This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
The incidence of cancerous disease recently increased in recent years, impacting the physical, mental, and social life of humans. The incidence varies from developed countries (1 to 2%) to fewer development countries (almost 5%) yearly. The estimate also showed that more than 7 million people die from cancer disease and is predicted to be 10 to 15 million new cases added by 2020. Meanwhile, this disease is an unexceptional malignant breast cancer found in women, with more than 1 million fresh cases added yearly [1]. Breast cancer was ranked as number 1 due to the number in Indian women with an age rate as high 25.8 per 100,000 and a mortality rate of 2.7 per 100,000 women [2]. There are lots of chemotherapy drugs available for breast cancer; however, Ribociclib (RBO) is one of them that has been recently approved by the US FDA, and it is given orally of 200 mg as shown in Figure 1. The mechanism of action of RBO is to hamper the activity of cyclin-dependent kinase (CDK) at different types of cyclins like CDK4 and CDK6 of cell cycle pathways which indirectly inhibit the retinoblastoma (Rb) protein phosphorylation. The hampering of Rb again slows down the CDK-mediated G1-S phase movement. Because of this, the cell cycle is apprehended, inhibiting the cancer cell growth [3, 4]. The RBO is associated with various adverse effects, and the most common reaction, with more than 20% incidence, is neutropenia, nausea, fatigue, diarrhea, leukopenia, alopecia, vomiting, constipation, headache, and back pain. Apart from adverse effect, the drug is also associated with low solubility and permeability as it is a BCS class IV drug and undergoes immense metabolism by hepatic cells mediated via CYP3A4 in the human liver. Ribociclib is also the substrate for P-gp efflux, which leads to an increase in the dose to achieve the therapeutic level [5]. Thus, these major factors which decrease the degree of absorption of Ribociclib lead to its poor in vivo prospects. The clinical efficacy of Ribociclib is not satisfactory at the therapeutic level, which often results in an increase in dose frequency, or high dose causes adverse effects [6]. However, being a recently approved drug (RBO), not much work has been done so far in the nanocarrier system to overcome the related issue. A recent study investigated by Fei and Yoosefian on the preparation of RBO micelles to increase the aqueous solubility of hydrophobic drug carried out using dodecylphosphocholine surfactant [7]. The remaining challenge still to overcome is the bioavailability issue by inhibiting P-gp efflux and bypassing the extensive first-pass metabolism [8]. The lipidic nanocarriers are being developed in order to overcome the predicaments like improving solubility, minimization of P-gp efflux, protection of the drug from enzymatic (CYP3A4) degradation, providing metabolic stability (stability in GIT), enhancing permeability and retention in the target tissue, enhancing the bioavailability, and minimization of toxic drug effects [9]. The drug integrated into nanostructured lipidic carrier (NLCs) formulations with specific lipid (Solid and Liquid lipids) and excipients can regulate P-gp-mediated efflux movement and has the potential to change the pharmacokinetics of Ribociclib largely. In addition, lipids used in Ribociclib-loaded NLCs will promote its systemic blood circulation via the intestinal lymphatic system rather than being directly absorbed into the portal circulation and consequently prevent its significant first-pass metabolism [10]. Thus, our objective is to develop NLCs as a formulation which exhibits great potential for improving and normalizing the absorption of Ribociclib and contributing significantly in enhancing its therapeutic efficacy up to a satisfactory level.
[figure omitted; refer to PDF]
Based on solubility data, Labrafil M2125 CS (HLB 4), Capmul MCM C8 (HLB 5-6), Maisine 3S1 (HLB 4), and Plurol oleique (HLB 6) were showing high solubility of RBO as it was anticipated that the structure containing carboxylic acid and succinate group favors the solubility of RBO in each liquid lipid. In addition, longer triglycerides have shown more solubility than the medium-chain due to more entrapment of drugs. Similarly, Stearic acid (HLB 17) and Capmul MCM C10 (HLB 5) have shown high solubility of RBO due to similar reasons. The solubility parameters of each lipid direct reflect the encapsulation efficiency. Thus, the high solubility associated with the drug in lipids is also attributed to the high encapsulation efficiency of the NLCs formulation [29]. Bang and coworkers investigated NLCs of anticancer drug paclitaxel, and similar methods were applied for the selection of solid and liquid lipid for the NLCs preparation [30, 31].
4.1.2. Compatibility Study of Lipids
The results of compatibility studies are given in Table 3. Labrafil M 2125 CS, Capmul MCM- C8, Maisine 351, Plurol oleique were selected as liquid lipids, and Stearic acid and Capmul MCM C10 were selected as solid lipids for further compatibility/miscibility study among lipids. Negi and coworkers develop a protocol for solid and liquid lipid-based compatibility on miscibility/affinity and the physical compatibility of lipids. So based on these parameters, Capmul MCM C8 and Compritol were selected [31].
Table 3
Results of miscibility study between liquid lipids and solid lipids.
Solid lipid | Liquid lipid | Observation | Inferences |
Stearic acid | Labrafil M 2125 CS | After 48 h, separate layers were observed. | Failed |
Capmul MCM-C8 | Uniform distribution and no phase separation were observed. | Good compatibility | |
Maisine 351 | Turbidity and phase separation was found after 48 h. | Failed | |
Plurol oleique | |||
Capmul MCM C10 | Labrafil M 2125 CS | No phase separation was observed. | Passed |
Capmul MCM-C8 | |||
Maisine 351 | After 48 h, all remain in the liquid phase. | Failed | |
Plurol oleique |
4.1.3. Thermal Inspection of a Binary Mixture by DSC
In a ratio of the binary mixture, as the % of liquid lipid increases by 10%, the enthalpy for the melting point decreases in respect of the solid lipid (100%) as shown in Figure 4. However, the crystallinity index (CI) has less variability in the ratio of 8 : 2 to 6 : 4 compared to 9 : 1. Therefore, the final ratio was selected based upon CI, i.e., 6 : 4, as shown in Figure 5 [32]. Iqbal and associates reported similar parameters for the selection of the best ratio of solid and liquid lipids for a suitable binary mixture. As the % of the liquid lipid (Sefsol 218) increases, the enthalpy of the binary mixture decreases with respect to the solid lipid (Geleol), and it was accompanied by a decrease in crystallinity index from 100.0 to 30.3% [13].
[figure omitted; refer to PDF][figure omitted; refer to PDF]4.1.4. Selection of Surfactant
The surfactants, namely, Cremophor RH 40, Cremophor EL, Tween 20, Tween 80, Poloxamer 188, and Solutol HS15, were used to evaluate for its emulsification property. The result of the screening is illustrated in Table 4. The higher emulsification property reflects to reduce the particle size of binary mixture lipids and increase the physical stability of lipid nanoparticles, sequentially preventing the aggregation of nanoparticles which gives higher transmittance [33]. Alam and coworkers developed isradipine NLCs. They have reported in their research about the surfactant selection criteria based on % of transmittance, which was examined by UV spectroscopy method at 510 nm [12].
Table 4
Observational study for the selection of surfactants.
Name of surfactants | % transmittance |
Cremophor EL | 64.23 |
Cremophor RH 40 | 91.61 |
Poloxamer 188 | 92.04 |
Solutol HS 15 | 98.28 |
Tween 20 | 45.95 |
Tween 80 | 63.69 |
4.2. Preparation and Optimization of NLCs
With regard to approaches to Pharmaceutical Development, the product should be designed to meet needs and the intended product performance. A researcher might select either an experimental approach, an organized approach, or both for product development. A more organized approach for product development also refers to the quality by design approach, which includes adding prior knowledge and their results using the design of experiments (DOE). Such an organized approach helps to get the final desired product with the desired quality. It also helps to understand all the regulations. With respect to the quality of the product, which is generally related to safety and efficacy, setup parameters were justified, which refers to the quality target product profile (QTPP) which gives the outlook summary of drug product characterization, which can be predetermined. The dependent variables like particle size, entrapment efficiency, and PDI are considered critical quality attributes (CQAs). For effective delivery at the targeted site, the particle size should be small; more entrapment of the drug leads to therapeutic efficacy even at a low dose and low PDI (≤0.7) [34] which is considered as a uniformly distributed formulation. These parameters of QTPP and CQAs are summarized in Tables 5 and 6, respectively [35, 36].
Table 5
Quality target product profile of the nanostructured lipidic carrier.
QTPP parameters | Target to achieve | Justification |
Dosage type | Nanoformulation | Escalate the drug permeation/bioavailability |
Route of administration | Oral | Easy to use/lymphatic uptake/stability/minimize side effects |
Physical state | Lyophilized powder | Easy administration/physical aspect |
Physiochemical characterization | Entrapment efficiency | Drug loading assurance |
Particle size | Influence the permeation and absorption | |
Zeta potential | Product stability assurance | |
Pharmacokinetics | Absorption | Required to achieve the desired efficacy |
Distribution | ||
Metabolism and targeting |
Table 6
List of various CQAs affecting the therapeutic efficacy of RBO-NLCs.
CQA parameters | Target to achieve | Justification |
Particle size | 100-200 nm | Assure the absorption, hence the increase in bioavailability |
Entrapment efficiency | ≥70% | To reach the optimum therapeutic efficacy |
PDI | ≤0.7 | Uniform drug distribution, hence content uniformity |
4.2.1. Risk Assessment
The profound parameters for further experiment for optimization of formulation using Box-Behnken design from preliminary assessed data were binary mixture concentration (1-3%), surfactant concentration (1-3%), and sonication time (1-5 min). The 100% equipped response surface fraction is present in the design space to signify its accuracy all over the region. Figure 6 demonstrated the relation among all factors for NLCs preparation prepared by software Minitab 19, Philadelphia, USA. Similarly, Kovacs and associates have reported for the salicylic acid-loaded NLCs in which they have investigated the risk assessment, which referred to both qualitative and quantitative estimates of the risk. An Ishikawa diagram was drawn to find out the key material and process attributes which affects the NLCs formulation preparation [37].
[figure omitted; refer to PDF]4.2.2. Box-Behnken Design (BBD)
The Box-Behnken design (BBD) response surface method tested the points which are within a predetermined range unlike the central composite design (CCRD) to get the total possible runs based on the optimized NLCs formulation data, which was prepared by probe sonication technique; 17 runs were obtained using the mentioned variables in Table 7. Table 8 gives the result derived from the experimental runs. The optimized formula obtained was a binary mixture, surfactant, and sonication time which were found as 1%
Table 7
Different variables selected for Box-Behnken design.
Independent variables | Factors | Unit | Level | |
Low | High | |||
A1 | Binary mixture | % | 1 | 3 |
A2 | Surfactant | % | 1 | 3 |
A3 | Sonication | Min | 1 | 5 |
Table 8
Experimental runs and observed responses from BBD.
Std | Runs | Factor 1 | Factor 2 | Factor 3 | Response 1 | Response 2 | Response 3 |
Binary mixture (% | Surfactant (% | Sonication time (min) | Particle size (nm) | PDI | Entrapment efficiency (% | ||
14 | 1 | 3 | 1 | 3 | 656 | 0.653 | 94.5 |
13 | 2 | 2 | 2 | 3 | 211.9 | 0.562 | 89.9 |
17 | 3 | 2 | 3 | 5 | 179.87 | 0.554 | 89.99 |
16 | 4 | 1 | 2 | 1 | 230.7 | 0.497 | 87.1 |
1 | 5 | 1 | 1 | 3 | 212.54 | 0.499 | 85.9 |
9 | 6 | 1 | 2 | 5 | 152.32 | 0.618 | 86.1 |
3 | 7 | 2 | 3 | 1 | 221.92 | 0.5 | 87.76 |
15 | 8 | 3 | 3 | 3 | 276.53 | 0.621 | 96.12 |
8 | 9 | 2 | 2 | 3 | 225.9 | 0.549 | 87.98 |
11 | 10 | 2 | 2 | 3 | 234.8 | 0.541 | 89.42 |
7 | 11 | 2 | 1 | 5 | 465.71 | 0.487 | 87.91 |
12 | 12 | 2 | 2 | 3 | 212.7 | 0.502 | 88.4 |
6 | 13 | 1 | 3 | 3 | 114.23 | 0.649 | 87.7 |
4 | 14 | 3 | 2 | 5 | 543.73 | 0.597 | 95.4 |
5 | 15 | 3 | 2 | 1 | 419.34 | 0.682 | 96.71 |
2 | 16 | 2 | 2 | 3 | 211.9 | 0.562 | 89.9 |
10 | 17 | 2 | 1 | 1 | 396.4 | 0.512 | 90.14 |
The quadratic equations obtained for different dependent variables and
Table 9
Interpretation of regression value of dependent variable responses B1, B2, and B3.
Dependent variables | Adequate precision | SD | % CV | |||
Adjusted value | Predicted value | |||||
Particle size (B1) | 0.9983 | 0.9962 | 0.9908 | 74.6866 | 9.27 | 3.17 |
PDI (B2) | 0.9500 | 0.8856 | 0.7640 | 12.2250 | 0.0211 | 3.75 |
EE (B3) | 0.9692 | 0.9297 | 0.7403 | 14.2001 | 0.9234 | 1.03 |
Particle Size (B1) = 219.44,
PDI (B2) = 0.5432,
Entrapment efficiency (B3) = 89.12,
In all the equations,
[figures omitted; refer to PDF]
[figures omitted; refer to PDF]
[figures omitted; refer to PDF]
4.2.3. Impact of Independent Variables on Responses
(1) Impact of the Binary Mixture on Dependent Variables. As the binary mixture concentration increases, the particle size also increases because of agglomeration of particles due to lack of emulsifying agent (surfactant) as more emulsifying agent is required if lipid concentration increases aqueous phase, and lack of it is attributed to an increase in particle size.
Entrapment efficiency was directly affected binary mixture quantity. This indicated that an increase in the lipid concentration leads to an increase in the amount of solid lipid, which can be, accommodated more drugs inside the NLCs formulation in the solid lipid [38]. Similarly, Subedi and associates investigated the Doxorubicin loaded into the lipidic carrier, which stated the factor affecting the quantity of lipid on particle size as well as encapsulation efficiency. So, the increment of the lipid content increases the encapsulation efficiency and particle size [39].
(2) Impact of Surfactant on Dependent Variables. The lipid nanoparticle particle size decreases as the concentration of surfactant increases above the critical micelle concentration due to the decrease in interfacial tension. This critical concentration of surfactant also forms a network on the surface of lipid nanoparticles which prevent its agglomeration of the small particle from forming a bigger particle size.
However, the increase in the concentration of surfactant on entrapment efficiency was found very minimal [40]. The results are in agreement with the research reported by Shtay and associates. They have studied the factorial design, which applied to variables that affecting their preparation for food application using lipid nanocarrier. They have reported the increase in concentration decrease the particle size explained by a decrease in interfacial tension [40].
(3) Impact of Sonication Time on Dependent Variables. The mechanism of probe sonication is based on the principle that high energy waves ultrasonically emitted cause the breakage of lipid nanoparticles into small nanoparticles. Thus, upon an increase in sonication time, the lipid nanoparticle size decreases and forms monodisperse globules because of the shear force generated by cavitation.
However, the impact of sonication on entrapment efficiency was observed to be very minimal with aggravation in the sonication time [41]. Ahad and coworkers formulate the lipidic nanocarrier using an experimental design technique for valsartan. They reported an inverse relationship between the sonication time and vesicle size [42].
4.2.4. Validation of Design of Experimental
The goal of the current study is to achieve minimum particle size with maximum entrapment efficiency. Design-Expert software, version 12.0.3.0 (Minneapolis, USA), was applied for the predicted value of response given in Table 10. Based on independent variable values, RBO-NLCs prepared and compared their data with a predicted response. The value obtained from the experiment was almost similar, which confirms the validity of the optimized process for preparing NLCs except for the particle size, which might be due to the wide variety of particle data obtained from a number of runs which give a broad mean value.
Table 10
Point prediction method to attain predicted and actual response (maximum entrapment efficiency, optimum PDI, and minimum particle size) [12].
Lipid (% | Surfactant (% | Sonication time (min) | Particle size (nm) | Entrapment efficiency (%) | PDI | |
Predicted | 1.0 | 3.0 | 3.0 | 219.44 | 89.12 | 0.543 |
Actual | 117.00 | 87.70 | 0.600 |
4.3. Characterization of Optimized RBO-NLCs
4.3.1. Particle Size, PDI, and Surface Charge
The nanoparticle size (Z-Average) and PDI were obtained at
4.3.2. Drug Entrapment (EE) and Drug Loading (DL)
The % drug entrapment efficiency of NLCs was calculated to be
The % DL was found to be
4.3.3. Structural Analysis by TEM
The TEM image shown in Figure 10 demonstrated that the RBO-NLCs particles were uniformly distributed with no sign of any aggregation of particles, and the shape of each nanoparticle was spherical. The result indicated that the NLCs particle was dispersed homogenously. A similar outcome was evaluated by Alam and coworkers where the NLCs particles were uniform and in spherical shape [45].
[figure omitted; refer to PDF]4.4. Powder X-Ray Diffraction (PXRD) Pattern Study
RBO was found to be crystalline in nature as it showed sharp, intense peaks located at 10.0, 14.8, 15.2, 15.7, 20.1, 22.6, and
[figures omitted; refer to PDF]
4.5. Structural Analysis by FT-IR Spectroscopy
The spectra obtained from FT-IR spectroscopy of drug alone and RBO-NLCs are demonstrated in Figures 12(a) and 12(b), respectively. The spectra of the drug alone have seen typical peaks at 3667.64 N–H stretch, 1727.66 C=O stretches, 1604.90–1458.69 C=C stretches, and 1391.78 C-N stretches. FTIR spectrum of RBO-NLCs showed a peak at 2916 C-H stretch, 2849.51 O-H stretch, and 1699.26 C=O stretch of carboxylic acid, which could be due to the existent of excipients in the NLCs formulation. In the FT-IR spectrum, the typical peak of RBO-NLCs was absent compared to the pure drug because of the complete entrapment of RBO inside the NLCs formulation [51]. Annu et al. have developed the chitosan nanoparticle. The result reported in the FT-IR study stated that the characteristic peaks of the drug in nanoparticles were absent compared to drug alone spectra, and it could be the possible reason for entrapment of the drug within the nanoformulation [52].
[figures omitted; refer to PDF]
4.6. In Vitro Drug Release Study
RBO-NLCs and RBO-SUS shown in Figure 13 indicated a sustained drug release at different pH 1.2, 4.5, and 6.8 from the initial time point as compared to RBO-SUS, which shows a gradual increase in release but not sustained for 24 h. The drug present inside the lipid matrix of NLCs undergoes surface erosion which is attributed to its sustained release from the formulation. Initial burst release in the NLCs was due to the unentrapped drug. The
[figures omitted; refer to PDF]
Table 11
Dissolution profile comparison for NLCs and suspension formulation in different dissolution media.
Parameters | Formulation | 0.1 N HCL (pH 1.2) | AB (pH 4.5) | PBS (pH 6.8) |
%DE | RBO-NLCs | |||
RBO-SUS | ||||
f2 similarity factor | — | 43.89 | 43.76 | 48.52 |
RBO-NLCs | 2.03 | 1.38 | 11.27 | |
RBO-SUS | 2.05 | 1.20 | — | |
RBO-NLCs | 4.47 | 6.53 | — | |
RBO-SUS | 4.49 | 3.53 | — |
Similarly, similarity factor, f2 was calculated. The f2 values below 50 for pH 1.2, 4.5, and 6.8 indicated the both RBO-NLCs and RBO-SUS release pattern were not similar. Sahibzada and coworkers investigated a similar comparison of drug nanoparticle dissolution parameters for the enchantment of oral bioavailability. The %DE and similarity factor f2 were calculated for two methods to prepare nanoparticles and compared for better dissolution [21].
4.7. Mechanism of Drug Release
The kinetic release model in the buffer of pH 1.2, 4.5, and 6.8 was calculated. Based on the
4.8. In Vitro Lipolysis Study
The drug should be solubilized near the absorptive membrane so that it is available for systemic absorption. Therefore, the drug should solubilize in an aqueous layer in lipolysis media, and the same will not happen in the case of the sediment layer. The percentage of drug content in a different layer of aqueous and sediment is depicted in Figure 14. The high solubilization potential of the drug in the NLCs formulation in the aqueous layer indicated higher absorption, i.e., approximately 76% after orally administering the RBO-NLCs formulation. Furthermore, drug content in the sediment phase of RBO-NLCs was also calculated and found to be
4.9. In Vitro Haemolysis Study
Haemolysis is detected in red blood cells treated with experimental agents, as shown in Figure 15. The positive control sample showed the least variability in red blood cells with respect to their structure or shape, which was almost similar to the sample treated with NLCs-placebo as the haemolysis count was significantly high in the case of Triton X100 treated sample which destabilizes erythrocytes membrane. The RBO-NLCs formulation showed well intact red blood cells, which were again similar to the NLCs placebo-treated sample (% haemolysis of 1.855%). However, slight structure modification occurred in the case of the RBO-SUS sample (% haemolysis of 2.712%) treated red blood cells, which disrupts the membrane of red blood cells. The result concluded that the NLCs formulation of RBO was safer compared to a pure drug suspension. So, the result comprises the in vivo fate of NLCs formulation of the selected drug. A similar investigation was reported by Chauhan and associates in which the curcumin nanoformulation evaluated their anticancer potential and evaluated direct nanoformulation erythrocyte interaction in which membrane disruption directly measures the toxicity of nanoformulation [56].
[figures omitted; refer to PDF]
4.10. In Vitro Gut Permeation Study
The intestinal permeability of RBO-NLCs and RBO-SUS is shown in Figure 16. After 2 h study setup, the flux of the drug in the rat intestine was calculated. The flux for RBO-NLCs and RBO-SUS formulations was found to be
4.11. Depth Permeation Study by Confocal Microscopy
The confocal microscopy images of rat intestine with Rhodamine B dye, as shown in Figure 17, showed the depth of penetration of RBO-NLCs and RBO-SUS, respectively, after 2 h of dose administration. The depth of penetration measured through the
[figures omitted; refer to PDF]
4.12. Cell Line Study
4.12.1. Effect of RBO on MCF-7 Breast Cancer Cells
The antitumor activity of RBO against the MCF-7 cells was investigated by MTT assay. As shown in Figure 18, RBO exhibited a broad spectrum of inhibition against cancer cell lines in a dose-dependent manner. The result from the MTT assay, the IC50 value (μg/ml) of RBO post-24 hrs and 48 hrs of treatment against the MCF-7 cell line at 37°C at 5% CO2, was found to be 39.51 and 13.36 μg/ml, respectively. After calculation of the IC50 value, similarly dose-dependent toxicity of the RBO-NLCs formulation was done as shown in Figure 19. The % cytotoxicity was significantly enhanced in the case of nanoformulation with lower IC50 value of 0.531 and 0.474 μg/ml post-24 hrs and 48 hrs, respectively. So, by formulating into NLCs of the drug, the cytotoxicity was enhanced and reduced their IC50 value by 74 times and 28 times after 24 h and 48 h, respectively. A similar study was also conducted by Marinelli and coworkers in which RBO alone or in combination with Everolimus was investigated in breast cancer lines [59].
[figure omitted; refer to PDF][figure omitted; refer to PDF]4.13. In Vivo Pharmacokinetic Study
In vivo pharmacokinetic study was conducted in female Wistar rats for the determination of bioavailability data, as depicted into Figure 20 after the oral administration of RBO suspension and RBO-loaded NLCs. The peak plasma concentration (
Table 12
Pharmacokinetic parameters of RBO-SUS and RBO-NLCs.
Pharmacokinetic parameters | RBO-SUS | RBO-NLCs |
4.00 | 4.00 | |
Data represented as
4.14. Stability Study
The parameters of optimized NLCs on the stability study were evaluated as per the ICH guidelines, and change in any physical and chemical was evaluated as shown in Table 13. A freshly prepared sample was kept for stability study after determining their particle size, and PDI, which was
Table 13
Result of stability study for RBO-NLCs at
Time (month) | Phase separation | Sedimentation | Particle size | PDI | Entrapment efficiency | |||
0 | No | No | nm | SD | PDI | SD | %EE | SD |
114.23 | 2.75 | 0.649 | 0.043 | 87.75 | 1.79 | |||
1 | No | No | 127.78 | 1.26 | 0.711 | 0.067 | 86.47 | 0.91 |
3 | No | No | 131.91 | 2.50 | 0.703 | 0.054 | 85.87 | 0.79 |
6 | No | No | 135.44 | 1.31 | 0.707 | 0.067 | 85.13 | 0.91 |
[figures omitted; refer to PDF]
5. Conclusion
RBO loaded NLCs was prepared using solvent evaporation followed by the probe sonication method. The NLCs formulation was optimized using the Box-Behnken design response surface method. The prepared NLCs exhibited mean particle size of
Acknowledgments
The authors would like to acknowledge the Indian Council of Medical Research, Govt. of India, New Delhi, India, for providing ICMR-SRF Research fellow to the first author (Grant Number: 45/36/2019-NAN-BMS).
Glossary
Abbreviations
RBO:Ribociclib
NLCs:Nanostructured lipid carriers
P-gp:P-Glycoprotein
BBD:Box-Behnken design
PDI:Polydispersity index
EE:Entrapment efficiency
TEM:Transmission electron microscopy
CDK:Cyclin-dependent kinase
IUPAC:International Union of Pure and Applied Chemistry
HLB:Hydrophilic and lipophilic balance
rcf:Relative centrifugal force
UV:Ultraviolet
QbD:Quality by design
p-XRD:Powder X-ray diffraction
FT-IR:Fourier-transform infrared spectroscopy.
[1] M. R. Ataollahi, J. Sharifi, M. R. Paknahad, A. Paknahad, "Breast cancer and associated factors: a review," Journal Of Medicine and Life, vol. 8, 2015.
[2] S. Malvia, S. A. Bagadi, U. S. Dubey, S. Saxena, "Epidemiology of Breast Cancer in Indian Women," Asia‐Pacific Journal of Clinical Oncology, vol. 13 no. 4, pp. 289-295, DOI: 10.1111/ajco.12661, 2017.
[3] E. Hamilton, J. R. Infante, "Targeting CDK4/6 in patients with cancer," Cancer Treatment Reviews, vol. 45, pp. 129-138, DOI: 10.1016/j.ctrv.2016.03.002, 2016.
[4] G. N. Hortobagyi, S. M. Stemmer, H. A. Burris, Y. S. Yap, G. S. Sonke, S. Paluch-Shimon, M. Campone, K. L. Blackwell, F. Andre, E. P. Winer, W. Janni, S. Verma, P. Conte, C. L. Arteaga, D. A. Cameron, K. Petrakova, L. L. Hart, C. Villanueva, A. Chan, E. Jakobsen, A. Nusch, O. Burdaeva, E. M. Grischke, E. Alba, E. Wist, N. Marschner, A. M. Favret, D. Yardley, T. Bachelot, L. M. Tseng, S. Blau, F. Xuan, F. Souami, M. Miller, C. Germa, S. Hirawat, J. O’Shaughnessy, "Ribociclib as first-line therapy for HR-positive, advanced breast cancer," The New England Journal Of Medicine, vol. 375 no. 18, pp. 1738-1748, DOI: 10.1056/NEJMoa1609709, 2016.
[5] J. I. Lai, Y. J. Tseng, M. H. Chen, C. Y. F. Huang, P. M. H. Chang, "Clinical perspective of FDA approved drugs with P-glycoprotein inhibition activities for potential cancer therapeutics," . 2020, http://www.frontiersin.org
[6] "Center for drug evaluation and research," . https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/209092Orig1s000ChemR.pdf
[7] Z. Fei, M. Yoosefian, "Design and development of polymeric micelles as nanocarriers for anti-cancer ribociclib drug," Journal of Molecular Liquids, vol. 329, article 115574,DOI: 10.1016/j.molliq.2021.115574, 2021.
[8] L. Brannon-Peppas, J. O. Blanchette, "Nanoparticle and targeted systems for cancer therapy," . 2004, https://pubmed.ncbi.nlm.nih.gov/15350294/
[9] N. Naseri, H. Valizadeh, P. Zakeri-Milani, "Solid lipid nanoparticles and nanostructured lipid carriers: structure preparation and application," Advanced Pharmaceutical Bulletin, vol. 5 no. 3, pp. 305-313, DOI: 10.15171/apb.2015.043, 2015.
[10] S. Khan, S. Baboota, J. Ali, S. Khan, R. S. Narang, J. K. Narang, "Nanostructured lipid carriers: an emerging platform for improving oral bioavailability of lipophilic drugs," International Journal of Pharmaceutical Investigation, vol. 5 no. 4, pp. 182-191, DOI: 10.4103/2230-973X.167661, 2015.
[11] S. Khan, M. Shaharyar, M. Fazil, S. Baboota, J. Ali, "Tacrolimus-loaded nanostructured lipid carriers for oral delivery - Optimization of production and characterization," European Journal of Pharmaceutics and Biopharmaceutics, vol. 108, pp. 277-288, DOI: 10.1016/j.ejpb.2016.07.017, 2016.
[12] T. Alam, S. Khan, B. Gaba, M. F. Haider, S. Baboota, J. Ali, "Adaptation of Quality by Design-Based Development of Isradipine Nanostructured-Lipid Carrier and Its Evaluation for In Vitro Gut Permeation and In Vivo Solubilization Fate," Journal of Pharmaceutical Sciences, vol. 107 no. 11, pp. 2914-2926, DOI: 10.1016/j.xphs.2018.07.021, 2018.
[13] B. Iqbal, J. Ali, S. Baboota, "Silymarin loaded nanostructured lipid carrier: from design and dermatokinetic study to mechanistic analysis of epidermal drug deposition enhancement," Journal of Molecular Liquids, vol. 255, pp. 513-529, DOI: 10.1016/j.molliq.2018.01.141, 2018.
[14] I. Jazuli, Annu, B. Nabi, T. moolakkadath, T. Alam, S. Baboota, J. Ali, "Optimization of nanostructured lipid carriers of lurasidone hydrochloride using box-behnken design for brain targeting: in vitro and in vivo studies," Journal of Pharmaceutical Sciences, vol. 108 no. 9, pp. 3082-3090, DOI: 10.1016/j.xphs.2019.05.001, 2019.
[15] A. A. Khan, J. Mudassir, S. Akhtar, V. Murugaiyah, Y. Darwis, "Freeze-dried lopinavir-loaded nanostructured lipid carriers for enhanced cellular uptake and bioavailability: statistical optimization, in vitro and in vivo evaluations," Pharmaceutics, vol. 11 no. 2,DOI: 10.3390/pharmaceutics11020097, 2019.
[16] E. González-Mira, S. Nikolić, M. L. García, M. A. Egea, E. B. Souto, A. C. Calpena, "Potential use of nanostructured lipid carriers for topical delivery of flurbiprofen," Journal of Pharmaceutical Sciences, vol. 100 no. 1, pp. 242-251, DOI: 10.1002/jps.22271, 2011.
[17] Z. Zhang, F. Gao, H. Bu, J. Xiao, Y. Li, "Solid lipid nanoparticles loading candesartan cilexetil enhance oral bioavailability: in vitro characteristics and absorption mechanism in rats," Nanomedicine: Nanotechnology, Biology, and Medicine, vol. 8 no. 5, pp. 740-747, DOI: 10.1016/j.nano.2011.08.016, 2012.
[18] S. Khan, M. Shaharyar, M. Fazil, M. Q. Hassan, S. Baboota, J. Ali, "Tacrolimus-loaded nanostructured lipid carriers for oral delivery- in vivo bioavailability enhancement," European Journal of Pharmaceutics and Biopharmaceutics, vol. 109, pp. 149-157, DOI: 10.1016/j.ejpb.2016.10.011, 2016.
[19] Q. Luo, J. Zhao, X. Zhang, W. Pan, "Nanostructured lipid carrier (NLCs) coated with chitosan oligosaccharides and its potential use in ocular drug delivery system," International Journal of Pharmaceutics, vol. 403 no. 1-2, pp. 185-191, DOI: 10.1016/j.ijpharm.2010.10.013, 2011.
[20] A. Innes, A. M. Farrell, R. P. Burden, A. G. Morgan, R. J. Powell, "Complement activation by cellulosic dialysis membranes," Journal of Clinical Pathology, vol. 47 no. 2, pp. 155-158, DOI: 10.1136/jcp.47.2.155, 1994.
[21] M. U. K. Sahibzada, A. Sadiq, S. Khan, H. S. Faidah, N. Ullah, M. U. A. Khurram, M. U. Amin, A. Haseeb, "Fabrication, characterization and in vitro evaluation of silibinin nanoparticles: an attempt to enhance its oral bioavailability," Drug Design, Development and Therapy, vol. 11, pp. 1453-1464, DOI: 10.2147/DDDT.S133806, 2017.
[22] A. C. Salome, C. O. Godswill, I. O. Ikechukwu, "Kinetics and mechanisms of drug release from swellable and non swellable matrices: a review," Research Journal of Pharmaceutical, Biological and Chemical Sciences, vol. 4 no. 2, pp. 97-103, 2013.
[23] Y. Liu, L. Wang, Y. Zhao, M. He, X. Zhang, M. Niu, N. Feng, "Nanostructured lipid carriers versus microemulsions for delivery of the poorly water-soluble drug luteolin," International Journal of Pharmaceutics, vol. 476 no. 1, pp. 169-177, 2014.
[24] M. M. Cruz Silva, V. M. C. Madeira, L. M. Almeida, J. B. A. Custódio, "Hemolysis of human erythrocytes induced by tamoxifen is related to disruption of membrane structure," Biochimica et Biophysica Acta (BBA)-Biomembranes, vol. 1464 no. 1, pp. 49-61, DOI: 10.1016/S0005-2736(99)00237-0, 2000.
[25] Z. Attari, A. Bhandari, P. C. Jagadish, S. Lewis, "Enhanced ex vivo intestinal absorption of olmesartan medoxomil nanosuspension: Preparation by combinative technology," Saudi Pharmaceutical Journal, vol. 24 no. 1, pp. 57-63, DOI: 10.1016/j.jsps.2015.03.008, 2016.
[26] A. Singh, Y. R. Neupane, B. Mangla, K. Kohli, "Nanostructured lipid carriers for oral bioavailability enhancement of exemestane: formulation design, in vitro , ex vivo , and in vivo Studies," Journal of Pharmaceutical Sciences, vol. 108 no. 10, pp. 3382-3395, DOI: 10.1016/j.xphs.2019.06.003, 2019.
[27] B. V. Jardim-Perassi, A. S. Arbab, L. C. Ferreira, T. F. Borin, N. R. S. Varma, A. S. M. Iskander, A. Shankar, M. M. Ali, D. A. P. de Campos Zuccari, "Effect of melatonin on tumor growth and angiogenesis in xenograft model of breast cancer," PLoS One, vol. 9 no. 1, article e85311,DOI: 10.1371/journal.pone.0085311, 2014.
[28] S. Reagan-Shaw, M. Nihal, N. Ahmad, "Dose translation from animal to human studies revisited," The FASEB Journal, vol. 22 no. 3, pp. 659-661, DOI: 10.1096/fj.07-9574LSF, 2008.
[29] B. Nabi, S. Rehman, S. Aggarwal, S. Baboota, J. Ali, "Quality by design adapted chemically engineered lipid architectonics for HIV therapeutics and intervention: contriving of formulation, appraising the in vitro parameters and in vivo solubilization potential," AAPS PharmSciTech, vol. 21 no. 7,DOI: 10.1208/s12249-020-01795-w, 2020.
[30] K. H. Bang, Y. G. Na, H. W. Huh, S. J. Hwang, M. S. Kim, M. Kim, H. K. Lee, C. W. Cho, "The delivery strategy of paclitaxel nanostructured lipid carrier coated with platelet membrane," Cancers, vol. 11 no. 6,DOI: 10.3390/cancers11060807, 2019.
[31] L. M. Negi, M. Jaggi, S. Talegaonkar, "Development of protocol for screening the formulation components and the assessment of common quality problems of nano-structured lipid carriers," International Journal of Pharmaceutics, vol. 461 no. 1–2, pp. 403-410, DOI: 10.1016/j.ijpharm.2013.12.006, 2014.
[32] C. Zhang, F. Peng, W. Liu, J. Wan, C. Wan, H. Xu, C. W. Lam, X. Yang, "Nanostructured lipid carriers as a novel oral delivery system for triptolide: induced changes in pharmacokinetics profile associated with reduced toxicity in male rats," International Journal of Nanomedicine, vol. 9 no. 1, pp. 1049-1063, 2014.
[33] Z. Teng, M. Yu, Y. Ding, H. Zhang, Y. Shen, M. Jiang, P. Liu, Y. Opoku-Damoah, T. J. Webster, J. Zhou, "Preparation and characterization of nimodipine-loaded nanostructured lipid systems for enhanced solubility and bioavailability," International Journal of Nanomedicine, vol. 14, pp. 119-133, DOI: 10.2147/IJN.S186899, 2019.
[34] M. Danaei, M. Dehghankhold, S. Ataei, F. Hasanzadeh Davarani, R. Javanmard, A. Dokhani, S. Khorasani, M. R. Mozafari, "Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems," Pharmaceutics, vol. 10 no. 2,DOI: 10.3390/pharmaceutics10020057, 2018.
[35] S. Cunha, C. P. Costa, J. N. Moreira, J. M. Sousa Lobo, A. C. Silva, "Using the quality by design (QbD) approach to optimize formulations of lipid nanoparticles and nanoemulsions: a review," Nanomedicine: Nanotechnology, Biology and Medicine, vol. 28, article 102206,DOI: 10.1016/j.nano.2020.102206, 2020.
[36] N. K. Garg, G. Sharma, B. Singh, P. Nirbhavane, R. K. Tyagi, R. Shukla, O. P. Katare, "Quality by Design (QbD)-enabled development of aceclofenac loaded-nano structured lipid carriers (NLCs): An improved dermatokinetic profile for inflammatory disorder(s)," International Journal of Pharmaceutics, vol. 517 no. 1–2, pp. 413-431, DOI: 10.1016/j.ijpharm.2016.12.010, 2017.
[37] A. Kovács, S. Berkó, E. Csányi, I. Csóka, "Development of nanostructured lipid carriers containing salicyclic acid for dermal use based on the quality by design method," European Journal of Pharmaceutical Sciences, vol. 99, pp. 246-257, DOI: 10.1016/j.ejps.2016.12.020, 2017.
[38] P. Pathak, M. Nagarsenker, "Formulation and evaluation of lidocaine lipid nanosystems for dermal delivery," AAPS PharmSciTech, vol. 10 no. 3, pp. 985-992, DOI: 10.1208/s12249-009-9287-1, 2009.
[39] R. K. Subedi, K. W. Kang, H. K. Choi, "Preparation and characterization of solid lipid nanoparticles loaded with doxorubicin," European Journal of Pharmaceutical Sciences, vol. 37 no. 3–4, pp. 508-513, DOI: 10.1016/j.ejps.2009.04.008, 2009.
[40] R. Shtay, C. P. Tan, K. Schwarz, "Development and characterization of solid lipid nanoparticles (SLNs) made of cocoa butter: a factorial design study," Journal of Food Engineering, vol. 231, pp. 30-41, DOI: 10.1016/j.jfoodeng.2018.03.006, 2018.
[41] M. Noroozi, S. Radiman, A. Zakaria, "Influence of sonication on the stability and thermal properties of Al 2 O 3 nanofluids," Journal of Nanomaterials, vol. 2014,DOI: 10.1155/2014/612417, 2014.
[42] A. Ahad, M. Aqil, K. Kohli, Y. Sultana, M. Mujeeb, A. Ali, "Formulation and optimization of nanotransfersomes using experimental design technique for accentuated transdermal delivery of valsartan," Nanomedicine: Nanotechnology, Biology and Medicine, vol. 8 no. 2, pp. 237-249, DOI: 10.1016/j.nano.2011.06.004, 2012.
[43] J. Varshosaz, M. A. Davoudi, S. Rasoul-Amini, "Docetaxel-loaded nanostructured lipid carriers functionalized with trastuzumab (herceptin) for HER2-positive breast cancer cells," Journal of Liposome Research, vol. 28 no. 4, pp. 285-295, DOI: 10.1080/08982104.2017.1370471, 2018.
[44] M. Elmowafy, H. M. Ibrahim, M. A. Ahmed, K. Shalaby, A. Salama, H. Hefesha, "Atorvastatin-loaded nanostructured lipid carriers (NLCs): strategy to overcome oral delivery drawbacks," Drug Delivery, vol. 24 no. 1, pp. 932-941, DOI: 10.1080/10717544.2017.1337823, 2017.
[45] T. Alam, J. Pandit, D. Vohora, M. Aqil, A. Ali, Y. Sultana, "Optimization of nanostructured lipid carriers of lamotrigine for brain delivery: in vitro characterization and in vivo efficacy in epilepsy," Expert Opinion on Drug Delivery, vol. 12 no. 2, pp. 181-194, DOI: 10.1517/17425247.2014.945416, 2015.
[46] Ratiopharm GmbH 89079 Ulm (DE), "Crystalline forms of ribociclib free base," . 2017, https://patentimages.storage.googleapis.com/58/4f/69/f2481b0c2868b1/EP3156406A1.pdf
[47] L. J. Jia, D. R. Zhang, Z. Y. Li, F. F. Feng, Y. C. Wang, W. T. Dai, C. X. Duan, Q. Zhang, "Preparation and characterization of silybin-loaded nanostructured lipid carriers," Drug Delivery, vol. 17 no. 1, pp. 11-18, DOI: 10.3109/10717540903431586, 2010.
[48] S. Torrado, S. Torrado, "Characterization of physical state of mannitol after freeze-drying: effect of acetylsalicylic acid as a second crystalline cosolute," Chemical & Pharmaceutical Bulletin, vol. 50 no. 5, pp. 567-570, DOI: 10.1248/cpb.50.567, 2002.
[49] R. K. Cavatur, N. M. Vemuri, A. Pyne, Z. Chrzan, D. Toledo-Velasquez, R. Suryanarayanan, "Crystallization behavior of mannitol in frozen aqueous solutions," Pharmaceutical Research, vol. 19 no. 6, pp. 894-900, DOI: 10.1023/A:1016177404647, 2002.
[50] G. Yang, F. Wu, M. Chen, J. Jin, R. Wang, Y. Yuan, "Formulation design, characterization, and in vitro and in vivo evaluation of nanostructured lipid carriers containing a bile salt for oral delivery of gypenosides," International Journal of Nanomedicine, vol. Volume 14, pp. 2267-2280, DOI: 10.2147/IJN.S194934, 2019.
[51] B. Shah, D. Khunt, M. Misra, H. Padh, "Non-invasive intranasal delivery of quetiapine fumarate loaded microemulsion for brain targeting: formulation, physicochemical and pharmacokinetic consideration," European Journal of Pharmaceutical Sciences, vol. 91, pp. 196-207, DOI: 10.1016/j.ejps.2016.05.008, 2016.
[52] Annu, S. Baboota, J. Ali, "In vitro appraisals and ex vivo permeation prospect of chitosan nanoparticles designed for schizophrenia to intensify nasal delivery," Polymer Bulletin,DOI: 10.1007/s00289-021-03598-w, 2021.
[53] V. A. Duong, T. T. L. Nguyen, H. J. Maeng, S. C. Chi, "Data on optimization and drug release kinetics of nanostructured lipid carriers containing ondansetron hydrochloride prepared by cold high-pressure homogenization method," Data in Brief, vol. 26, article 104475,DOI: 10.1016/j.dib.2019.104475, 2019.
[54] A. S. Joshi, H. S. Patel, V. S. Belgamwar, A. Agrawal, A. R. Tekade, "Solid lipid nanoparticles of ondansetron HCl for intranasal delivery: development, optimization and evaluation," Journal of Materials Science: Materials in Medicine, vol. 23 no. 9, pp. 2163-2175, 2012.
[55] S. Rehman, B. Nabi, S. Baboota, J. Ali, "Tailoring lipid nanoconstructs for the oral delivery of paliperidone: Formulation, optimization and in vitro evaluation," Chemistry and Physics of Lipids, vol. 234,DOI: 10.1016/j.chemphyslip.2020.105005, 2021.
[56] S. Chauhan, M. Yallapu, Ebeling, Chauhan, Jaggi, "Interaction of curcumin nanoformulations with human plasma proteins and erythrocytes," International Journal of Nanomedicine, vol. 6,DOI: 10.2147/IJN.S25534, 2011.
[57] L. Wu, L. Zhao, X. Su, P. Zhang, G. Ling, "Repaglinide-loaded nanostructured lipid carriers with different particle sizes for improving oral absorption: preparation, characterization, pharmacokinetics, andin situintestinal perfusion," Drug Delivery, vol. 27 no. 1, pp. 400-409, DOI: 10.1080/10717544.2019.1689313, 2020.
[58] K. Soni, M. Rizwanullah, K. Kohli, "Development and optimization of sulforaphane-loaded nanostructured lipid carriers by the Box-Behnken design for improved oral efficacy against cancer:in vitro,ex vivoandin vivoassessments," Artificial Cells, Nanomedicine, and Biotechnology, vol. 46 no. sup1, pp. 15-31, DOI: 10.1080/21691401.2017.1408124, 2018.
[59] O. Marinelli, E. Romagnoli, F. Maggi, M. Nabissi, C. Amantini, M. B. Morelli, M. Santoni, N. Battelli, G. Santoni, "Exploring treatment with ribociclib alone or in sequence/combination with everolimus in ER+HER2−Rb wild-type and knock-down in breast cancer cell lines," BMC Cancer, vol. 20 no. 1,DOI: 10.1186/s12885-020-07619-1, 2020.
[60] S. Jain, S. R. Patil, N. K. Swarnakar, A. K. Agrawal, "Oral delivery of doxorubicin using novel polyelectrolyte-stabilized liposomes (layersomes)," Molecular Pharmaceutics, vol. 9 no. 9, pp. 2626-2635, DOI: 10.1021/mp300202c, 2012.
[61] A. K. Jain, N. K. Swarnakar, C. Godugu, R. P. Singh, S. Jain, "The effect of the oral administration of polymeric nanoparticles on the efficacy and toxicity of tamoxifen," Biomaterials, vol. 32 no. 2, pp. 503-515, DOI: 10.1016/j.biomaterials.2010.09.037, 2011.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2022 Ali Sartaj et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
Purpose. The current investigation is on the explicit development and evaluation of nanostructured lipidic carriers (NLCs) through the oral route to overcome the inherent lacuna of chemotherapeutic drug, in which Ribociclib (RBO) was used for breast cancer to diminish the bioavailability issue. Method. The RBO-NLCs were prepared using the solvent evaporation method and optimized method by the Box–Behnken design (BBD). Various assessment parameters characterized the optimized formulation and their in vivo study. Results. The prepared NLCs exhibited mean particle size of
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details


1 Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
2 Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi 110007, India
3 Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences and Research, New Delhi 110017, India