Vitamin-Derived Nanolipoidal Carriers for Brain ... - ACS Publications

Feb 3, 2017 - Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Distt. Ajmer,. Rajasth...
0 downloads 0 Views 4MB Size
Research Article pubs.acs.org/chemneuro

Vitamin-Derived Nanolipoidal Carriers for Brain Delivery of Dimethyl Fumarate: A Novel Approach with Preclinical Evidence Pramod Kumar,† Gajanand Sharma,‡ Rajendra Kumar,§ Ruchi Malik,† Bhupinder Singh,‡,§ O. P. Katare,‡ and Kaisar Raza*,† †

Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Distt. Ajmer, Rajasthan, India-305817 ‡ Division of Pharmaceutics, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India-160014 § UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles and Nanocomposites, Panjab University, Chandigarh, India-160014 S Supporting Information *

ABSTRACT: Various oral treatment options have been reported for relapsing multiple sclerosis. Recently, dimethyl fumarate (DMF) has been approved for the management of the same. Though effective, DMF is associated with concerns like multiple dosing, patient incompliance, gastrointestinal flushing, lower brain permeation, and economic hurdles. Henceforth, the objective of the present study was to develop vitamin-based solid lipid nanoparticles (SLNs) for effective brain delivery of DMF with a promise of once-a-day dosing. The developed SLNs were characterized for micromeritics, morphology, entrapment efficiency, drug loading and in vitro drug release. Caco-2 and SH-SY5Y cell lines were used to assess the intestinal permeability and neuronal uptake. Pharmacokinetic and biodistribution studies were performed on rats. The developed nanometeric lipidparticles were able to control the drug release and substantially enhance the Caco-2 as well as SH-5YSY cell permeability. The developed systems not only enhanced the oral bioavailability of the drug, but also offered substantially elevated brain drug levels to that of plain drug. The drug was protected from liver and biological residence was increased, indicating promising potential of the carriers in effective brain delivery of DMF. Enhanced bioavailability and elevated bioresidence of DMF by vitamin-based SLNs provided the evidence for once-a-day delivery potential for DMF in the management of neurological disorders. KEYWORDS: Vitamin D3, vitamin A acetate, SLNs, multiple sclerosis, drug delivery, once-a-day delivery, SH-SY5Y cells

1. INTRODUCTION Oral delivery of pharmaceuticals is always preferred, owing to the inherent benefits of noninvasive nature and patient compliance. Approx. 90% of the active pharmaceutical ingredients (APIs) are being administered by oral route.1 Oral delivery of APIs for neurological disorders is generally limited, due to blood brain barrier and blood-cerebrospinal fluid barrier.2 These barriers are naturally meant to maintain homeostasis and protect the brain from toxins.3 The targeting of molecule to brain is difficult due to endothelial efflux of molecules.4 To overcome these challenges, various strategies have been employed, most of them are based on nanotechnology.5 Variety of nanotechnology-based products like DaunoXome, Doxil and Depocyt are in market indicating the success of the nanomedicine approach.6 A plethora of colloidal carriers like liposomes, polymeric micelles, nanostructured lipidic carriers, and polymeric nanoparticles have been reported for effective brain delivery.7−10 Solid lipid nanoparticles (SLNs) are regarded as one of the © 2017 American Chemical Society

ideal carriers for brain delivery due to biocompatible nature, scalability, and bypassing of first pass metabolism. It has been established by means of various scientific studies that lipid solubility, oral bioavailability, and brain availability of the drugs can be enhanced using SLNs.11,12 Fat soluble vitamins are generally recommended for the management of the neurological disorders, especially in multiple sclerosis (MS).13,14 Vitamin A, E, and D are specially used as immunomodulator in MS. Henceforth, incorporation of these fat-soluble vitamins in the nanocarriers can provide an extra edge over conventional SLNs.15,16 Fumaric acid esters have been marketed since 1994 under the trade name of Fumaderm in Germany for the management of plaque psoriasis.17 Esters of fumaric acid, especially dimethyl fumarate (DMF), has recently been approved by various federal Received: January 27, 2017 Accepted: February 3, 2017 Published: February 3, 2017 1390

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396

Research Article

ACS Chemical Neuroscience

Figure 1. TEM microphotographs of (A) DMF-cholecalciferol SLNs and (B) DMF-retinol acetate SLNs.

agencies for the management of relapsing MS.18 DMF is used to suppress the relapsing of MS in the patients by reforming the new white matter lesions.19 DMF can be regarded as a prodrug, which gets metabolized into monomethyl fumarate (MMF) and fumarate by the esterases.20 Oral DMF has been proved for its efficacy in MS management in DEFINE and CONFIRM trials.19 Litjens et al. performed the oral pharmacokinetic studies and reported instant rise in the MMF concentration in blood, instead of DMF. In one other observation, reduced absorption of DMF with food has been reported. DMF is generally administered with the dose frequency of 240 mg twice or thrice a day by oral routes.21 Recently, Kumar et al. developed nanolipidic carriers of DMF along with tocopherol acetate, and reported enhanced oral absorption, bioavailability, brain distribution and neuronal cellular uptake.22 In the present study, it was envisioned to develop DMFloaded cholecalciferol/retinol acetate-based SLNs for effective brain delivery. The developed SLNs were characterized for particle size, zeta potential, PDI, drug release, morphology, entrapment efficiency, and drug loading. Oral in vivo pharmacokinetic and organ distribution studies were also performed, further supplemented with neuroblastoma cellular uptake studies.

2.2. In Vitro Drug Release Studies. The pattern of drug release from the dialysis bag containing pure drug and the developed SLNs is shown in Figure 2. It is revealed from this

Figure 2. Graphical representation of percent cumulative drug release from pure DMF, DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs.

release profile that sustained drug release was offered by both the SLNs to that of pure DMF. Zero-order drug release flux values from pure DMF, DMF-cholecalciferol SLNs and DMFretinol acetate SLNs were observed to be 72.6, 38.5, and 44.2 μg·h−1·cm−2 for the initial 12 h. The release flux of DMF from the developed system was observed to be significantly retarded to that from the pure drug (p < 0.05). On the other hand, these systems released approximately 91%, 60.02%, and 73.09% drug in the initial 6 h, respectively. Higuchian drug release profile was followed by DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs (please refer to the Supporting Information). Findings of this research are in consonance with earlier reports, as the Higuchian release pattern is the frequently reported drug release profile for the systems promising sustained drug delivery.24,25 2.3. Caco-2 and SH-SY5Y Cellular Uptake Studies. Caco-2 cellular permeability studies of pure coumarin-6 dye, DMF-cholecalciferol SLNs, and DMF-retinol acetate SLNs were performed to understand the intestinal absorption at cellular level.26,27 Plain dye was unable to penetrate the cells, while coumarin-6 tagged formulations were able to penetrate the cells, as shown in Figure 3. Blue color inside the cells is due to the fluorescence from DAPI, and green florescence indicates the coumarin-6 presence inside the cells. It can be concluded that DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs

2. RESULTS AND DISCUSSION 2.1. Physicochemical Characterization. DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs were found to possess average size of 118.8 and 198.7 nm, respectively. PDI and zeta potential of the DMF-cholecalciferol SLNs and DMFretinol acetate SLNs were observed to be 0.338 and 0.608, and −1.96 and −0.309 mV, respectively. Lower particle size and zeta potential assured prolonged circulation and better brain bioavailability, with a promise to bypass mononuclear phagocytic system.23 As shown in Figure 1, both kinds of SLNs were spherical in shape with smooth surface. The microphotographs also assured segregated nature of the nanoparticles. Percent of entrapment efficiency (EE) of DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs was observed to be 83.99 ± 3.36% and 88.22 ± 3.97%, respectively, and percent of drug loading (DL) was found to be 20.99 ± 0.84% and 22.06 ± 0.88, respectively. Higher entrapment and drug loading assured better drug carrying capacity of SLNs to the desired organ.11 1391

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396

Research Article

ACS Chemical Neuroscience

Figure 3. Confocal laser scanning microphotographs (60×) of (A) control with DAPI-stained nuclei, (B) plain dye treated group, (C) DMFcholecalciferol SLNs, and (D) DMF-retinol acetate SLNs.

laser scanning microscopic photographs confirmed that DAPI dye was able to stain the cell nuclei, but coumarin-6 dye was not able to cross the neuroblastoma cells. On the contrary, coumarin-6 tagged DMF-cholecalciferol SLNs and DMFretinol acetate SLNs were able to access the cell cytoplasm as well as cell nuclei, as shown in Figure 5. The neuroblastoma uptake of dye tagged DMF-cholecalciferol SLNs and DMFretinol acetate SLNs was observed to be enhanced by 52.84 times and 68.68 times, respectively to that of plain dye. On the other hand, uptake of dye tagged DMF-cholecalciferol SLNs was found to be 1.30 times higher than that of dye-tagged DMF-retinol acetate loaded SLNs, as shown in Figure 6. Better in vitro uptake of the dye-tagged nanocarriers inferred enhanced in vivo neuronal uptake.22

were able to cross cell cytoplasm as well as cell nuclei. DMFcholecalciferol SLNs and DMF-retinol acetate SLNs were observed to offer enhanced Caco-2 cellular permeability by 15.22 and 10.87 times to that of plain dye, as shown in Figure 4.

Figure 4. Graphical representation of the Caco-2 cellular permeability of the pure dye, DMF-cholecalciferol SLNs, and DMF-retinol acetate SLNs. Asterisk (*) indicates the statistical significant difference in t test at p < 0.001 from the pure dye group.

Elevated Caco-2 cellular permeability indicates the better lacteal absorption of developed SLNs. The plausible reason for higher Caco-2 cellular permeability might be the nanometric size range and biocompatible nature of the carrier composed of cholecalciferol and retinol acetate.28,29 SH-SY5Y cells, human neuroblastoma cells, were used to understand the neuronal uptake as an in vitro model. Confocal

Figure 6. Graphical representation of the SH-SY5Y cellular permeability of the pure dye, DMF-cholecalciferol SLNs, and DMFretinol acetate SLNs. Asterisk (*) indicates the statistical significant difference in t test at p < 0.001 to that of plain dye.

Figure 5. Confocal laser scanning microphotographs of SH-SY5Y cells (60×) of (A) control with DAPI-stained nuclei, (B) DMF-cholecalciferol SLNs, and (D) DMF-retinol acetate SLNs. 1392

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396

Research Article

ACS Chemical Neuroscience 2.4. Oral in Vivo Pharmacokinetic Studies. Oral in vivo pharmacokinetic studies of pure DMF, DMF-cholecalciferol SLNs, and DMF-retinol acetate SLNs were performed on Wistar rats. Graphical representation between plasma MMF concentration and time is shown in Figure 7. It can be observed

model was employed to determine various pharmacokinetic parameters, and the parameters have been compiled in Table 2. The brain bioavailability was enhanced by 16.74-fold and 19.51fold by DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs, respectively, in comparison to that of plain drug. The findings of reduced drug availability in the liver tissues by both of the SLNs supported the liver bypassing phenomenon, as hypothesized in pharmacokinetic studies. Both of the nanocarriers were observed to enhance the brain biological half-life of the order of 1.8 times (DMF-cholecalciferol SLNs) and 1.5 times (DMF-retinol acetate SLNs) and also evaluate the Cmax values in brain. As envisioned, both of the developed systems were able to enhance the brain delivery of DMF; however, performance of the DMF-retinol acetate SLNs was observed to be slightly superior to that of DMF-cholecalciferol SLNs, as in pharmacokinetic studies.

3. CONCLUSIONS Our findings report first time combination of cholecalciferol and retinol acetate in the SLNs for better brain delivery of DMF. DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs have now been established for better brain delivery by in vitro and in vivo modes. Developed formulations not only enhanced oral bioavailability, but also elevated the biological half-life, which is desired parameter to develop once-a-day oral product. The developed scalable nanoparticles with capability to enhance the Cmax, bioavailability, and biological residence time in the central compartment as well as brain provide a scientific evidence for an effective once-a-day formulation of DMF for the management of neurological disorder. DMFcholecalciferol loaded SLNs and DMF-retinol acetate loaded SLNs have been established and proves once-a-day dose concept in the neurological disorders. Developed Present findings open a future platform for researchers for better delivery of DMF along with neuroprotectives like cholecalciferol and retinol acetate.

Figure 7. Graphical depiction of MMF plasma concentration as a function of time in animals: pure DMF, DMF-cholecalciferol SLNs, and DMF-retinol acetate SLNs.

from Figure 7 that the plasma drug concentration in the group receiving DMF-retinol acetate was higher than that of the group administered DMF-cholecalciferol SLNs; however, both of the SLNs were able to maintain higher plasma levels vis-à-vis plain DMF group. Bioavailability of DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs were enhanced by 4.39 and 5.09 times, respectively. Cmax from the DMF-cholecalciferol SLNs group and the DMF-retinol acetate SLNs group was elevated by 2.16-fold and 1.97-fold, respectively, in comparision to that of the pure DMF. Biological half-life for DMF-cholecalciferol SLNs group and DMF-retinol acetate SLNs group was enhanced by 1.86-fold and 1.79-fold, respectively, in comparison to that of the pure DMF group. Substantially enhanced in Cmax and AUC, it is inferred the absorption phenomenon involving bypassing of first-pass metabolism. Interestingly, Vd, tmax, and Cl were reduced by the developed formulations than the pure DMF, as shown in Table 1. The findings are unique in nature, as it shows that liver metabolism was bypassed, resulting in enhanced biological half-life, decreased volume of distribution, and longer systemic circulation. All these pharmacokinetic outcomes promise an effective once-a-day DMF formulation based on these vitamin-derived SLNs.30 2.5. Biodistribution Studies. Biodistribution of MMF in liver, kidney and heart were evaluated at different time points to understand the sojourn of drug in the biological milieu. The amount of drug at various time points available in liver, heart, and brain is shown in Figure 8. One CBM oral pharmacokinetic

4. METHODS 4.1. Materials. Phospholipid 90G (PL90G) was provided as an ex gratis sample from Messrs (M/s) Phospholipid GmbH, Nattermannallee, Germany. Monomethyl fumarate (MMF) and dimethyl fumarate (DMF) were purchased from M/s Sigma-Aldrich, Banglore, India. Palmitic acid and sodium hydroxide were purchased from M/s Molychem, Mumbai, India. Stearic acid, potassium dihydrogen phosphate, disodium hydrogen ortho phosphate, dipotassium hydrogen ortho phosphate, and hydrochloric acid were procured from M/s Central Drug House, New Delhi, India. Methanol, water and acetonitrile were bought from M/s Spectrochem Pvt. Limited, Mumbai, India. Polysorbate 80 and minimum essential medium (Gibco) was delivered by M/s Fisher Scientific India Pvt. Limited,

Table 1. Tabular Representation of the Various in Vivo Pharmacokinetic Parametersa

a

parameters (unit)

pure DMF

DMF-cholecalciferol SLNs

DMF-retinol acetate SLNs

−1 [AUC]∞ 0 (ng·mL ·h) −1 Cmax (ng·mL ) Tmax (h) K (h‑1) Ka (h‑1) T1/2 (h) Vd (mL) Cl (h‑1·mL)

7529.05 1201.60 1.53 0.19 1.52 3.47 4.59 × 10−1 9.17 × 10−2

33 050* 2368.02* 1.89* 0.11* 1.49 6.20* 2.5 × 10−1* 2.87 × 10−2*

38 319.23* 2598.18* 1.38* 0.11* 2.34* 6.47* 2.4 × 10−1* 2.67 × 10−2*

Asterisk (*) indicates statistically significant difference from plain DMF group at P < 0.05. 1393

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396

Research Article

ACS Chemical Neuroscience

Figure 8. Biodistribution of MMF in brain, liver, and heart in the studied Wistar rats group (n = 6). maintained at 37 °C. After completion of the dialysis, the release medium was filtered and quantified for unentrapped drug by RPHPLC method. The mobile phase was composed of acetonitrile and water (60:40 v/v) with a flow rate of 1 mL/min. The chromatogram was recorded at 216 nm, using C18 column on an autosampler HPLC system (LC-2010C HT, M/s Schimadzu Co. Ltd., Chiyoda-ku, Tokyo, Japan).33 4.2.3. In Vitro Drug Release Studies. In vitro drug release studies of the pure DMF, DMF-cholecalciferol SLNs, and DMF-retinol acetate SLNs were performed using the dialysis bag method. Pure DMF (2.5 mg/mL), DMF-cholecalciferol SLNs, and DMF-retinol acetate SLNs (1 mL; equivalent to 2.5 mg of DMF) were sealed in separate dialysis bags. The sealed dialysis bag were kept in 30 mL of 0.1 N HCl for 2 h and than in phosphate buffered saline (PBS) pH 6.8 for further 24 h. Samples were withdrawn at predetermined time intervals of 0, 0.08, 0.17, 0.25, 0.50, 0.75, 1, 2, 4, 6, 12, and 24 h, and the sink condition was maintained by addition of fresh medium, post each sample.27 The samples were quantified by RP-HPLC.34 4.2.4. Caco-2 and SH-SY5Y Cellular Uptake Studies. Coumarin-6 dye was added in the DMF-cholecalciferol SLNs and DMF-retinol acetate SLNs by physical mixing process.13 Dye-tagged formulations were incubated with Caco-2/SH-SY5Y cells until 2 h at 37 °C. PBS pH 7.4 was used for the washing of cells. DAPI solution of 300 nM was used to stain the nuclei of the fixed cells. The excess dye was washed, and the cells were observed under a confocal laser scanning microscope (CLSM; Nikon C2 Plus, with NIS Elements version 4.3 Software, M/s NIKON Instruments INC., Melville, NY), installed at UGC-Centre of Excellence in Applications of Nanomaterials, Nanoparticles & Nanocomposites, Panjab University, Chandigarh, India for the florescence intensity. Florescence intensity was recorded at emission wavelengths of 500−550 nm and 417−477 nm, and excitation wavelengths of 488 and 405 nm, respectively for coumarin-6 and DAPI.27 4.2.5. In Vivo Pharmacokinetic Studies. Wistar rats (male/female, 200−250 g, 4−6 weeks old; n = 6) were used to perform oral in vivo pharmacokinetic studies. The respective oral dose (Pure DMF, DMFcholecalciferol SLNs and DMF-retinol acetate SLNs; equivalent to 3 mg/kg of DMF) was administered using oral cannula. Wistar rats were anaesthetized and blood (0.3 mL) was collected in heparin coated vials from retro-orbital plexus at different time intervals (0, 0.25, 0.5, 0.75, 1, 1.5, 2, 4, 6, 8, 10, and 12 h). Blood was centrifuged and the supernatant was mixed with 0.2 mL of methyl tert-butyl ether to precipitate the plasma proteins. pH of the plasma was adjusted to 8.0 or 8.5 and air-dried at 40 °C. The sample was reconstituted in mobile

Mumbai, India. Cetyl pamitate, cholecalciferol, retinol acetate and dialysis bag were obtained from M/s Himedia Laboratories Pvt. Limited, Nashik, India. Ethanol was bought from M/s Jai Chemicals and Pharma Works, Jaipur, India. Diethyl ether was supplied by M/s S D Fine Chemicals Limited, Ambala, Haryana. The Caco-2 cell line (human colon adenocarcinoma cells) was purchased from National Centre for Cell Science (NCCS), Pune, India. SH-SY5Y cells were provided by National Institute of Immunology, New Delhi. Oyster BDS premium C18 (250 × 4.6, 5 μm; batch no. 43/053) HPLC column was purchased from M/s Merck Specialties Pvt. Ltd., Mumbai, India. Reagents and chemicals are being used of analytical grade and used as such. 4.2. Experimental Methods. 4.2.1. Preparation of DMF-Loaded Vitamin-Based SLNs. Hot microemulsification technique was employed to formulate the DMF-loaded cholecalciferol-based SLNs and DMF-loaded retinol acetate-based SLNs. Stearic acid and vitamins (cholecalciferol/retinol acetate) in mass ratio of 70:30 were placed in a preheated beaker at 70 °C to give a clear solution. DMF (250 mg) was weighed and transferred into the transparent lipid. PL 90G (442 mg) was dispersed in double distilled water (14 g) until a white milky dispersion was obtained. Polysorbate 80 (4.55 g) was added dropwise to the milky dispersion. This aqueous phase was heated to 70 °C and isothermally transferred dropwise into the lipidic phase. The mixture was heated at 70 °C to give a clear microemulsion. The hot microemulsion was poured into 80 mL of saline, maintained at 2−4 °C, and stirred at 2000 ± 50 rpm for 30 min. The developed SLNs were kept in a refrigerator, until further use.31 4.2.2. Physiochemical Characterization. A Malvern Zetasizer instrument (Nano ZS, M/s Malvern Instruments Limited, Worcestershire, UK) installed at Dr. S. S. Bhatnagar University Institute of Chemical Engineering & Technology, Panjab University, Chandigarh, India was employed to determine size, polydispersity index (PDI), and zeta potential of the developed systems. Final results of the samples were reported as the average of three successive observations. A transmission electron microscope (TEM; Hitachi H7000, Tokyo, Japan) was used to explore the surface morphology of the developed formulations, which is installed at Central Instrumental Laboratory, Panjab University, Chandigarh. The samples were negatively stained with 1% phosphotungistic acid on the copper grids. Microphotographs were captured at suitable magnification.32 For determination of entrapment efficiency (EE) and drug loading (DL), the dialysis bag method was used. SLNs (1 mL, equivalent to 2.5 mg of DMF) were packed in a dialysis bag and kept in 30 mL of methanol for 2 h stirring. The system was continuously stirred and 1394

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396

Research Article

phase. All samples were filtered using 0.22 μm nylon syringe filter and analyzed for MMF content employing RP-HPLC. One CBM per oral pharmacokinetic approach was used to determine the pharmacokinetic parameters like area under curve (AUC), volume of distribution (Vd), biological half-life (t1/2), elimination rate (K), and clearance (Cl), employing MS Office Excel software.22 4.2.6. Biodistribution Studies. Wistar rats (Male/Female, 200−250 g, 4−6 weeks old; n = 6) were employed to determine the drug content in brain, liver, and heart at various time points. Dose administration was analogous to the pharmacokinetic studies. Animals were sacrificed at each sampling time by cervical dislocation. Harvesting of brain, liver, and heart was done, and the tissues were scrapped off with tissue paper to remove the sticky materials. Brain, liver, and heart were separately meshed in saline using a homogenizer. Homogenized organs were added into the acetonitrile and centrifuged at 15 000g. Sample processing and analysis was done, as per the method reported in pharmacokinetic studies.34 4.2.7. Statistical Analyses. Data represented in this paper are the average values of the replicates with standard deviation, and the level of significance was fixed at p < 0.05, unless stated.

0.18 0.34* 2728.74* 3248.89* 3.94* 67.41* 0.67* 2.14* 3.75 × 10−1*



ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acschemneuro.7b00041. Results of model fitting for the data obtained from release studies and results of model fitting for the data obtained from organ biodistribution studies (PDF) Asterisk (*) indicates statistically significant difference from the plain DMF receiving group at P < 0.05.



AUTHOR INFORMATION

Corresponding Author

*E-mail: [email protected]; [email protected]. ORCID

Kaisar Raza: 0000-0001-8159-8005 Author Contributions

P.K. designed, executed the experiments and prepared the manuscript; O.P.K. and G.S. helped in pharmacokinetic studies; R.K. and B.S. supported in cellular uptake studies; K.R. and R.M. supervised in hypothesis building, experimentation, and final drafting of the manuscript, and also arranged the chemicals and facilities, required for the experimentation. Funding

University Grants Commission (UGC), New Delhi provided partial contingent support and senior research fellowship to Pramod Kumar, in the form of UGC-National Fellowship (F./ 2014-15/NFO-2014-15-OBC-RAJ-8108/(SA-III/Web site). The financial support availed from UGC, New Delhi in the form of Start-Up-Grant to K.R. is also duly acknowledged. Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS M/s Phospholipid GmbH, Nattermannallee, Germany is being acknowledged for ex gratis supply of phospholipids. Dr. S. Gopalan Sampathkumar, National Institute of Immunology, New Delhi, India is humbly acknowledged for the kind support in the form of neuroblastoma cells and subsequent culture guidance.

a

0.15 0.38* 2073.55* 2548.53* 4.48* 73.64* 0.88* 2.68* 4.14 × 10−1* 0.17 0.22 3912 4484.20 4.02 10.06 0.24 3.66 6.31 × 10−1 0.02* 1.72* 1075.76* 4669.12* 30.70* 95.44* 4.34* 7.21* 1.62x 10−1* 0.05* 1.38* 9995.69* 24 281.40* 11.68* 1291.80* 3.31* 5.03 × 10−1* 2.55 × 10−2* 0.05* 0.81* 8701.03* 20 832.95* 13.93* 889.37* 2.79* 6.87 × 10−1* 3.42x 10−2* K (h‑1) Ka h‑1) ‑1 [AUC]12h 0 (ng·mL ·h) ‑1 [AUC]∞ 0 (ng·mL ·h) T1/2 (h) Cmax (ng·mL−1) Tmax (h) Vd (mL) Cl (mL·h‑1) 1 2 3 4. 5 6 7 8 9

0.09 0.68 770.06 1244.43 7.79 72.58 3.43 7.6 6.77 × 10−1

0.28 1.62 2261.19 2336.55 2.49 731.26 1.76 6.86 × 10−1 1.91 × 10−1

0.12* 0.57* 1813.91* 2508.49* 5.71* 151.82* 1.55* 2.61* 3.17 × 10−1*

DMF-cholecalciferol SLNs pure DMF DMF-retinol acetate SLNs DMF-cholecalciferol SLNs DMF-retinol acetate SLNs DMF-cholecalciferol SLNs parameters S. no.

pure DMF

pure DMF

heart brain

Table 2. Tabular Representation of the Pharmacokinetic Parameters for Kidney, Liver, and Brain of Wistar Ratsa

liver

DMF-retinol acetate SLNs

ACS Chemical Neuroscience

1395

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396

Research Article

ACS Chemical Neuroscience



fumarate employing tocopherol acetate-based nanolipidic carriers: Evidences from pharmacokinetic, biodistribution and cellular uptake studies. ACS Chem. Neurosci., DOI: 10.1021/acschemneuro.6b00428. (23) Kulkarni, S. A., and Feng, S.-S. (2013) Effects of Particle Size and Surface Modification on Cellular Uptake and Biodistribution of Polymeric Nanoparticles for Drug Delivery. Pharm. Res. 30, 2512− 2522. (24) Dash, S., Murthy, P. N., Nath, L., and Chowdhury, P. (2010) Kinetic modeling on drug release from controlled drug delivery systems. Acta Polym. Pharm. 67, 217−223. (25) Madhwi Kumar, R., Kumar, P., Singh, B., Sharma, G., Katare, O. P., and Raza, K. (2017) In vivo pharmacokinetic studies and intracellular delivery of methotrexate by means of glycine-tethered PLGA-based polymeric micelles. Int. J. Pharm. 519, 138−144. (26) Angelis, I. D., and Turco, L. (2011) Caco-2 cells as a model for intestinal absorption. Curr. Protoc. Toxicol., 20.6. (27) Kumar, P., Kumar, R., Singh, B., Malik, R., Sharma, G., Chitkara, D., Katare, O. P., and Raza, K. (2016) Biocompatible phospholipidbased mixed micelles for tamoxifen delivery: Promising evidences from in -vitro anticancer activity and dermatokinetic studies. AAPS PharmSciTech, DOI: 10.1208/s12249-016-0681-1. (28) Van Breemen, R. B., and Li, Y. (2005) Caco-2 cell permeability assays to measure drug absorption. Expert Opin. Drug Metab. Toxicol. 1, 175−185. (29) Beg, S., Raza, K., Kumar, R., Chadha, R., Katare, O. P., and Singh, B. (2016) Improved intestinal lymphatic drug targeting via phospholipid complex-loaded nanolipospheres of rosuvastatin calcium. RSC Adv. 6, 8173−8187. (30) Raza, K., Kumar, D., Kiran, C., Kumar, M., Guru, S. K., Kumar, P., Arora, S., Sharma, G., Bhushan, S., and Katare, O. P. (2016) Conjugation of docetaxel with multiwalled carbon nanotubes and codelivery with piperine: Implications on pharmacokinetic profile and anticancer activity. Mol. Pharmaceutics 13, 2423−32. (31) Raza, K., Singh, B., Singla, S., Wadhwa, S., Garg, B., Chhibber, S., and Katare, O. P. (2013) Nanocolloidal carriers of isotretinoin: antimicrobial activity against Propionibacterium acnes and dermatokinetic modeling. Mol. Pharmaceutics 10, 1958−1963. (32) Kumar, P., Sharma, G., Kumar, R., Singh, B., Malik, R., Katare, O. P., and Raza, K. (2016) Promises of a biocompatible nanocarrier in improved brain delivery of quercetin: Biochemical, pharmacokinetic and biodistribution evidences. Int. J. Pharm. 515, 307−314. (33) Raza, K., Kumar, N., Misra, C., Kaushik, L., Guru, S. K., Kumar, P., Malik, R., Bhushan, S., and Katare, O. P. (2016) Dextran-PLGAloaded docetaxel micelles with enhanced cytotoxicity and better pharmacokinetic profile. Int. J. Biol. Macromol. 88, 206−212. (34) Gennari, O., Montesano, D., Salzano, A., Albrizio, S., and Grumetto, L. (2011) Determination of dimethyl fumarate in desiccant and antimould sachets by reversed-phase liquid chromatography. Biomed. Chromatogr. 25, 1315−1318.

REFERENCES

(1) Das, S., and Chaudhury, A. (2011) Recent advances in lipid nanoparticle formulations with solid matrix for oral drug delivery. AAPS PharmSciTech 12, 62−76. (2) Banks, W. A. (2016) From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nat. Rev. Drug Discovery 15, 275−292. (3) Abbott, N. J., Patabendige, A. A. K., Dolman, D. E. M., Yusof, S. R., and Begley, D. J. (2010) Structure and function of the blood-brain barrier. Neurobiol. Dis. 37, 13−25. (4) Upadhyay, R. K. (2014) Drug Delivery Systems, CNS Protection, and the Blood Brain Barrier. BioMed Res. Int. 2014, 869269. (5) Scherrmann, J. M. (2002) Drug delivery to brain via the bloodbrain barrier. Vasc. Pharmacol. 38, 349−354. (6) Pillai, G. (2014) Nanomedicines for Cancer Therapy: An Update of FDA Approved and Those under Various Stages of Development. SOJPPS 1, 13. (7) Garcia-Garcia, E., Andrieux, K., Gil, S., and Couvreur, P. (2005) Colloidal carriers and blood-brain barrier (BBB) translocation: a way to deliver drugs to the brain? Int. J. Pharm. 298, 274−292. (8) Kumar, P., Raza, K., Kaushik, L., Malik, R., Arora, S., and Katare, O. P. (2016) Role of Colloidal Drug Delivery Carriers in Taxanemediated Chemotherapy: A Review. Curr. Pharm. Des. 22, 5127−5143. (9) Tseng, Y.-Y., Kao, Y.-C., Liao, J.-Y., Chen, W.-A., and Liu, S.-J. (2013) Biodegradable drug-eluting poly[lactic-co-glycol acid] nanofibers for the sustainable delivery of vancomycin to brain tissue: in vitro and in vivo studies. ACS Chem. Neurosci. 4, 1314−1321. (10) Kundu, P., Das, M., Tripathy, K., and Sahoo, S. K. (2016) Delivery of Dual Drug Loaded Lipid Based Nanoparticles across the Blood-Brain Barrier Impart Enhanced Neuroprotection in a Rotenone Induced Mouse Model of Parkinson’s Disease. ACS Chem. Neurosci. 7, 1658−1670. (11) Blasi, P., Giovagnoli, S., Schoubben, A., Ricci, M., and Rossi, C. (2007) Solid lipid nanoparticles for targeted brain drug delivery. Adv. Drug Delivery Rev. 59, 454−477. (12) Kaur, I. P., Bhandari, R., Bhandari, S., and Kakkar, V. (2008) Potential of solid lipid nanoparticles in brain targeting. J. Controlled Release 127, 97−109. (13) Torkildsen, Ø., Løken-Amsrud, K. I., Wergeland, S., Myhr, K.M., and Holmøy, T. (2013) Fat-soluble vitamins as disease modulators in multiple sclerosis. Acta Neurol. Scand. Suppl. 127, 16−23. (14) DeLuca, G. C., Kimball, S. M., Kolasinski, J., Ramagopalan, S. V., and Ebers, G. C. (2013) Review: the role of vitamin D in nervous system health and disease. Neuropathol. Appl. Neurobiol. 39, 458−484. (15) Bitarafan, S., Saboor-Yaraghi, A., Sahraian, M.-A., Nafissi, S., Togha, M., Beladi Moghadam, N., Roostaei, T., Siassi, F., Eshraghian, M.-R., Ghanaati, H., Jafarirad, S., Rafiei, B., and Harirchian, M.-H. (2015) Impact of Vitamin A Supplementation on Disease Progression in Patients with Multiple Sclerosis. Arch. Iran. Med. 18, 435−440. (16) Anonymous (2010) Vitamin D: hope on the horizon for MS prevention? Lancet Neurol. 9, 555. (17) Mrowietz, U., Altmeyer, P., Bieber, T., Röcken, M., Schopf, R. E., and Sterry, W. (2007) Treatment of psoriasis with fumaric acid esters (Fumaderm). J. Dtsch. Dermatol. Ges. 5, 716−717. (18) Linker, R. A., and Gold, R. (2013) Dimethyl fumarate for treatment of multiple sclerosis: mechanism of action, effectiveness, and side effects. Curr. Neurol. Neurosci. Rep. 13, 394. (19) Papadopoulou, A., D’Souza, M., Kappos, L., and Yaldizli, Ö . (2010) Dimethyl fumarate for multiple sclerosis. Expert Opin. Invest. Drugs 19, 1603−1612. (20) Bomprezzi, R. (2015) Dimethyl fumarate in the treatment of relapsing−remitting multiple sclerosis: an overview. Ther. Adv. Neurol. Disord. 8, 20−30. (21) Litjens, N. H. R., Burggraaf, J., van Strijen, E., van Gulpen, C., Mattie, H., Schoemaker, R. C., van Dissel, J. T., Thio, H. B., and Nibbering, P. H. (2004) Pharmacokinetics of oral fumarates in healthy subjects. Br. J. Clin. Pharmacol. 58, 429−432. (22) Kumar, P., Sharma, G., Kumar, R., Malik, R., Singh, B., Katare, O. P., and Raza, K. (2017) Enhanced brain delivery of dimethyl



NOTE ADDED AFTER ISSUE PUBLICATION This paper was published on February 14, 2017 with an incorrect Figure 7. The corrected version was published on the Web on September 7, 2017. An Addition and Correction was published in volume 8, issue 9.

1396

DOI: 10.1021/acschemneuro.7b00041 ACS Chem. Neurosci. 2017, 8, 1390−1396