (Delfin, Finland) before treatment and subsequently for 4 days during treatment so as to assess skin integrity. The skin irritation was assessed on the basis of redness or erythema based on visual observations such as follows: 0, no erythema; 1, slight erythema (light pink); 2, moderate erythema (dark pink); 3, moderate to severe erythema (light red); and 4, severe erythema (extreme redness).

282

309

Morphology of NE

310

Figure 1 shows the TEM image of the resultant o/w NE. The spherical oil droplets were distributed uniformly and homogeneously throughout the formulation with no aggregates present in the system.

303 304 305 306 307

311 312 313

U N

308

CP–CT NE and NE-gel were prepared wherein, optimized CP–CT loaded NE depicted the average droplet size of 35.45 ± 2.68 nm and lower polydispersity values of 0.080 ± 0.024 with uniform and homogenous distribution. Similarly, by incorporation of NE into gel, the properties of gel were unaltered and led to the formation of stable NE-gel formulation. Detailed results for the section could be retrieved through electronic supplementary data attached with the article.

302

F

270

R O O

269

Drug release and cell line studies

314

Drug release studies The in vitro release of CP and CT from NE and NE-gel compared to free drugs solution is shown in Figure 2. The figure clearly indicates complete release of free drugs within 6 h; whereas CP–CT loaded NE showed slower release up to 10 h. The CP–CT loaded NE-gel showed controlled release of both the drugs for up to 36 h.

315

P

268

D

267

E

266

T

265

316 317 318 319 320

Ex vivo HaCaT cell line studies The % growth inhibition of cells was measured as shown in Figure 3, A. The IC50 value of the groups is depicted in Table 1. On the other hand, 1% Triton-X 100 solution (utilized as positive control) showed N90% growth inhibition. Further, the results depict relatively higher efficacy of formulation as compared to free drug(s). The evidence of higher uptake in HaCaT cell line was generated from microscopic images using C6, as shown in Figure 3, B, where aqueous dispersion of coumarin-6 showed less fluorescence than C6 loaded into NE as seen in Figure 3, A(a).

321

Ex vivo studies

331

Skin permeation Non-detectable amounts of CP and CT permeated through the pig ear skin during the period of exposure of skin to the various formulations.

332

Dermal pharmacokinetic The penetration of drugs from various formulations into the pig ear skin is shown in Figure 3. The free CP–CT solution and gel showed limited drug penetration. As can be seen in the tape strip profiles in Figure 4, A-D, the penetration of NE-gel is slightly lower than NE due to controlled release of drug from the gel. The penetration of CP from NE increased by 5.48 and 8.08 in SC and viable layers respectively, as compared to free drugs, whereas the NE-gel increased 4.18 times and 5.20 times the penetration of CP in SC and viable layers respectively. Similarly, for CT, NE showed 2.6 and 3.1 times increased penetration into SC and viable layers respectively. The NE-gel showed 1.88 and 2.86 times enhanced penetration of CT into SC and viable layers respectively.

336

322 323 324 325 326 327 328 329 330

333 334 335

337 338 339 340 341 342 343 344 345 346 347 348

5

R O O

F

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

C

O R

R

E

C

T

E

D

P

Figure 2. In vitro drug release profile of free drug mixture CP-NE and CP–CT loaded NE-gel. Each value is represented as mean ± SD (n = 3).

U N

Figure 3. HaCaT cell lines studies with graph showing % growth inhibition of cells by CP–CT NE compared with free drugs (CP/CT) and their mixture (CP– CT). Each value is represented as mean ± SD (n = 3). Optical microscopic images of coumarin-6 loaded NE- (A) nanoemulsion globules upon incubation at 1 μg/ml for 24 h. In all images, panel (a) images under green fluorescence channel; (b) corresponding differential interference contrast images of HaCaT cells; (c) superimposition of panels (a) and (b). (d) Image showing horizontal lines series analysis of fluorescence along the white line. (B) Free coumarin-6 treated cells (** signifies P b 0.01 in comparison to free drugs).

349 350 351 352 353 354 355 356

Mechanistic understanding of skin distribution using confocal laser scanning microscopy (CLSM) The results of sections of pig ear skin treated with aqueous dispersion of C6, C6 loaded into optimized NE and NE-gel are show in Figure 4, E. As evident from Figure 4, E(a), the aqueous dispersion of C6 did not show any penetration into the skin but was majorly confined to the upper layer of skin. Figure 4, E(b), shows the results of C6 loaded NE, wherein the dye showed equal distribution

t1:1 t1:2

Table 1 IC50 value of different study groups in MTT assay. Study group

IC50 value (μg/ml)

t1:3

Free CP Free CT Free CP–CT CP–CT NE

199.80 634.53 111.10 46.56

t1:4 t1:5 t1:6 t1:7

± ± ± ±

21.35 83.19 13.28 6.96

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

D

P

R O O

F

6

357

360

In vivo studies

361

369

In vivo anti-psoriatic activity As shown in Figure 5, B, there was a reduction in skin thickness, redness and ear inflammation for different groups. The NE-gel showed maximum reduction in skin inflammation and scaly lesions when compared to the positive control group, negative control group and groups treated with free CP–CT loaded gel. The augmented action could be attributed to higher uptake of CP–CT from NE and NE-gel into epidermal and dermal layers of the skin as evident from the skin penetration studies.

370

Measurement of ear thickness

371

375

Figure 5, G depicts the measurements of ear thickness. The negative group showed left ear thickness of N400 μm in comparison to the control right ear thickness (≈250 μm). All treated groups showed a significant decrease in left ear thickness, but it was lowest for the NE-gel treated group.

376

Scoring severity of skin inflammation

377

The psoriatic scaling of all the groups is mentioned in Table 2 and the value for the negative control group is ≥2, due to marked development of psoriatic lesions. The severity of psoriatic

365 366 367 368

372 373 374

378 379

E

R

O R

364

C

363

U N

362

C

359

into SC and viable layers. Similarly, Figure 4, E(c) shows the penetration of dye loaded into NE-gel. The gel base also enhanced the retention of dye into the deeper layers of the skin.

358

T

E

Figure 4. Skin distribution of CP and CT from NE and NE-Gel (A). Comparison of penetration profiles of CP in skin layers. (B) Comparison of penetration profiles of CT in skin layers. (C) Depth of penetration of CP. (D) Depth of penetration of CT. (E) CLSM images of skin sections treated with (a) coumarin-6 aqueous dispersion, (b) coumarin-6 loaded NE, and (c) coumarin-6 NE loaded gel (** signifies P b 0.01 in comparison to free drugs).

lesions is significantly less (≤2) for the NE-gel, positive and free CP–CT loaded gel treated groups.

380

Histology and SEM analysis of skin

382

The SEM images of skin for all groups are shown in Figure 5, C. The SEM images of the sham group shows the presence of intact skin with hair follicles. The negative control group showed complete disruption of skin with dispersed psoriatic lesions, scales and no visible hair follicles. The positive control group and free CP–CT loaded gel treated group depicted signs of disrupted skin lipids compared to the sham group after treatment. However, the NE-gel treated group resembled the normal physiology of skin with revival of hair follicles as well. It can be clearly seen in the images that the NE-Gel treated group showed disappearance of psoriatic lesions. The histological images of skin of mice from different groups are shown in Figure 5, E, which also depicts similar results, wherein the NE-gel treated group restored normal physiology of skin and it resembled to that of the sham group.

383

Histology of the spleen

397

The histology of the spleen, as depicted in Figure 5, D, showed complete remission of spleen tissue in the case of the NE-gel treated group with reappearance of white pulp (blue in color) and red pulp (pink in color) areas of the spleen as in the case of normal spleen tissue. The red pulp and white pulp areas cannot be distinguished in the negative control group. Similar, but insignificant restoration of splenic tissue could be seen in the

398

381

384 385 386 387 388 389 390 391 392 393 394 395 396

399 400 401 402 403 404

7

E

C

T

E

D

P

R O O

F

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

Psoriatic scaling

t2:3 t2:4

Animal

Sham group

t2:5 t2:6 t2:7 t2:8

1 2 3 4

0 0 0 0

405 406 407 408 409 410

C

t2:2

Table 2 Score chart of severity of psoriatic inflammation for all the animals in different groups.

U N

t2:1

O R

R

Figure 5. (A) Progressive scaling of mice skin over the period of 7 days after application of 5% Imiquimod cream. (B) Visual analysis of improvement in psoriatic lesions after 6 days of treatment for different groups. (C) SEM images of skin samples of different groups. (D) Histological images of skin for different groups. (E) Histology of spleen for different groups. (F) Dimensions of the spleen for various treated groups after 5 days of treatment. (G) Comparison of ear thickness (right and left ear) for different treated groups. Each value is represented as mean ± SD. (** signifies P b 0.01 in comparison to the sham group [healthy control] and “ns” as non-significant change).

Negative control

Positive control

Free CP–CT gel

NE-gel

3 3 2 4

1 2 1 1

2 2 1 2

1 2 1 1

case of marketed formulation and free CP–CT loaded gel. The dimensions of the spleen after treatment were also compared as shown in Figure 5, F. The size of the spleen for the negative control group was the largest followed by a decreasing order for the positive control group, free CP–CT gel treated group, sham group and NE-gel treated group being nearly equal.

Analysis of TNF-α and IL-6 levels in serum

411

The levels of TNF-α and IL-6 found in serum of animals of different groups are depicted in Figure 6. The marked increase in levels of IL-6 and TNF-α (with concentration ≈ 1000 pg/ml and 90 pg/ml respectively) in the case of the negative control group shows a good model development. Following treatment, there was an approximately 95% reduction in IL-6 levels for the NE-gel treated group compared to negative control. Similarly, the NE-gel treated group showed a 77.77% reduction in TNF-α concentration compared to the negative control group. The analyzed levels of IL-6 and TNF-α are depicted in Figure 6.

412

In vivo skin irritation

422

As can be seen in Figure 7, capsaicin showed higher values of percentage increase in TEWL and maximum irritation potential. Free CP–CT when loaded into gel had higher irritation potential as compared to NE-Gel, which was better tolerated. The scoring

423

413 414 415 416 417 418 419 420 421

424 425 426

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

R O O

F

8

D

P

Figure 6. IL-6 and TNF-α levels in blood serum of animals from the different study groups (** signifies P b 0.01 in comparison to the sham group [healthy control] and “ns” as non-significant change).

E

Table 3 Score chart of severity of skin irritation potential for all the animals in different groups.

429

430

Discussion

431

In view of the high dose of CP, selection of oil was based on its maximum solubility. CP showed high solubility in almost all the oils, with maximum solubility in Caproyl 90 (Supplementary data Figure S1). The latter, however, is hard to emulsify and required a large amount of surfactant for its emulsification, which is undesirable for topical delivery in psoriasis due to irritation tendency of surfactants. 37 Similarly, CP had high solubility in Caproyl PGMC, which possesses surfactant properties of its own and resulted in nanoemulsion with no

432 433 434 435 436 437 438 439

t3:2

Animal

Capsaicin

Free CP–CT in gel

NE-gel

t3:3 t3:4

1 2 3 4

4 4 3 4

1 1 1 2

1 0 0 1

t3:5 t3:6 t3:7 t3:8

T C E R O R

428

of skin irritation potential of the various formulations is depicted in Table 3 with highest values in capsaicin followed by the free CP–CT gel and NE-gel treated group.

U N

427

C

Figure 7. Comparison of % increase in TEWL for NE-gel with free drug mixtures (CP–CT) loaded gel and capsaicin (** signifies P b 0.01 in comparison to negative control).

Psoriatic scaling

t3:1

trend in globule size, and PDI due to interactions with the surfactant. Thus, Capmul MCM C8 EP, though had lower solubility of CP (40.68 ± 2.83 mg/g), was selected for formulation development. Capmul MCM C8 EP was able to solubilize low dose of CT (0.005% w/w) as well, hence, it served as common oil for both the drugs. The two drugs were found to be compatible in oil as revealed by HPLC analysis (data not shown here). The mechanism of solubilization is due to the interaction of the hydrophobic groups of both the drugs with the hydrophobic chains of the oil. Similar mechanism has been reported for solubilization of Tmx and QT by Capmul MCM EP in the literature. 38 Cremophor RH 40 was selected as surfactant due to formation of small droplets in the emulsion system coupled with narrow PDI (Supplementary data Table S1A). Co-surfactants were also added to the nanoemulsion, which further lowered the globule size and PDI. The co-surfactants are reported to act by imparting flexibility to the interfacial film formed by surfactants and further reducing the interfacial tension. 39,40 Labrafil 1944 CS was selected as co-surfactant since it produced sub-micron emulsion (Supplementary data Table S1B). In in vitro release study (Figure 2), it is evident that the NE and NE-gel follows sustained release pattern for both the drugs in comparison to free drug. This can be explained on the basis that the drug is encapsulated into the

440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520

F

474

Appendix A. Supplementary data

R O O

473

521 522 523 524 525 526 527 528 529 530

531

Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.nano.2017.02.009.

532

References

534

1. Duffin KC, Chandran V, Gladman DD, Krueger GG, Elder JT, Rahman P. Genetics of psoriasis and psoriatic arthritis: update and future direction. J Rheumatol 2008;35:1449-53. 2. Lowes MA, Bowcock AM, Krueger JG. Pathogenesis and therapy of psoriasis. Nature 2007;445:866-73. 3. Nickoloff BJ. Keratinocytes regain momentum as instigators of cutaneous inflammation. Trends Mol Med 2006;12:102-6. 4. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet 2007;370:263-71. 5. Raychaudhuri SK, Maverakis E, Raychaudhuri SP. Diagnosis and classification of psoriasis. Autoimmun Rev 2014;13:490-5. 6. Gupta M, Gupta A. Depression and suicidal ideation in dermatology patients with acne, alopecia areata, atopic dermatitis and psoriasis. Br J Dermatol 1998;139:846-50. 7. Laws PM, Young HS. Topical treatment of psoriasis. Expert Opin Pharmacother 2010;11:1999-2009. 8. Katoh N, Kishimoto S. Combination of calcipotriol and clobetasol propionate as a premixed ointment for the treatment of psoriasis. Eur J Dermatol 2003;13:382-4. 9. Austad J, Bjerke JR, Gjertsen BT, Helland S, Livden JK, Morken T, et al. Clobetasol propionate followed by calcipotriol is superior to calcipotriol alone in topical treatment of psoriasis. J Eur Acad Dermatol Venereol 1998;11:19-24. 10. Devaux S, Castela A, Archier E, Gallini A, Joly P, Misery L, et al. Topical vitamin D analogues alone or in association with topical steroids for psoriasis: a systematic review. J Eur Acad Dermatol Venereol 2012;26:52-60. 11. Olsen EA, Cornell RC. Topical clobetasol-17-propionate: review of its clinical efficacy and safety. J Am Acad Dermatol 1986;15:246-55. 12. Jegasothy B, Jacobson C, Levine N, Millikan L, Olsen E, Pinnell S, et al. Clobetasol propionate versus fluocinonide creams in psoriasis and eczema. Int J Dermatol 1985;24:461-5. 13. Miller J, Munro D. Topical corticosteroids: clinical pharmacology and therapeutic use. Drugs 1980;19:119-34. 14. Kragballe K. Treatment of psoriasis with calcipotriol and other vitamin D analogues. J Am Acad Dermatol 1992;27:1001-8. 15. Segaert S, Duvold LB. Calcipotriol cream: a review of its use in the management of psoriasis. J Dermatolog Treat 2006;17:327-37. 16. Ashcroft DM, Po ALW, Williams HC, Griffiths CE. Systematic review of comparative efficacy and tolerability of calcipotriol in treating chronic plaque psoriasis. BMJ 2000;320:963-7. 17. Pradhan M, Singh D, Singh MR. Novel colloidal carriers for psoriasis: current issues, mechanistic insight and novel delivery approaches. J Control Release 2013;170:380-95.

P

472

D

471

E

470

formulations have limited skin access. The combination therapy approach in the present work causes good healing process in psoriasis. The present approach has reduction in skin irritation as compared to the free drug in gel. Also, the local topical concentration improved compared to the other groups mentioned in the study. This approach would offer selective as well as satisfactory safety profile. With less irritation potential of formulation, long term treatment is possible with high patient compliance. This nurtures the future application and commercialization of the developed formulation.

T

469

C

468

E

467

R

466

O R

465

lipidic system in the NE and thus release is hindered as the drug is more hydrophobic and tends to retain in the lipidic system. Further, NE-Gel retarded the release even more since the NE is retained more inside the 3-D network structure of the gel system and takes a lag time in release of the system. The developed nanoemulsion showed higher growth inhibition in HaCaT cell line and penetration of drugs in SC and viable layers (Figure 3). The above benefits are probably due to enhanced penetration and controlled release of drugs from the nanoemulsion system (Figure 5). The first basic component of nanoemulsion, i.e., oil, carries the drug in solubilized form, could be involved in breaking the thermodynamic and kinetic barrier for diffusion of drug molecules. 41 The other basic components are surfactants and co-surfactants which act by solubilizing and disrupting the lipidic structure of skin. 42 The water in the NE also contributes toward enhanced penetration by increasing hydration of the skin leading to swelling of lipids, paving a way for drugs to diffuse through the pores in the skin. 43 The small globules of size b100 nm for nanoemulsion also contribute to penetration by exploiting conventional routes of intracellular, intercellular and transappendageal transport. 44 Another mechanism reported in literature is that nanoemulsion penetrates into the skin by extracting the lipids and disrupting the H-bonds of lipid bilayers. 45 The results of retention of nanoemulsion locally into the viable layers of skin was desirable as pathogenesis of psoriasis involves epidermal keratinocytes and angiogenesis of deeper layers. 24 The higher penetration into deeper layers is desirable, as the drug can be expected to penetrate efficiently through the scaly psoriatic skin. Since psoriasis is an autoimmune disorder, generation of auto-antibodies takes place further increasing the levels of the cytokines (IL-6 and TNF-α). The process takes place in the spleen being the major organ related to immunity. This leads to higher burden on the spleen and increased cytokine production, which lead to swelling of the spleen or spleenomegaly. 39–42,46 The higher uptake of drugs, i.e., CT had a marked effect on reduction in levels of IL-6. 15 Furthermore, CT inhibits keratinocytes in the SC thereby, suppressing the production of cytokines in the psoriatic skin. 47 CP, an anti-inflammatory compound, acts on inflammatory cells like macrophages and neutrophils and inhibits the maturation of dendritic cells ultimately leading to suppression of the production of various pro-inflammatory cytokines. 7,13 This leads to decreased spleenomegaly condition which is validation for higher activity of formulation in diseased condition. The higher penetration of drugs was anticipated to cause irritation on the rat skin, however, negligible irritation was observed in the case of the NE-gel treated group (Figure 7). CT has tendency to cause skin irritation, but higher dose of CP penetrated into the skin could mask the irritation caused by CT. The other reasons for minimum irritation could be sustained release and controlled exposure of drugs to the skin. Additionally, NE-gel contains drugs in encapsulated form with minimal direct contact with the skin. The developed formulation improved the local concentration of the two drugs reducing their systemic side effects, in specific higher penetration into the skin layers where conventional

C

464

U N

463

9

533

535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578

R

E

U N

C

670

D

P

R O O

F

33. Ciapetti G, Cenni E, Pratelli L, Pizzoferrato A. In vitro evaluation of cell/ biomaterial interaction by MTT assay. Biomaterials 1993;14:359-64. 34. Touitou E, Meidan VM, Horwitz E. Methods for quantitative determination of drug localized in the skin. J Control Release 1998;56:7-21. 35. Escobar-Chavez JJ, Merino-Sanjuán V, López-Cervantes M, UrbanMorlan Z, Piñón-Segundo E, Quintanar-Guerrero D, et al. The tapestripping technique as a method for drug quantification in skin. J Pharm Pharm Sci 2008;11:104-30. 36. Sun J, Zhao Y, Hu J. Curcumin inhibits imiquimod-induced psoriasislike inflammation by inhibiting IL-1beta and IL-6 production in mice. PLoS One 2013;8:e67078. 37. Charaf UK, Hart GL. Phospholipid liposomes/surfactant interactions as predictors of skin irritation. J Soc Cosmet Chem 1991;42:71-86. 38. Jain AK, Thanki K, Jain S. Solidified self-nanoemulsifying formulation for oral delivery of combinatorial therapeutic regimen: part I. Formulation development, statistical optimization, and in vitro characterization. Pharm Res 2014;31:923-45. 39. Sarkhejiya N, Nakum M, Patel V, Atara S, Desai T. Emerging trend of microemulsion in formulation and research. Int Bull Drug Res 2012;1:54-83. 40. Sarkhejiya Naimish A, Nakum Mayur A, Patel Vipul P, Atara Samir A, Desai Thusarbindu R. Emerging trend of microemulsion in formulation and research. Int Bull Drug Res 2000;1:54-83. 41. van der Fits L, Mourits S, Voerman JS, Kant M, Boon L, Laman JD, et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J Immunol 2009;182:5836-45. 42. Shakeel F, Baboota S, Ahuja A, Ali J, Shafiq S. Skin permeation mechanism and bioavailability enhancement of celecoxib from transdermally applied nanoemulsion. J Nanobiotechnol 2008;6:1. 43. Williams AC, Barry BW. Penetration enhancers. Adv Drug Deliv Rev 2012;64:128-37. 44. Yilmaz E, Borchert H-H. Effect of lipid-containing, positively charged nanoemulsions on skin hydration, elasticity and erythema—an in vivo study. Int J Pharm 2006;307:232-8. 45. Frelichowska J, Bolzinger M-A, Pelletier J, Valour J-P, Chevalier Y. Topical delivery of lipophilic drugs from o/w Pickering emulsions. Int J Pharm 2009;371:56-63. 46. Shakeel F, Baboota S, Ahuja A, Ali J, Shafiq S. Skin permeation mechanism of aceclofenac using novel nanoemulsion formulation. Pharmazie 2008;63:580-4. 47. Dam T, Møller B, Hindkjaer J, Kragballe K. The vitamin D3 analog calcipotriol suppresses the number and antigen-presenting function of Langerhans cells in normal human skin. J Invest Dermatol Symp Proc 1996:72-7.

E

C

18. McNeill SC, Potts RO, Francoeur ML. Local enhanced topical delivery (LETD) of drugs: does it truly exist? Pharm Res 1992;9:1422-7. 19. Schreier H, Bouwstra J. Liposomes and niosomes as topical drug carriers: dermal and transdermal drug delivery. J Control Release 1994;30:1-15. 20. Cevc G, Blume G. New, highly efficient formulation of diclofenac for the topical, transdermal administration in ultradeformable drug carriers, Transfersomes. BBA-Biomembranes 2001;1514:191-205. 21. Bhalaria M, Naik S, Misra A. Ethosomes: a novel delivery system for antifungal drugs in the treatment of topical fungal diseases. Indian J Exp Biol 2009;47:368. 22. Souto E, Wissing S, Barbosa C, Müller R. Development of a controlled release formulation based on SLN and NLC for topical clotrimazole delivery. Int J Pharm 2004;278:71-7. 23. Baroli B, López-Quintela MA, Delgado-Charro MB, Fadda AM, Blanco-Méndez J. Microemulsions for topical delivery of 8methoxsalen. J Control Release 2000;69:209-18. 24. Khandavilli S, Panchagnula R. Nanoemulsions as versatile formulations for paclitaxel delivery: peroral and dermal delivery studies in rats. J Invest Dermatol 2007;127:154-62. 25. Vaddi H, Wang L, Ho P, Chan S. Effect of some enhancers on the permeation of haloperidol through rat skin in vitro. Int J Pharm 2001;212:247-55. 26. Alves MP, Scarrone AL, Santos M, Pohlmann AR, Guterres SS. Human skin penetration and distribution of nimesulide from hydrophilic gels containing nanocarriers. Int J Pharm 2007;341:215-20. 27. Alves P, Pohlmann A, Guterres S. Semisolid topical formulations containing nimesulide-loaded nanocapsules, nanospheres or nanoemulsion: development and rheological characterization. Pharmazie 2005;60:900-4. 28. Varma VNSK, Maheshwari P, Navya M, Reddy SC, Shivakumar H, Gowda D. Calcipotriol delivery into the skin as emulgel for effective permeation. Saudi Pharm J 2014;22:591-9. 29. Chen H, Chang X, Du D, Li J, Xu H, Yang X. Microemulsion-based hydrogel formulation of ibuprofen for topical delivery. Int J Pharm 2006;315:52-8. 30. Sonawane R, Harde H, Katariya M, Agrawal S, Jain S. Solid lipid nanoparticles-loaded topical gel containing combination drugs: an approach to offset psoriasis. Expert Opin Drug Deliv 2014;11:1833-47. 31. Rückert R, Asadullah K, Seifert M, Budagian VM, Arnold R, Trombotto C, et al. Inhibition of keratinocyte apoptosis by IL-15: a new parameter in the pathogenesis of psoriasis? J Immunol 2000;165:2240-50. 32. Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983;65:55-63.

O R

579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623

A. Kaur et al / Nanomedicine: Nanotechnology, Biology, and Medicine xx (2017) xxx–xxx

T

10

624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669