ISSN : 0970 - 020X, ONLINE ISSN : 2231-5039
     FacebookTwitterLinkedinMendeley

Formulation Development and Characterization of Lovastatin Nanogel for the Treatment of Hyperlipidemia

Md. Ali Mujtaba1*, Md. Sarfaraz Alam2, Nawaf M. Alotaibi3

1Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Arar, Saudi Arabia.

2Department of Pharmaceutics, HIMT group of institutions, Knowledge Park –1, Greater Noida, Distt - Gautam Budh Nagar (U.P.) – 201301, India.

3Department of Clinical Pharmacy, Faculty of Pharmacy, Northern Border University, Arar, Saudi Arabia.

Corresponding Author E-mail: sajanqa@gmail.com

DOI : http://dx.doi.org/10.13005/ojc/400404

Article Publishing History
Article Received on : 17 May 2024
Article Accepted on : 01 Jul 2024
Article Published : 09 Jul 2024
Article Metrics
Article Review Details
Reviewed by: Dr. Gihan Dissanayake
Second Review by: Dr. Gavat Cristian
Final Approval by: Dr. Malinee Sriariyanun
ABSTRACT:

Lovastatin (LS) is the cholesterol-lowering drug in the statin class, but it has poor oral bioavailability due to its high metabolism and low solubility, which affect its clinical efficacy. To overcome limitations associated with LS, the current study sought to develop a transdermal nanoemulsion using linseed oil and finally convert it into a nanogel formulation. Nanoemulsion (NE) was prepared using the spontaneous titration method. Different components of NE were selected based on solubility study and pseudo ternary phase diagrams were constructed using the titration method to determine the concentration range of components. Carbopol 934 was used to convert NE to nanogel (NG). The NE was selected based on the stability study and the composition of optimized NE consists of oil phase as 10%w/w linseed oil, 35%w/w Tween 80, and Polyethylene glycol 400 in 1:1 as Smix, and 55%w/w aqueous phase as water. The optimized NE (NE3) was characterized for various parameters and the formulation NE3 was found with desired globular size (108.9 ± 3.12 nm), polydispersity index 0.257 ± 0.015, zeta potential (−16.93 ± 1.12 mV), and spherical morphology. NE3 was combined with carbopol 934 to convert into NG and further characterized for pH, rheological behavior, and in vitro permeation study. The in vitro drug permeation study showed that the NG (33.69± 0.75 µg/cm2/hr) and NE (36.63± 0.55 µg/cm2/hr) have maximum permeation flux rate as compared to LS suspension (6.41±1.13 µg/cm2/hr). These results conclude that the NG formulation of LS can be a safe and effective alternative to an oral formulation of LS with enhanced permeation characteristics for transdermal delivery.

KEYWORDS:

Lovastatin; Nanoemulsion; Nanogel; Solubility; Stability; Permeation

Download this article as: 

Copy the following to cite this article:

Mujtaba M. A, Alam M. S, Alotaibi N. M. Formulation Development and Characterization of Lovastatin Nanogel for the Treatment of Hyperlipidemia. Orient J Chem 2024;40(4).


Copy the following to cite this URL:

Mujtaba M. A, Alam M. S, Alotaibi N. M. Formulation Development and Characterization of Lovastatin Nanogel for the Treatment of Hyperlipidemia. Orient J Chem 2024;40(4). Available from: https://bit.ly/3S1R5yR


Introduction

Hyperlipidemia is the main risk factor for the development of atherosclerosis, which eventually results in cardiovascular disease. The increased prevalence of dyslipidemia is mainly due to food habits and lifestyle factors brought about by the Westernization of food in many developing nations including Saudi Arabia1. Lovastatin (LS) is one of the most effective drugs for lowering cholesterol in the statin class of drugs. The drug LS inhibits the HMG-CoA reductase enzymes and prevents the formation of cholesterol2. Moreover, LS showed the ability to lower LDL levels while not affecting HDL levels. LS has been shown in numerous studies to reduce the death rate from coronary heart disease3,4. The Biopharmaceutical Classification System (BCS) classifies LS as a class II drug that has low solubility and high solubility. LS is available in both immediate-release and extended-release tablet dosage forms, however, due to its high metabolism in the liver and gut and its low solubility (0.0004 mg/ml in water), it has a poor oral bioavailability (<5%)4. Due to these characteristics, LS is a potential drug candidate for transdermal delivery in an attempt to increase drug bioavailability. A greater amount of the drug can enter into the systemic circulation when LS is delivered transdermally since it avoids the effects of hepatic metabolization.

Transdermal drug delivery system (TDDS), is a cutting-edge technique for drug delivery that can be utilized to increase absorption and prevent hepatic first-pass metabolism for systemic effects. Antihyperlipidemic drugs can be applied topically or systemically via transdermal delivery5. Due to the first pass effect being avoided, bioavailability can be increased. The gastrointestinal discomfort associated with statin drugs can also be prevented by using TDDS. For the transdermal delivery of drugs, nanoemulsions (NEs) are being investigated extensively. NEs are isotropically transparent, thermodynamically stable mixtures of oils, surfactants, co-surfactants, and water in the appropriate quantities that have globule sizes from 5 to 200 nm6. They have nanoscale globules, which make them optically transparent. Apart from these, they provide many benefits, including greater ability to dissolve, improved efficiency of drug-loading for both hydrophilic and lipophilic molecules, and high thermodynamic stability. NEs are preferable in TDDS as they can penetrate into deeper layers of skin7. However, their low viscosity, makes the composition difficult to apply transdermally. To avoid this problem, different gelling agents are added in NEs which increase formulation viscosity and make transdermal application easier. Gelling agents help in transdermal application by creating a three-dimensional hydrogel network that traps nanoemulsion globules8.

Transdermal administration of LS was also investigated by some researchers to increase the bioavailability of the drug. Soujanya et al., 2018 developed proniosome-based transdermal formulation of LS by the coacervation phase separation method. LS-loaded proniosomal formulation showed high percentage entrapment efficiency and controlled drug release9. Gupta et al., 2022 developed LS-loaded solid lipid nanoparticles (SLN) for transdermal delivery using glyceryl monostearate as lipid using solvent emulsification diffusion method10. Spoorthy et al., 2023 prepared polymeric nanoparticles and SLN of LS and then included them in a transdermal patch to overcome the problem associated with oral delivery of LS11. Very few articles have been published related to the NE gel formulation of LS. Kaur et al., 2019 prepared the NEs gel with tween 80, labrafac PG, and transcutol and evaluated its potential for osteoporosis8. Linseed oil has cardioprotective effect. It has rich sources of ω-3 fatty acid, which primarily improve lipid profile and act on the cardiovascular system12. There is no reported NEs gel formulation of LS prepared using linseed oil as the oil phase. Therefore, this research aims to formulate a nanoemulsion-based nanogel formulation using linseed oil for transdermal delivery of LS which can bypass the liver metabolism of the LS and increase the permeability of the LS which ultimately increases the bioavailability of LS. Therefore, the prepared nanogels (NG) formulation would increase the medication’s efficacy.

Material and Method

Materials

The drug (LS) was obtained from Jamjoom Pharmaceutical Co. Ltd., Saudi Arabia as a gift sample. The oils, surfactants, and co-surfactants were bought from SD Fine Chemicals, India, and Sigma-Aldrich, India. Analytical grade chemicals and reagents were used in this study.

Methods

Screening of components for nanoemulsions

Screening on different components for NEs preparation is based on an equilibrium solubility study13. The solubility of the drug was examined by mixing an excess of the drug into two milliliters of various oils (Linseed oil, Sunflower oil, Eucalyptus oil and Olive oil), surfactants such as Tween 20, Tween 60, Tween 80 and Cremophor RH 40), and co-surfactants such as Myoglyol, Isopropyl myristate, PEG 400, and Propylene glycol. The mixtures were mixed on an isothermal orbital shaker for 72 h at a temperature of 25 ± 2oC for equilibrium. After that, samples were obtained, and centrifugation was done for 15 mins at 3000 rpm. Supernatants were collected and passed through membrane filters with a 0.22 µ pore size which were further analyzed at 238 nm using UV spectrophotometer14.

Formulation of nanoemulsions

Linseed oil was selected as the oil phase, whereas, PEG 400 as a co-surfactant, and Tween 80 as a surfactant based on the solubility investigation as it showed the highest solubility of the LS (Table 1).  Different weight ratios of 1:1, 1:2, and 2:1%w/w of surfactant and co-surfactant were employed to make Smix. To determine the optimal ratio of Smix, we created ternary phase diagrams for each Smix. The optimum Smix was chosen for the nanoemulsion preparation process. In the oil phase (linseed oil) the drug was mixed by selecting formulations from the NEs regions of the prepared ternary phase diagram. The formulation was chosen based on the need to emulsify the most oil with the least amount of Smix. The resulting mixture was gradually mixed with double-distilled water and stirred gently until equilibrium was reached. Five NEs formulations (NE1 – NE5) were prepared with different weights of oil, Smix, and water phase (Table 2). The NEs were vortexed and monitored for up to 24 hours to detect any phase separation. Different physical stability tests such as the freeze-thaw cycle and centrifugation study were performed on the formulations6.

Physical stability testing

The stability tests of the NEs (NE1 – NE5) were performed to solve the metastable formulation issue. The formulations chosen from pseudo-ternary phase diagrams underwent additional testing, including centrifugation, freeze-thaw, and heating-cooling cycles. For the centrifugation study, the drug-loaded NE formulations (NE1–NE5) underwent centrifugation for 30 minutes at 5000 rpm. Further, the NEs formulations were exposed to 6 cycles of heating and cooling at 45oC and 4oC respectively for 48h. Freeze-thaw cycle: the storage cycles (-21oC and +25oC) were applied to the NEs formulations for 48 hours15,16. visual observation was conducted to assess how the appearance of NEs has changed. Phase separation and creaming were seen in NEs and the selected formulations were discarded. For further analysis, the thermodynamically stable NE formulation was selected.

Physicochemical characterization of NEs formulation

Droplet size (DS), polydispersity index (PDI), and zeta potential (ZP) analysis

Using photon correlation spectroscopy, DS, PDI, and ZP of NE formulation were determined17. The DS, PDI, and ZP measurements were performed through Malvern Zetasizer Nano-ZS90 (Malvern Analytical Ltd., United Kingdom). Appropriate dilution of the samples was done and the diluted samples were analyzed at a detection angle of 90o and temperature of 25 oC for PS and PDI measurement. The PDI was determined as it represents particle size distribution. The diluted sample was also used for ZP measurement with the same instrument using a second electrode to measure the ZP. ZP is used to determine the dispersion ability of globules in NEs. All measurements were done in triplicate and expressed as mean ± SD.

Transmission electron microscopy (TEM) analysis

The shape of dispersed globules in NEs was examined through TEM (JEOL JEM1010, Tokyo, Japan) operated at 100 kV accelerated voltage. On the carbon-coated grid, diluted NE with water (1:10) was applied which was further treated with 2% phosphotungstic acid droplets. Then the coated grid was kept for drying at room temperature after that it was observed under a microscope and the image was taken on the AMT image–capture engine18.

Preparation of nanogel

The thermodynamically stable formulation (NE3) was chosen for conversion into nanogel (NG) using the previously published method15. To prepare the NG, the stable nanoemulsion (NE3) was mixed with the carbopol 934 polymer dispersion. 1 gm of carbopol 934 was mixed with 100 milliliters of distilled water in a beaker, and the mixture was allowed to completely dissolve. 0.5 g of triethanolamine was added dropwise until a clear gel composition was achieved following full dispersion. The gel base and nanoemulsion were mixed 1:1 while being continuously stirred. The prepared NG was subjected to observation for different parameters. The viscosity, pH, homogeneity, and spreadability of the NG were determined. The content uniformity was tested to ensure that the amount of drug in each portion of NG is consistent.

Characterization of nanogel

pH, Spreadability, and Rheological evaluation

The physical homogeneity, color, and consistency of the NG were determined. The pH of NG was measured before use to ensure stability and skin comfort. The pH of the prepared NG was determined at a temperature of 25 ± 2 °C using a pH meter. The viscosity of NG formulation was measured with a Brookfield viscometer (GallenKamp, England), and the results were given as mean ± SD. The spreadability of the NG formulation was determined by the previously described method16. The sample was placed between two glass slides to test the spreadability of the NG. 1 gm of NG was placed on the pre-marked glass slide had a circle with a diameter of 1 cm on its lower side, and the upper glass slide was covered on it. The weight of 200 g was placed on the upper glass slide for five minutes and the spreading of NG was measured using equation 1.

Spreadability (%) = Increase in diameter / Initial diameter × 100                 (1)

Drug content determination

Drug content determination was performed by dissolving the NG sample in 10 ml of ethanol and then diluting it with distilled water q.s 100 ml. Using 0.22μ membrane filters, the sample was filtered and thereafter subjected to analysis at 238 nm using a UV spectrophotometer13. Equation 2 was used to calculate the %drug content:

%Drug content = Observed drug content/Total amount of drug taken x 100                        (2)

In vitro drug permeation study

Franz diffusion cells were used to measure the in vitro drug permeation19. The cellophane membrane (MW 12-14000 Da) was used with a surface area of 4.9 cm2. The membrane was initially treated with pH 6.8 phosphate buffer for 30 minutes at 25oC. The treated cellophane membrane was kept between the donor and receptor compartments of the Franz diffusion cell. Then 1 gm of NG formulation was placed in the donor compartment whereas, pH 6.8 phosphate buffer was placed in the receptor compartment which was kept at 37 ± 0.5oC. 500 μl of aliquots were removed through the sampling port of the diffusion cell at predetermined intervals (0.5, 1, 2, 3, 4, 5, and 8 hours) and replenished with the same quantity of fresh receptor solution to maintain the sink condition. The aliquots were filtered and analysis was done at 238 nm using a UV spectrophotometer8.

Statistical analysis

The data are shown as mean ± SD. Graph Pad Prism version 5.0 (Graph Pad Software Inc., USA) was used to analyze all the data using one-way ANOVA.

Results and Discussions

Equilibrium solubility study

Linseed oil showed the highest solubility of LS (26.09 ± 0.234 mg/ml) among the various oils used for the solubility analysis. Similarly, the drug solubility was found 26.84 ± 1.23 mg/ml in the surfactant tween 80 and 30.59 ± 2.067 mg/ml in the co-surfactant PEG 400. Based on solubility analysis, linseed oil, Tween 80, and PEG 400 were used as the oil phase, surfactant, and co-surfactant to produce pseudo-ternary phase diagrams, as shown in Table 1.

Table 1: Solubility analysis of LS

Oil

Solubility of LS (mg/ml) ± SD (n=3)

Almond oil

12.44 ± 0.123

Sunflower oil

11.29 ± 0.32

Eucalyptus oil

12.48 ± 0.513

Linseed oil

26.09 ± 0.234

Olive oil

12.28 ± 1.09

Surfactants

Solubility of LS (mg/ml) ± SD (n=3)

Cremophor RH 40

19.29 ± 0.321

Tween 20

16.37 ± 0.34

Tween 60

14.06 ± 1.01

Tween 80

26.84 ± 1.23

Co-surfactants

Solubility of LS (mg/ml) ± SD (n=3)

Myoglyol

12.46 ± 0.147

Isopropyl myristate

12.74 ± 0.198

PEG 400

30.59 ± 2.067

Propylene glycol

15.54 ± 0.201

 

Formulation of nanoemulsions

To identify the NE region that resulted from combining the Smix and oil phases, ternary phase diagrams were prepared. When titrating the oil with water, Smix emulsifies it at specific ratios that cause NE production. Here, Smix was prepared by combining Tween 80 and PEG 400 at various ratios of 1:1, 1:2, and 2:1. As shown in Fig. 1, a broader region of the NE was produced when we used Smix at a 1:1 ratio. The larger area in the ternary phase diagram suggested that the nanoemulsifying effectiveness of formulation is better at the selected ratio (1:1) and better interaction between the aqueous phase, Smix, and the oil phase. The formulation should contain the minimum amounts of surfactants and co-surfactants because it has been documented that using too much surfactant can induce skin irritation and toxicity-related problems20,21. However, as co-surfactant concentration increased, the nanoemulsion zone reduced significantly. Whereas the nanoemulsion region in the ternary phase diagram increases as the surfactant concentration in the Smix increases. Since a high surfactant concentration irritates the skin18, therefore, we chose Smix in a 1:1 ratio. Table 2 represents the composition of different formulations at the Smix of 1:1 ratio. 

Figure 1: Ternary phase diagram prepared using Smix in 1:1

Click here to View Figure

Table 2: Composition of various nanoemulsion formulations having Smix (1:1).

Formulations

Oil

Smix

Distilled water

LS

NE1

5

20

75

2.5

NE2

5

35

60

2.5

NE3

10

35

55

2.5

NE4

15

30

55

2.5

NE5

15

35

50

2.5

NE6

20

30

50

2.5

* Weight taken as %w/w

Physical stability of nanoemulsion formulations  

The physical stability of the NEs (NE1–NE6) during centrifugation testing, freeze-thaw cycles, and heating-cooling cycles is shown in Table 3. When subjected to the centrifugation test and the freeze-thaw cycles, NE1 did not pass the physical stability study. When subjected to centrifugation, NE2 was not stable. When subjected to the freeze-thaw and heating-cooling cycles, NE4 and NE6 failed the stability investigation. However, when the NE5 formulation was subjected to the freeze-thaw cycle, its stability failed. The single formulation, NE3, is the one that passes all of the thermodynamic stability tests and was used for further characterization.

Table 3: Thermodynamic stability of selected formulation of nanoemulsion

Formulations

Heating-cooling cycles

Centrifugation cycles

Freeze-thaw cycles

Inferences

NE1

x

x

Failed

NE2

x

Failed

NE3

Passed

NE4

x

x

Failed

NE5

x

Failed

NE6

x

x

Failed

 

Characterization of nanoemulsions

DS, PDI, and ZP analysis

DS is an important characteristic of NE as it shows how the NE droplet behaves. Droplet penetration will occur more quickly and deeply in the skin when droplets are smaller in size22. The optimized LS-loaded NEs (NE3) have an average DS of 108.9 ± 3.12 nm with the PDI value of 0.257 ± 0.015 (Fig. 2). NE droplet size is within the intended range i.e. less than 200 nm. PDI indicates the uniformity of droplet size in the NEs formulation. A low value of PDI (≤ 0.5) indicates the higher stability of NE formulation23. It was possible to identify the homogeneous droplet size distribution in the NE as the PDI of the NE3 formulation was less than 0.5. ZP quantifies the charge on the NE droplets’ surface; the prepared NE3 formulation had a negative ZP of −16.93 ± 1.12 mV (Table 4).

Table 4: DS, PDI, and ZP of NE3 (n = 3).

Optimized Formulation Code

DS

(nm)

PDI

ZP (mV)

NE3

108.9 ± 3.12

0.257± 0.015

− 16.93 ± 1.12

 

Figure 2: Size distribution of NE3 formulation

Click here to View Figure

TEM analysis

The morphology and shape of the NE’s nanoscale globules are visible in TEM photomicrographs. The obtained NE3 formulation photomicrographs show that the sphere-shaped globules and their nano size were confirmed as shown in Fig. 3. Additionally, there was no evidence of globule coalescence, demonstrating the formulation’s physical stability24.

Figure 3: TEM images of optimized NE3 batch NE

Click here to View Figure

Preparation and characterization of nanogel

Thermodynamic stability and nanosize range of NE3 nanoemulsion formulations led to their inclusion in the gel matrix to prepare NG. To prepare the NG, the NE3 nanoemulsion formulation was mixed in a 1:1 ratio with the carbopol 934 gel base. At room temperature, the mixture was stirred until the nanogels became transparent and uniform. For transdermal preparations, pH is a crucial parameter since the formulation’s pH should be close to the skin’s pH. The prepared NG formulations had a pH of 6.6 ± 0.11. Further, the viscosity of the NG formulations was found 23286 ± 3.85 cp. In the NG formulation, the drug content of LS was 98.78 ± 1.39% which showed that the drug was uniformly distributed throughout the NG system. The gel extrusion test indicates how easily the formulation can be extruded from the tube. Extrudability was found 15.46 ± 1.82 g/cm2 which shows that the NG formulation can extrude out of the tube. Table 5 shows the results of drug content, pH, viscosity, and extrudability of developed NG.

Table 5: pH, viscosity, drug content, and extrudability of the NE gel formulation (n= 3).

Parameters

NE gel formulation

pH

6.6 ± 0.11

Viscosity (cP)

23286 ± 3.85

Drug content uniformity (%)

98.78 ± 1.39

Extrudability (g/cm2)

15.46 ± 1.82

 

In-vitro drug permeation study

Fig. 4 shows the % drug penetration from NE, NGs, and drug suspension during the in vitro permeation study. It was observed that the formulation of NE and NG has a high permeation rate as compared to drug suspension. The formulation with the highest flux value was NE (36.63± 0.55 µg/cm2/hr), followed by NGs (33.69± 0.75 µg/cm2/hr). Compared to the formulations, the drug suspension permeation flux value (6.41±1.13 µg/cm2/hr) was much lower. It is clear from the flux value that LS permeation has been greatly increased by nanosized droplets. Therefore, the formulation exhibited improved permeation capabilities due to the addition of nano-sized droplets of NE, which aid in crossing different stratum corneum barriers.

Figure 4: Cumulative % of LS permeated from NE and NGs formulations in 8 hours

Click here to View Figure

Conclusion

LS-loaded NG formulation was developed using linseed oil, tween 80 along with PEG 400 as surfactant, co-surfactant (Smix in 1:1), and cabopol 934 as a gelling agent for transdermal delivery of LS to improve its bio-performance and overcome the drawback associated with its oral delivery. Different components of the formulation were selected based on solubility and the NEs were prepared by spontaneous emulsification method. The thermodynamically stable NE was selected and characterized for various parameters. The NEs formulation was found in the nano range (< 200 nm) which is suitable for transdermal delivery of LS. Finally, NE3 formulation was converted into NG using cabopol 934. In vitro permeation study was performed for NE, NG, and pure drug suspension. The NG and NE formulation demonstrated a more than 5-fold higher permeation flux rate than that of pure LS suspension in the permeation study. Therefore, the LS-loaded NG formulation was found effective in terms of in vitro permeation performance. So, it is concluded that the NG-based formulation of LS is a potential approach for treating hyperlipidemia and could be an alternative to oral formulation. Moreover, detailed pharmacokinetic and pharmacodynamic studies are required for the clinical outcomes of this investigation.

Acknowledgment

The authors gratefully acknowledge the approval and the support of this research study by grant no. PHAR-2023-12-2300 from the Deanship of Scientific Research at Northern Border University, Arar, K.S.A.

Conflict of Interest

The authors report no conflicts of interest.

References

  1. Mujtaba, M. A.; Alotaibi, N. M. J. Pharm. Res. Int. 2020, 32, 50–56.
    CrossRef
  2. Gesto, D. S.; Pereira, C. M. S.; Cerqueira, N. M. F. S.; Sousa, S. F. Molecules. 2020, 25(17), 3891.
    CrossRef
  3. Andersson, T.; Nåtman, J.; Mourtzinis, G.; Bager, J. E.; Bengtsson, B. K.; Franzén, S.; Hjerpe, P. Eur J Prev Cardiol. 2023, 30(17), 1883-1894.
    CrossRef
  4. Gaber, D. A. Int J Nanomedicine. 2020, 15,4225-4236.
    CrossRef
  5. Akl, M.A.; Ryad, S.; Ibrahim, M. F.; Kassem, A. A. Int J Pharm. 2023, 638, 122917.
    CrossRef
  6. Alam, M. S.; Ali, M. D.; Ahmad, S.; Banu, N.; Ali, M. S.; Alam, N.; Ali, M.; Ansari, M. S.; Shamim, M.; Mujtaba, M. A.  Pak J Pharm Sci. 2021, 34, 1385-1392.
  7. Ahad, A.; Al-Saleh, A. A.; Al-Mohizea, A. M.; Al-Jenoobi, F. I.; Raish, M.; Yassin, A. E. B.; Alam, M. A. Saudi Pharm J. 2017, 25(7), 1040-1046.
    CrossRef
  8. Kaur, R.; Ajitha, M. J Drug Deliv Sci Technol. 2019, 52, 968–978.
    CrossRef
  9. Soujanya, C.; Ravi, P. P. Int. J. Pharm. Sci. Nanotechnol. 2018, 11(4), 4196–4207.
  10. Gupta, D. K.; Sharma, S. K.; Gaur, P. K.; Singh, A. K. Res J Pharm Technol. 2022, 15(3), 1085-1089.
  11. Spoorthy, M.; Ramesh, K.; Kumar, G.V. Res J Pharm Technol.2023, 1391-1400.
  12. Avelino, A.P.; Oliveira, G.M.; Ferreira, C.C.; Luiz, R.R.; Rosa, G. Clin Interv Aging. 2015, 10, 1679-1685.
  13. Mahajan, N.; Mujtaba, M. A.; Fule, R.; Thakre, S.; Akhtar, M. S.; Alavudeen, S. S.; Anwer, M. K.; Aldawsari, M. F.; Mahmood, D.; Alam, M. S. ACS omega. 2024, 9(7), 8139–8150.
  14. Zhang Y, Zhang H, Che E, Zhang L, Han J, Yang Y, Wang S, Zhang M, Gao C. (2015). Colloids Surf B Biointerfaces. 2015, 128, 77-85.
    CrossRef
  15. Khan, R. U.; Shah, S. U.; Rashid, S. A.; Naseem, F.; Shah, K. U.; Farid, A.; Hakeem, K.R.; Kamli, M. R.; Althubaiti, E. H. Alamoudi, S. A. Polymers. 2022, 14(9), 1922.
    CrossRef
  16. Salawi, A.; Almoshari, Y.; Sultan, M. H.; Madkhali, O. A.; Bakkari, M. A.; Alshamrani, M.; Safhi, A. Y.; Sabei, F. Y.; Al Hagbani, T.; Ali, M. S.; Alam, M. S. Pharmaceuticals. 2023, 16(4), 490.
    CrossRef
  17. Mujtaba, M. A.; Alotaibi, N. M. Pak J Pharm Sci. 2023, 36(2), 535-540.
    CrossRef
  18. Mujtaba, M. A.; Alotaibi, N. M.; Alshehri, S. M.; Yusuf, M.; Anwer, M. K.; Rahman, M. A.; Parveen, A. Polymers. 2022, 14, 4344. 
    CrossRef
  19. Alam, M. S.; Algahtani, M. S.; Ahmad, J.; Kohli, K.; Shafiq-Un-Nabi, S.; Warsi, M. H.; Ahmad, M. Z. Ther Deliv. 2020, 11, 767-778.
    CrossRef
  20. Ahmad, J.; Gautam, A.; Komath, S.; Bano, M.; Garg, A.; Jain, K. Recent Pat. Antiinfect Drug Discov. 2019, 14, 36–48.
    CrossRef
  21. Roy, A.; Nishchaya, K.; Rai, V.K. Expert Opin Drug Deliv. 2022, 19(3), 303-319.
    CrossRef
  22. Anjani, Q.K.; Sabri, A. H. B.; Utomo, E. Mol Pharm. 2022, 19(4), 1191-1208. 
    CrossRef
  23. Bhardwaj, S.; Bhatia, S.; Gupta, P.S.; Singh, S. Iran J Basic Med Sci. 2022, 25(3), 352-363.
  24. Aqil, M.; Kamran, M.; Ahad, A.; Imam, S. S. J. Mol. Liq. 2016, 214, 238–248.
    CrossRef

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

About The Author