Next Article in Journal
From Lab to Upscale—Boosting Formulation Performance through In Vitro Technologies
Previous Article in Journal
Theoretical Models of the Most Promising Needle-Free Electrospinning Systems for Drug Delivery Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

Enhancement of the Solubility of Rosuvastatin Calcium by Nanovesicular Formulation: A Systematic Study Based on a Quality by Design Approach †

by
Marwa H. S. H. S. Dawoud
1,*,
Ahmed M. M. Fayez
2,
Reem A. A. Mohamed
2 and
Nabila M. M. Sweed
1
1
Department of Pharmaceutics, Faculty of Pharmacy, October University for Modern Sciences and Arts, 6th of October, 12451 Egypt
2
Department of Pharmacology, Faculty of Pharmacy, October University for Modern Sciences and Arts, 6th of October, 12451 Egypt2 [email protected] (A.M.F.)
*
Author to whom correspondence should be addressed.
Presented at the 1st International Electronic Conference on Pharmaceutics, 1–15 December 2020; Available online: https://iecp2020.sciforum.net/.
Proceedings 2021, 78(1), 34; https://doi.org/10.3390/IECP2020-08698
Published: 1 December 2020
(This article belongs to the Proceedings of The 1st International Electronic Conference on Pharmaceutics)

Abstract

:
Rosuvastatin calcium (Rsv) is an effective statin, with a potent antihyperlipidemic effect. However, it suffers poor bioavailability owing to its poor solubility. Thus; encapsulating Rsv into a nanovesicular structure could overcome this problem. The aim of this work is to investigate the potential of solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) in enhancing the solubility of Rsv, using the quality by design (QbD) concept. A complete risk assessment study has been conducted, where the critical process parameters (CPPs), material attributes (MAs), and critical quality attributes have been identified using Ishikawa diagrams. Selected CPPs/MAs were screened and further upgraded to a 24 full-factorial design to develop a design space with the optimized formula. The screened CPPs/MAs were tested on the particle size, the polydispersity index (PDI), the zeta potential (ζ-pot), and the entrapment efficiency (EE%). A comprehensive approach for Rsv nanovesicular carriers has been conducted, where the NLCs showed better results than the SLNs. The optimized formula was prepared with 3% total lipid content, 0.154% surfactant, and 9.4 mg drug. The optimized formula had a particle size of 310.5 nm, with 0.243 PDI, a ζ-pot of −24.7 mV, and an EE% of 93.87%, and showed a sustained release of the drug for up to 72 h. It successfully lowered total cholesterol, low density lipoprotein, and triglycerides, and elevated the levels of high density lipoprotein in rats, with better results as compared to the standard drug. Thus, a complete QbD study was conducted to explore experimental regions for many successful nanovesicular carriers for the enhancement of the solubility of poorly soluble drugs.

1. Introduction

Quality by design (QbD) is a systematic science and risk-based approach that plays a great role in product and process understanding in order to achieve a safe product. The use of QbD in pharmaceutical formulation assures the quality of a pharmaceutical product through the use of scientific development and risk management tools, producing a high quality product in the most efficient manner [1]. Rosuvastatin calcium (Rsv) is one of the most effective statins, with the potential of reducing low-density lipoprotein (LDL), triglycerides (TGs), and increasing high-density lipoprotein (HDL). However, Rsv suffers from poor solubility and extensive first-pass effect, resulting in its poor bioavailability [2]. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) are vesicular nanoparticles made from physiologically accepted and biodegradable lipid fractions [3]. The objective of the current study is the application of a QbD approach in the optimization and formulation of Rsv; in an attempt to improve its solubility.

2. Experiments

2.1. Materials

Rosuvastatin calcium, A32700, was as a generous gift from the Global Napi Pharmaceutical Company (Cairo, Egypt), and Precirol® ATO 5 and Compritol® 888 ATO were received as gifts from GatteFosse (Lyon, France). Tween® 20 was purchased from Sigma (St. Louis, MO, USA) while Tween® 80 from Scharlau (Barcelona, Spain). Stearic acid was obtained from Piochem (Giza, Egypt), Oleic acid from Oxford Labchem (Maharashtra, India), and Castor oil from UCCMA (Cairo, Egypt). Poloxamer® 188 was from Caisson (Smithfield, UT, USA). All other chemicals and reagents were of analytical grade. Fructose for the in vivo study was obtained from UNIPHARMA Co. (El-Obour City, Cairo, Egypt), while sheep tail fat and hydrogenated oil were from obtained commercial sources. Sodium carboxymethyl cellulose was obtained from Chemajet Pharmaceutical Industries (Cairo, Egypt).

2.2. Methods

2.2.1. Preparation of the Nanovesicular Carrier

SLNs and NLCs were prepared by emulsification–ultrasonication method as reported by Das et al. [4].

2.2.2. Characterization of Nanoparticles

Particle size (PS), polydispersity index (PDI), and zeta potential (ζ-pot) measurements were performed using dynamic light scattering using a zetasizer after suitable dilution [5].The entrapment efficiency (EE%) was measured indirectly by analyzing the free drug in the supernatant after centrifugation of the dispersion [5].

2.2.3. Quality by Design Paradigm

The quality target product profile (QTPP) of the current study is to enhance the solubility of Rsv. The average PS, PDI, ζ-pot, EE%, and drug release profile were taken as the influential critical quality attributes (CQAs) for the current study [6].
Ientification of the failure modes was performed using Ishikawa diagrams, to figure out the critical process parameters (CPPs) and material attributes (MAs) affecting the QTPP [7].
Screening of different solid lipids, liquid lipids, and surfactants for nanovesicular formulation: saturated solubility of the drug in different liquid lipids (oleic acid and castor oil) and SAA (Tween® 20, Tween® 80 and Poloxamer® 188) was measured [8]. Compritol® 888 ATO, Precirol® ATO 5, and Stearic acid were tested for their ability to solubilize Rsv [9].
Optimization of Rsv-loaded SLN/NLC with the selected variables: a 24 full factorial design was used for the optimization steps shown in Table 1.
Data optimization and model validation: a design space was established based on the product desirability. An optimized formula (O1) was prepared as suggested by the program and was evaluated, and compared with the expected results.

2.2.4. In-Vitro Drug Release

An in-vitro drug release study was tested using the dialysis membrane method in PBS at pH 7.4. The optimized formula was compared to the standard Rsv in distilled water (both containing 10 mg Rsv), where the samples were withdrawn over a period of 72 h [10].

2.2.5. In-Vivo Pharmacodynamics Study

The in-vivo study was conducted on 24 male Wistar rats (170–200 g), which were allowed free access to water and food [11]. The rats were divided into two dietary groups. The normal-fat diet (NFD) group consisted of six rats fed on a NFD, and the high-fat diet (HFD) group consisted of 18 rats fed on a HFD. This diet regimen was continued for six weeks, and at week seven, the rats were fasted, anesthetized, and blood samples were withdrawn to measure triglycerides (TGs) and total cholesterol (TC) [12].
Animals were then grouped into four groups, the first group being the negative control rats, which were the NFD group and received plain sodium carboxymethyl cellulose aqueous solution. The second group was the hyperlipidemic positive control group, a HFD group in which the rats received plain sodium carboxymethyl cellulose aqueous solution, while the third group were a HFD group receiving Rsv in sodium carboxymethyl cellulose aqueous solution, and the last group were a HFD group receiving the optimized formula (O1). After two weeks, animals were then anesthetized and fasting blood samples were taken to measure TGs, TC, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) [12].

3. Results and Discussion

3.1. Quality Target Product Profile and Risk Analysis

Potential causes of each of the CQAs were outlined using Ishikawa diagrams, as represented in Figure 1.

3.2. Screening of Different Solid Lipids, Liquid Lipids, and Surfactants (SAA) for Nanovesicular Formulation

Rsv was found to be most soluble in Precirol® ATO 5 among the solid lipids, in oleic acid among the liquid lipids, and in Tween® 20 among the SAA. Accordingly, these were the ingredients of choice for the nanovesicular preparations. The high solubility of Rsv in Precirol® ATO 5 may be due to the highly porous structure of Precirol® ATO 5 which allows more drug accommodation and solubility. The presence of methane sulfonamide hydrophilic moiety in Rsv resulted in the imparting of a slight hydrophilic nature, which in turn leads to its better solubility in Tween® 20, than Tween® 80 [13]. However, the hydrophobic nature of the drug allows Tween® 20 to solubilize it more than Poloxamer® 188 [10].

3.3. Response Surface Design Analysis

Further analysis using ANOVA indicated that all models were significant, with the significant effect of CPPs/MAs on the measured CQAs at (p < 0.05).

3.3.1. Particle Size Analysis

The effect of the CPPs/MAs on the PS can be described as:
PS = 361.1−67.6 * X1 − 44.7 * X2 − 57.6 * X3 − 6.2 * X4 + 20.6 * X12 + 82.7 * X13 + 31.3 * X14 + 55.9 * X23 − 43.1 * X24 + 7.6 * X34
An increase in X1 or X2 resulted in a significant reduction in the particle size. Probably, this may be due to the reduction in the surface tension when SAA% increases, making the oil droplets smaller. In addition, SAA would be able to coat the oil droplets, which would stabilize the dispersion [4]. A larger particle size was observed when X3 was decreased, i.e., the NLC was prepared, which may be attributed to the presence of the liquid lipid which might have increased the hydrodynamic diameter [14]. The effect of the drug amount was insignificant.

3.3.2. PDI Analysis

The effect of the CPPs/MAs on the PDI can be described as:
PDI = +0.41 − 0.11* X1 + 0.051 * X2 + 9.344E − 003 * X3 − 0.048 * X4 + 0.053 * X13 − 0.067 * X24
A lower lipid content (X1) resulted in a higher PDI value, which may be due to the insufficient amount of lipid to enclose Rsv, leading to the heterogenicity of the system [15]. The PDI was bigger at a high SAA%, which could be attributed to the excess amount of SAA that may accumulate on the surface of the vesicles, resulting in an increase in the system’s heterogenicity [8]. Moreover, the excess SAA might lower the surface tension to an extreme extent, which might rupture the vesicles and increase the system’s heterogenicity [4]. A higher drug amount had a statistical significant lowering effect on the PDI, while the ratio between solid lipid and liquid lipid was insignificant.

3.3.3. Zeta Potential Analysis

The effect of the CPPs/MAs on the ζ-pot can be described as:
ζ-pot = +14 − 1.10 * X1 + 0.80 * X2 + 1.51 * X3 − 0.98 * X4 − 0.56 * X13 + 0.92 * X23 − 1.18 * X34
The increase in the ζ-pot by decreasing X1 could be due to the reduction in the particle size by the increase in the total lipid content, which in turn reduces the surface area of the vesicles with less charge accommodation [16]. Moreover, the ζ-pot was increased by the increase in the SAA%, which could be due to molecular polarization and the adsorption of the surface acting agent in the water. The adsorbed SAA in the water could be absorbed to the emulsifier layer of the particle/water interface and form an electric double layer that is similar to an ionic state [17].

3.3.4. EE% Analysis

The effect of the CPPs/MAs on the EE% can be described as:
EE% = 76.46 + 4.33 * X1 − 6.25 * X2 − 5.02 * X3 + 5.38 * X4 − 4.49 * X13 + 4.61 * X23 − 5.63 * X24
A significant increase in the EE% was observed with the increase in the lipid content, which might be due to sufficient amounts of lipids that can be used to encapsulate the drug [15]. Moreover, a higher SAA% resulted in a lower EE%, which may be due to the disruption of the vesicles at a high SAA concentration [4]. When the NLC was prepared the EE% increased, which could be due to the incorporation of a liquid lipid into a solid lipid, leading to a number of crystalline sequence disturbances and defects in the crystal lattice, which could in turn create space for the encapsulation of the Rsv molecules [10]. Finally, an increase in the EE% was observed by the increase in the drug amount, which may be due to the increased availability of the Rsv, which consequently improves its retention within the vesicles [17].

3.4. Model Validation, Data Optimization, and Control Strategy Establishment

An optimized formula (O1), with desirability 0.893, was prepared (Table 2). The validity of the design was established by comparing the observed results with the expected ones, which were found to statistically insignificant. A successful design space was established with a control space that ensures the reproducibility of the product.

3.5. In-Vitro Drug Release Analysis

The NLC optimized formula was able to release the drug in a sustained manner, as compared to the standard Rsv (Figure 2). The optimized formula was able to sustain the release for up to 72 h. The drug on the surface of the vesicles resulted in an initial burst release which was followed by a sustained release pattern [18].

3.6. In-Vivo Pharmacodynamics Study

In agreement with previous reports [19], the hyperlipidemic positive control group exhibited a significant increase in TGs, TC, and LDL when compared to the negative control. Compared to standard Rsv, the optimized formula O1 significantly reduced serum TC level by 26.6% and LDL by 46%. The elevation in HDL (13.95%) and reduction in TGs (39%) were not significant.
Statins have been reported to have side effects among quite a significant number of patients (5–20%), with more side effects appearing at higher doses [20]. The use of the nanovesicular formulation of Rsv may increase its solubility, and hence its bioavailability. Moreover, the lipid formulation could induce bile secretion in the small intestine where the NLC would be associated with the bile salts, forming mixed micelles, and thus ensuring the NLC transition to the lymphatic circulation directly, bypassing the first-pass effect and promoting its better absorption [21]. All this resulted in improving and sustaining the antihyperlipidemic activity of Rsv NLC when compared to the standard Rsv.

4. Conclusions

The QbD approach was found to be very useful in formulation of a nanovesicular carrier loaded with Rsv. Several tools have been used in the risk assessment and design of experiments for the screening and the optimization of the nanovesicular carriers. A design space was established which defines the control strategy for the formulation of the Rsv-nanovesicular carrier. This control strategy gives the permitted ranges of the total lipid content, SAA%, type of the nanovesicle, and the drug amount that produced the nano-vesicle with optimal PS, PDI, ζ-potential, and EE% for any further studies. The prepared optimized formula managed to significantly lower each of the TC, TGs, and LDL, and to elevate the HDL as compared to the positive control, thus proving the potential use of the Rsv-NLC as a successful antihyperlipidemic agent. A full and successful practical use of the QbD approach in pharmaceutical development was applied by the use of several advanced techniques, which could be considered as reliable reference for further nanovesicular carriers formulations.

Author Contributions

Conceptualization, M.H.S.D. and N.M.S.; methodology, M.H.S.D., N.M.S., A.M.F. and R.A.M.; resources, M.H.S.D., N.M.S., A.M.F. and R.A.M.; analysis, M.H.S.D., A.M.F. and R.A.M.; data curation, M.H.S.D.; writing—original draft preparation, M.H.S.D., A.M.F. and R.A.M.; writing—reviewing and editing, M.H.S.D. and N.M.S. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the MSA University ethical committee, Egypt with approval number (PT15/EC15/2018F).

Informed Consent Statement

This work contains no study involving humans.

Data Availability Statement

The study didn’t report any data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xu, X.; Khan, M.A.; Burgess, D.J. A quality by design (QbD) case study on liposomes containing hydrophilic API: I. Formulation, processing design and risk assessment. Int. J. Pharm. 2011, 419, 52–59. [Google Scholar] [CrossRef] [PubMed]
  2. Kulkarni, N.; Ranpise, N.; Mohan, G. Development and Evaluation of Solid Self Nano-Emulsifying Formulation of Rosuvastatin Calcium for Improved Bioavailability. Trop. J. Pharm. Res. 2015, 14, 575. [Google Scholar] [CrossRef]
  3. Bahari, L.A.S.; Hamishehkar, H. The impact of variables on particle size of solid lipid nanoparticles and nanostructured lipid carriers; A comparative literature review. Adv. Pharm. Bull. 2016, 6, 143–151. [Google Scholar] [CrossRef] [PubMed]
  4. Das, S.; Ng, W.K.; Tan, R.B.H. Are nanostructured lipid carriers (NLCs) better than solid lipid nanoparticles (SLNs): Development, characterizations and comparative evaluations of clotrimazole-loaded SLNs and NLCs? Eur. J. Pharm. Sci. 2012, 47, 139–151. [Google Scholar] [CrossRef]
  5. Patil-Gadhe, A.; Pokharkar, V. Pulmonary targeting potential of rosuvastatin loaded nanostructured lipid carrier: Optimization by factorial design. Int. J. Pharm. 2016, 501, 199–210. [Google Scholar] [CrossRef]
  6. Xu, X.; Costa, A.P.; Khan, M.A.; Burgess, D.J. Application of quality by design to formulation and processing of protein liposomes. Int. J. Pharm. 2012, 434, 349–359. [Google Scholar] [CrossRef]
  7. Amasya, G.; Badilli, U.; Aksu, B.; Tarimci, N. Quality by design case study 1: Design of 5-fluorouracil loaded lipid nanoparticles by the W/O/W double emulsion—Solvent evaporation method. Eur. J. Pharm. Sci. 2016, 84, 92–102. [Google Scholar] [CrossRef]
  8. Basalious, E.B.; El-Sebaie, W.; El-Gazayerly, O. Application of Pharmaceutical QbD for Enhancement of the Solubility and Dissolution of a Class II BCS Drug using Polymeric Surfactants and Crystallization Inhibitors: Development of Controlled-Release Tablets. AAPS PharmSciTech 2011, 12, 799–810. [Google Scholar] [CrossRef]
  9. Singh, H.; Gupta, R.D.; Gautam, G. Formulation Development, Characterization, and in Vitro-in Vivo Study of Antihyperlipidemic Drug Rosuvastatin Calcium—Solid Lipid Nanoparticles. Asian J. Pharm. Clin. Res. 2018, 11, 436. [Google Scholar] [CrossRef]
  10. Patil-Gadhe, A.; Pokharkar, V. Montelukast-loaded nanostructured lipid carriers: Part i Oral bioavailability improvement. Eur. J. Pharm. Biopharm. 2014, 88, 160–168. [Google Scholar] [CrossRef]
  11. Mohamed, R.A.; Galal, O.; Mohammed, A.R.; El-Abhar, H.S. Tropisetron modulates peripheral and central serotonin/insulin levels via insulin and nuclear factor kappa B/receptor for advanced glycation end products signalling to regulate type-2 diabetes in rats. RSC Adv. 2018, 8, 11908–11920. [Google Scholar] [CrossRef]
  12. Schaalan, M.; El-Abhar, H.S.; Barakat, M.; El-Denshary, E.S. Westernized-like-diet-fed rats: Effect on glucose homeostasis, lipid profile, and adipocyte hormones and their modulation by rosiglitazone and glimepiride. J. Diabetes Complic. 2009, 23, 199–208. [Google Scholar] [CrossRef]
  13. Sarfraz, R.M.; Ahmad, M.; Mahmood, A.; Minhas, M.U.; Yaqoob, A. Development and Evaluation of Rosuvastatin Calcium Based Microparticles for Solubility Enhancement: An In Vitro Study. Adv. Polym. Technol. 2017, 36, 433–441. [Google Scholar] [CrossRef]
  14. Hu, X.; Zhang, Y.; Yang, J.; Wan, H. Influence of liquid lipid content on the properties of puerarin-loaded lipid nanoparticles. J. Chin. Adv. Mater. Soc. 2014, 2, 9–19. [Google Scholar] [CrossRef]
  15. Gokce, E.H.; Korkmaz, E.; Dellera, E.; Sandri, G.; Bonferoni, M.C.; Ozer, O. Resveratrol-loaded solid lipid nanoparticles versus nanostructured lipid carriers: Evaluation of antioxidant potential for dermal applications. Int. J. Nanomed. 2012, 7, 1841. [Google Scholar] [CrossRef]
  16. Khames, A.; Khaleel, M.A.; El-Badawy, M.F.; El-Nezhawy, A.O.H. Natamycin solid lipid nanoparticles—sustained ocular delivery system of higher corneal penetration against deep fungal keratitis: Preparation and optimization. Int. J. Nanomed. 2019, 14, 2515–2531. [Google Scholar] [CrossRef]
  17. Wang, X. Development of antibody arrays for measuring biosimilar conformational comparability at molecular level. J. Bioequiv. Availab. 2012, 1, 24–26. [Google Scholar] [CrossRef]
  18. Sahu, A.K.; Kumar, T.; Jain, V. Formulation optimization of erythromycin solid lipid nanocarrier using response surface methodology. Biomed Res. Int. 2014, 2014, 689391. [Google Scholar] [CrossRef]
  19. Carmena, R. Type 2 diabetes, dyslipidemia, and vascular risk: Rationale and evidence for correcting the lipid imbalance. Am. Heart J. 2005, 150, 859–870. [Google Scholar] [CrossRef]
  20. Martin, P.D.; Warwick, M.J.; Dane, A.L.; Brindley, C.; Short, T. Absolute Oral Bioavailability of Rosuvastatin in Healthy White Adult Male Volunteers. Clin. Ther. 2003, 25, 2553–2563. [Google Scholar] [CrossRef]
  21. Khan, S.; Baboota, S.; Ali, J.; Khan, S.; Narang, R.S.; Narang, J.K. Nanostructured lipid carriers: An emerging platform for improving oral bioavailability of lipophilic drugs. Int. J. Pharm. Investig. 2015, 5, 182–191. [Google Scholar]
Figure 1. Ishikawa diagrams for different CQAs; (A) particle size (PS) and polydispersity index (PDI), (B) the zeta potential (ζ-pot), and (C) the entrapment efficiency (EE%).
Figure 1. Ishikawa diagrams for different CQAs; (A) particle size (PS) and polydispersity index (PDI), (B) the zeta potential (ζ-pot), and (C) the entrapment efficiency (EE%).
Proceedings 78 00034 g001
Figure 2. In-vitro release of Rsv from the optimized Rsv formula and standard Rsv.
Figure 2. In-vitro release of Rsv from the optimized Rsv formula and standard Rsv.
Proceedings 78 00034 g002
Table 1. The studied critical process parameters (CPPs)/ material attributes (Mas), their levels, and the composition of the 16 formulae as obtained from the factorial design, with the results of the critical quality attributes (CQAs).
Table 1. The studied critical process parameters (CPPs)/ material attributes (Mas), their levels, and the composition of the 16 formulae as obtained from the factorial design, with the results of the critical quality attributes (CQAs).
FactorsLow Level (−1)High Level (+1)
X1 Lipid (%)13
X2 Surfactant (%)0.10.3
X3 Solid lipid (SL): Liquid lipid (LL)7:310:0
X4 Drug amount (mg)5 10
CodeX1X2X3X4Y1 = Particle Size (nm)Y23 = Polydispersity indexY3 =Zeta potential(mV)Y4 = Entrapment efficiency (%)
F1−1−11−1279.2 ± 1.560.452 ± 0.05−14.3 ± 2.0745.43 ± 5.98
F21−1−1−1232.8 ± 3.570.175 ± 0.09−19.2 ± 1.8776.71 ± 8.83
F3111−1308.9 ± 3.540.589 ± 0.13−14.1 ± 4.0881.37 ± 3.78
F4−111−1400.4 ± 1.650.643 ± 0.08−12.9 ± 2.9885.57 ± 2.98
F51−111400.3 ± 2.560.384 ± 0.04−14.7 ± 0.9867.12 ± 4.14
F61−1−11300.0 ± 2.730.228 ± 0.05−21.4 ± 3.0994.40 ± 9.06
F711−11255.0 ± 2.900.292 ± 0.09−16.6 ± 4.0989.20 ± 3.87
F8−11−11256.5 ± 1.650.400 ± 0.04−16.3 ± 1.8777.39 ± 5.76
F9−1−1−11806.1 ± 2.630.538 ± 0.06−18.1 ± 3.0478.67 ± 4.31
F101111313.1 ± 0.950.270 ± 0.08−13.5 ± 1.9882.96 ± 9.31
F11−1−1−1−1736.2 ± 1.620.479 ± 0.02−11.8 ± 3.5062.23 ± 5.98
F121−11−1245.0 ± 1.160.237 ± 0.04−10.3 ± 2.0546.44 ± 2.74
F1311−1−1280.8 ± 3.070.262 ± 0.05−12.3 ± 2.0893.33 ± 4.87
F14−1111237.0 ± 3.600.445 ± 0.07−11.1 ± 1.9572.60 ± 2.09
F15−11−1−1478.5 ± 2.760.815 ± 0.04−11.0 ± 1.0864.29 ± 5.87
F16−1−111262.1 ± 2.840.348 ± 0.04−10.9 ± 1.1075.91 ± 4.21
Table 2. Composition of the optimized formula with the expected and the observed results.
Table 2. Composition of the optimized formula with the expected and the observed results.
CPPs/MAsLevel in Coded Value
Total lipid content (X1)
SAA% (X2)
SL: LL ratio (X3)
Drug amount (X4)
+1
−0.623
−1
+0.992
CQA Results
ExpectedObserved
PS (nm)352.345310.5
PDI0.2590.243
ζ-pot (mv)−20.803−24.7
EE (%)94.66393.87
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dawoud, M.H.S.H.S.; Fayez, A.M.M.; Mohamed, R.A.A.; Sweed, N.M.M. Enhancement of the Solubility of Rosuvastatin Calcium by Nanovesicular Formulation: A Systematic Study Based on a Quality by Design Approach. Proceedings 2021, 78, 34. https://doi.org/10.3390/IECP2020-08698

AMA Style

Dawoud MHSHS, Fayez AMM, Mohamed RAA, Sweed NMM. Enhancement of the Solubility of Rosuvastatin Calcium by Nanovesicular Formulation: A Systematic Study Based on a Quality by Design Approach. Proceedings. 2021; 78(1):34. https://doi.org/10.3390/IECP2020-08698

Chicago/Turabian Style

Dawoud, Marwa H. S. H. S., Ahmed M. M. Fayez, Reem A. A. Mohamed, and Nabila M. M. Sweed. 2021. "Enhancement of the Solubility of Rosuvastatin Calcium by Nanovesicular Formulation: A Systematic Study Based on a Quality by Design Approach" Proceedings 78, no. 1: 34. https://doi.org/10.3390/IECP2020-08698

Article Metrics

Back to TopTop