We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Quercetin nanocrystals prepared by a novel technique improve the dissolution rate and antifibrotic activity of quercetin

    Pegah Cheshmehnoor

    Department of Pharmaceutics & Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran

    ,
    Shahram Rabbani

    Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran

    &
    Azadeh Haeri

    *Author for correspondence: Tel.: +98 218 820 0212;

    E-mail Address: a_haeri@sbmu.ac.ir

    Department of Pharmaceutics & Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran

    Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran

    Published Online:https://doi.org/10.2217/nnm-2022-0032

    Aim: To develop quercetin nanocrystals by a simple approach and to evaluate their in vivo antifibrotic efficacy. Materials & methods: Nanosuspensions were fabricated by a thin-film hydration technique and ultrasonication. The influence of process variables on the average diameter of quercetin nanoparticles was investigated. Moreover, in vivo efficacy was investigated in an established murine CCl4-induced fibrosis model. Results: Nanocrystals showed a particle size of <400 nm. The optimized formulations showed an increase in dissolution rate and solubility. Quercetin nanocrystals markedly prevented fibrotic changes in the liver, as evidenced by mitigated histopathological changes and diminished aminotransferase levels and collagen accumulation. Conclusion: The findings reflect the promising potential of quercetin nanocrystals for liver fibrosis prevention.

    Graphical abstract

    Papers of special note have been highlighted as: •• of considerable interest

    References

    • 1. Parola M, Pinzani M. Liver fibrosis: pathophysiology, pathogenetic targets and clinical issues. Mol. Aspects Med. 65, 37–55 (2019). •• Provides an extensive review of the pathophysiology and clinical aspects of liver fibrosis.
    • 2. Berumen J, Baglieri J, Kisseleva T, Mekeel K. Liver fibrosis: pathophysiology and clinical implications. WIREs Mech. Dis. 13(1), e1499 (2021).
    • 3. Luangmonkong T, Suriguga S, Mutsaers HAM et al. Targeting oxidative stress for the treatment of liver fibrosis. Rev. Physiol. Biochem. Pharmacol. 175, 71–102 (2018).
    • 4. Li H. Advances in anti-hepatic fibrotic therapy with traditional Chinese medicine herbal formula. J. Ethnopharmacol. 251, 112442 (2020).
    • 5. Fraga CG, Croft KD, Kennedy DO, Tomás-Barberán FA. The effects of polyphenols and other bioactives on human health. Food Funct. 10(2), 514–528 (2019).
    • 6. Pingili RB, Challa SR, Pawar AK et al. A systematic review on hepatoprotective activity of quercetin against various drugs and toxic agents: evidence from preclinical studies. Phytother. Res. 34(1), 5–32 (2020). •• Provides a review of the hepatoprotective activity of quercetin.
    • 7. Li X, Jin Q, Yao Q et al. The flavonoid quercetin ameliorates liver inflammation and fibrosis by regulating hepatic macrophages activation and polarization in mice. Front. Pharmacol. 9, 72 (2018).
    • 8. Li X, Jin Q, Yao Q et al. Quercetin attenuates the activation of hepatic stellate cells and liver fibrosis in mice through modulation of HMGB1-TLR2/4-NF-κB signaling pathways. Toxicol. Lett. 261, 1–12 (2016).
    • 9. Wang R, Zhang H, Wang Y, Song F, Yuan Y. Inhibitory effects of quercetin on the progression of liver fibrosis through the regulation of NF-κB/IκBα, p38 MAPK, and Bcl-2/Bax signaling. Int. Immunopharmacol. 47, 126–133 (2017).
    • 10. Guo Y, Bruno RS. Endogenous and exogenous mediators of quercetin bioavailability. J. Nutr. Biochem. 26(3), 201–210 (2015).
    • 11. Lai F, Schlich M, Pireddu R, Fadda AM, Sinico C. Nanocrystals as effective delivery systems of poorly water-soluble natural molecules. Curr. Med. Chem. 26(24), 4657–4680 (2019). •• Provides a review of nanocrystals as promising delivery systems for hydrophobic natural agents.
    • 12. Chin WW, Parmentier J, Widzinski M, Tan EH, Gokhale R. A brief literature and patent review of nanosuspensions to a final drug product. J. Pharm. Sci. 103(10), 2980–2999 (2014).
    • 13. Wang Y, Zheng Y, Zhang L, Wang Q, Zhang D. Stability of nanosuspensions in drug delivery. J. Control. Rel. 172(3), 1126–1141 (2013).
    • 14. Verma V, Ryan KM, Padrela L. Production and isolation of pharmaceutical drug nanoparticles. Int. J. Pharm. 603, 120708 (2021).
    • 15. Zhang X, Li LC, Mao S. Nanosuspensions of poorly water soluble drugs prepared by top-down technologies. Curr. Pharm. Des. 20(3), 388–407 (2014).
    • 16. Sinha B, Müller RH, Möschwitzer JP. Bottom-up approaches for preparing drug nanocrystals: formulations and factors affecting particle size. Int. J. Pharm. 453(1), 126–141 (2013).
    • 17. Han X, Wang M, Ma Z, Xue P, Wang Y. A new approach to produce drug nanosuspensions CO2-assisted effervescence to produce drug nanosuspensions. Colloids Surf. B Biointerfaces 143, 107–110 (2016).
    • 18. Wang Y, Han X, Wang J, Wang Y. Preparation, characterization and in vivo evaluation of amorphous tacrolimus nanosuspensions produced using CO2-assisted in situ nanoamorphization method. Int. J. Pharm. 505(1–2), 35–41 (2016).
    • 19. Wang Y, Wang C, Zhao J, Ding Y, Li L. A cost-effective method to prepare curcumin nanosuspensions with enhanced oral bioavailability. J. Colloid Interface Sci. 485, 91–98 (2017).
    • 20. Katara R, Majumdar DK. Eudragit RL 100-based nanoparticulate system of aceclofenac for ocular delivery. Colloids Surf. B Biointerfaces 103, 455–462 (2013).
    • 21. Lewinska A, Adamczyk-Grochala J, Bloniarz D et al. AMPK-mediated senolytic and senostatic activity of quercetin surface functionalized Fe(3)O(4) nanoparticles during oxidant-induced senescence in human fibroblasts. Redox Biol. 28, 101337 (2020).
    • 22. Cai X, Fang Z, Dou J, Yu A, Zhai G. Bioavailability of quercetin: problems and promises. Curr. Med. Chem. 20(20), 2572–2582 (2013). •• Provides a review of approaches for enhancing the poor bioavailability of quercetin.
    • 23. Al-Kassas R, Bansal M, Shaw J. Nanosizing techniques for improving bioavailability of drugs. J. Control. Rel. 260, 202–212 (2017).
    • 24. Niu X, Wang X, Niu B et al. New IMB16-4 nanoparticles improved oral bioavailability and enhanced anti-hepatic fibrosis on rats. Pharmaceuticals (Basel) 15(1), (2022).
    • 25. Abdullah AS, Sayed I, El-Torgoman AMA et al. Green synthesis of silymarin–chitosan nanoparticles as a new nano formulation with enhanced anti-fibrotic effects against liver fibrosis. Int. J. Mol. Sci. 23(10), (2022).
    • 26. Russo E, Villa C. Poloxamer hydrogels for biomedical applications. Pharmaceutics 11(12), (2019).
    • 27. McCartney F, Jannin V, Chevrier S et al. Labrasol® is an efficacious intestinal permeation enhancer across rat intestine: ex vivo and in vivo rat studies. J. Control. Rel. 310, 115–126 (2019).
    • 28. Asadi A, Pourfattah F, Miklós Szilágyi I et al. Effect of sonication characteristics on stability, thermophysical properties, and heat transfer of nanofluids: a comprehensive review. Ultrason. Sonochem. 58, 104701 (2019).
    • 29. Malamatari M, Taylor KMG, Malamataris S, Douroumis D, Kachrimanis K. Pharmaceutical nanocrystals: production by wet milling and applications. Drug Discov. Today 23(3), 534–547 (2018).
    • 30. Lestari ML, Müller RH, Möschwitzer JP. Systematic screening of different surface modifiers for the production of physically stable nanosuspensions. J. Pharm. Sci. 104(3), 1128–1140 (2015).
    • 31. Leone F, Cavalli R. Drug nanosuspensions: a ZIP tool between traditional and innovative pharmaceutical formulations. Expert Opin. Drug Deliv. 12(10), 1607–1625 (2015).
    • 32. Fonte P, Reis S, Sarmento B. Facts and evidences on the lyophilization of polymeric nanoparticles for drug delivery. J. Control. Rel. 225, 75–86 (2016).
    • 33. Mohammady M, Mohammadi Y, Yousefi G. Freeze-drying of pharmaceutical and nutraceutical nanoparticles: the effects of formulation and technique parameters on nanoparticles characteristics. J. Pharm. Sci. 109(11), 3235–3247 (2020).
    • 34. Falsafi SR, Rostamabadi H, Assadpour E, Jafari SM. Morphology and microstructural analysis of bioactive-loaded micro/nanocarriers via microscopy techniques; CLSM/SEM/TEM/AFM. Adv. Colloid Interface Sci. 280, 102166 (2020).
    • 35. Chadha R, Bhandari S. Drug–excipient compatibility screening – role of thermoanalytical and spectroscopic techniques. J. Pharm. Biomed. Anal. 87, 82–97 (2014).
    • 36. Jacob S, Nair AB, Shah J. Emerging role of nanosuspensions in drug delivery systems. Biomater. Res. 24, 3 (2020). •• Provides a review of nanocrystals as promising delivery systems.
    • 37. Kaur N, Narang A, Bansal AK. Use of biorelevant dissolution and PBPK modeling to predict oral drug absorption. Eur. J. Pharm. Biopharm. 129, 222–246 (2018).
    • 38. Gupta R, Chen Y, Xie H. In vitro dissolution considerations associated with nano drug delivery systems. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 13(6), e1732 (2021).
    • 39. Hattori Y, Haruna Y, Otsuka M. Dissolution process analysis using model-free Noyes–Whitney integral equation. Colloids Surf. B Biointerfaces 102, 227–231 (2013).
    • 40. Shah DA, Murdande SB, Dave RH. A review: pharmaceutical and pharmacokinetic aspect of nanocrystalline suspensions. J. Pharm. Sci. 105(1), 10–24 (2016).
    • 41. Truzzi F, Tibaldi C, Zhang Y, Dinelli G, Amen ED. An overview on dietary polyphenols and their biopharmaceutical classification system (BCS). Int. J. Mol. Sci. 22(11), (2021).
    • 42. Khursheed R, Singh SK, Wadhwa S et al. Development of mushroom polysaccharide and probiotics based solid self-nanoemulsifying drug delivery system loaded with curcumin and quercetin to improve their dissolution rate and permeability: state of the art. Int. J. Biol. Macromol. 189, 744–757 (2021).
    • 43. Singh J, Mittal P, Bonde GV, Ajmal G, Mishra B. Design, optimization, characterization and in vivo evaluation of quercetin enveloped Soluplus®/P407 micelles in diabetes treatment. Artif. Cells Nanomed. Biotechnol. 46(Suppl. 3), S546–S555 (2018).
    • 44. Teixeira MC, Carbone C, Souto EB. Beyond liposomes: recent advances on lipid based nanostructures for poorly soluble/poorly permeable drug delivery. Prog. Lipid Res. 68, 1–11 (2017).
    • 45. Abdelkader H, Fathalla Z. Investigation into the emerging role of the basic amino acid L-lysine in enhancing solubility and permeability of BCS class II and BCS class IV drugs. Pharm. Res. 35(8), 160 (2018).
    • 46. Manzoor MF, Hussain A, Sameen A et al. Novel extraction, rapid assessment and bioavailability improvement of quercetin: a review. Ultrason. Sonochem. 78, 105686 (2021).
    • 47. Charalabidis A, Sfouni M, Bergström C, Macheras P. The Biopharmaceutics Classification System (BCS) and the Biopharmaceutics Drug Disposition Classification System (BDDCS): beyond guidelines. Int. J. Pharm. 566, 264–281 (2019).
    • 48. Li J, Wang Z, Zhang H, Gao J, Zheng A. Progress in the development of stabilization strategies for nanocrystal preparations. Drug Deliv. 28(1), 19–36 (2021).
    • 49. Bhattacharjee S. DLS and zeta potential – what they are and what they are not? J. Control. Rel. 235, 337–351 (2016).
    • 50. Abdelbary AA, Li X, El-Nabarawi M, Elassasy A, Jasti B. Effect of fixed aqueous layer thickness of polymeric stabilizers on zeta potential and stability of aripiprazole nanosuspensions. Pharm. Dev. Technol. 18(3), 730–735 (2013).
    • 51. Xu J, Liu X, Koyama Y et al. The types of hepatic myofibroblasts contributing to liver fibrosis of different etiologies. Front. Pharmacol. 5, 167 (2014).
    • 52. Sookoian S, Pirola CJ. Liver enzymes, metabolomics and genome-wide association studies: from systems biology to the personalized medicine. World J. Gastroenterol. 21(3), 711–725 (2015).
    • 53. Limdi JK, Hyde GM. Evaluation of abnormal liver function tests. Postgrad. Med. J. 79(932), 307–312 (2003).