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
ReviewOpen Accesscc iconby icon

The significance of quercetin-loaded advanced nanoformulations for the management of diabetic wounds

    Vijay Kumar Panthi‡

    Department of Pharmacy, College of Pharmacy & Natural Medicine Research Institute, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea

    ‡Both authors contributed equally

    Search for more papers by this author

    ,
    Mohammad Imran‡

    Therapeutic Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia

    ‡Both authors contributed equally

    Search for more papers by this author

    ,
    Arshi Chaudhary

    Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India

    ,
    Keshav Raj Paudel

    Department of Oriental Medicine Resources, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea

    &
    Yousuf Mohammed

    *Author for correspondence: Tel.: +61 433 853 534;

    E-mail Address: y.mohammed@uq.edu.au

    Therapeutic Research Group, Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia

    School of Pharmacy, The University of Queensland, Brisbane, 4102, Australia

    Published Online:https://doi.org/10.2217/nnm-2022-0281

    Quercetin is a well-known plant flavanol that exhibits multiple biological activities, including antioxidant, anti-inflammatory and anticancer activities. The role of quercetin in wound healing has been widely explored by a range of researchers in different models. However, the physicochemical properties, such as solubility and permeability, of this compound are low, which ultimately limits its bioavailability on the target site. To overcome these limitations for successful therapy, scientists have developed a range of nanoformulations that provide effective therapeutic potential. In this review, the broad mechanism of quercetin for acute and chronic wounds is covered. A compilation of recent advances on the horizon of wound healing via quercetin is incorporated with several advanced nanoformulations.

    Graphical abstract

    References

    • 1. Wan R, Weissman JP, Grundman K et al. Diabetic wound healing: the impact of diabetes on myofibroblast activity and its potential therapeutic treatments. Wound Repair Regen. 29(4), 573–581 (2021).
    • 2. Fu J, Huang J, Lin M et al. Quercetin promotes diabetic wound healing via switching macrophages from M1 to M2 polarization. J. Surg. Res. 246, 213–223 (2020).
    • 3. Kant V, Jangir BL, Sharma M et al. Topical application of quercetin improves wound repair and regeneration in diabetic rats. Immunopharmacol. Immunotoxicol. 43(5), 536–553 (2021).
    • 4. Bai Q, Han K, Dong K et al. Potential applications of nanomaterials and technology for diabetic wound healing. Int. J. Nanomed. 15, 9717–9743 (2020).
    • 5. Pradeepa R, Mohan V. Epidemiology of type 2 diabetes in India. Indian J. Ophthalmol. 69(11), 2932–2938 (2021).
    • 6. Ighodaro OM. Molecular pathways associated with oxidative stress in diabetes mellitus. Biomed. Pharmacother. 108, 656–662 (2018).
    • 7. Wadhwa K, Kadian V, Puri V et al. New insights into quercetin nanoformulations for topical delivery. Phytomedicine Plus 2(2), 100257 (2022).
    • 8. Vilian AE, Puthiaraj P, Kwak CH et al. Electrochemical determination of quercetin based on porous aromatic frameworks supported Au nanoparticles. Electrochimica Acta. 216, 181–187 (2016).
    • 9. Gao Y, Wang Y, Ma Y et al. Formulation optimization and in situ absorption in rat intestinal tract of quercetin-loaded microemulsion. Colloids Surf. B Biointerfaces 71(2), 306–314 (2009).
    • 10. Qi M, Zhou Q, Zeng W et al. Growth factors in the pathogenesis of diabetic foot ulcers. Front. Biosci. Landmark 23(2), 310–317 (2018).
    • 11. Kant V, Sharma M, Jangir BL, Kumar V. Acceleration of wound healing by quercetin in diabetic rats requires mitigation of oxidative stress and stimulation of the proliferative phase. Biotechnic Histochem. 97(6), 461–472 (2022).
    • 12. Ahmad M, Sultana M, Raina R et al. Hypoglycemic, hypolipidemic, and wound healing potential of quercetin in streptozotocin-induced diabetic rats. Pharmacogn. Mag. 13(Suppl. 3), S633–S639 (2017).
    • 13. Padhi S, Nayak AK, Behera A. Type II diabetes mellitus: a review on recent drug based therapeutics. Biomed. Pharmacother. 131, 110708 (2020).
    • 14. Uppal S, Italiya KS, Chitkara D, Mittal A. Nanoparticulate-based drug delivery systems for small molecule anti-diabetic drugs: an emerging paradigm for effective therapy. Acta Biomater. 81, 20–42 (2018).
    • 15. Vicentini FT, Simi TR, Del Ciampo JO et al. Quercetin in w/o microemulsion: in vitro and in vivo skin penetration and efficacy against UVB-induced skin damages evaluated in vivo. Eur. J. Pharm. Biopharm. 69(3), 948–957 (2008).
    • 16. Liu D, Hu H, Lin Z et al. Quercetin deformable liposome: preparation and efficacy against ultraviolet B induced skin damages in vitro and in vivo. J. Photochem. Photobiol. Biol. 127, 8–17 (2013).
    • 17. Hasan N, Imran M, Sheikh A et al. Cannabis as a potential compound against various malignancies, legal aspects, advancement by exploiting nanotechnology and clinical trials. J. Drug Target. 30(7), 709–725 (2022).
    • 18. Rai VK, Mishra N, Yadav KS, Yadav NP. Nanoemulsion as pharmaceutical carrier for dermal and transdermal drug delivery: formulation development, stability issues, basic considerations and applications. J. Control. Rel. 270, 203–225 (2018).
    • 19. Tapfumaneyi P, Imran M, Mohammed YH, Roberts M. Recent advances and future prospective of topical and transdermal delivery systems. Front. Drug Deliv. 2, 957732 doi: 10.3389/fddev.2022.957732 (2022).
    • 20. Kong M, Chen XG, Kweon DK, Park HJ. Investigations on skin permeation of hyaluronic acid based nanoemulsion as transdermal carrier. Carbohydrate Polymers 86(2), 837–843 (2011).
    • 21. Yilmaz E, Borchert H-H. Design of a phytosphingosine-containing, positively-charged nanoemulsion as a colloidal carrier system for dermal application of ceramides. Eur. J. Pharm. Biopharm. 60(1), 91–98 (2005).
    • 22. Bose S, Michniak-Kohn B. Preparation and characterization of lipid based nanosystems for topical delivery of quercetin. Eur. J. Pharm. Sci. 48(3), 442–452 (2013).
    • 23. Park JW, Hwang SR, Yoon I-S. Advanced growth factor delivery systems in wound management and skin regeneration. Molecules. 22(8), 1259 (2017).
    • 24. Pereira RF, Bartolo PJ. Traditional therapies for skin wound healing. Adv. Wound Care 5(5), 208–229 (2016).
    • 25. Groeber F, Holeiter M, Hampel M et al. Skin tissue engineering – in vivo and in vitro applications. Adv. Drug Deliv. Rev. 63(4–5), 352–366 (2011).
    • 26. Guo S, DiPietro LA. Factors affecting wound healing. J. Dental Res. 89(3), 219–229 (2010).
    • 27. Krasner D. Minimizing factors that impair wound healing: a nursing approach. Ostomy/Wound Manage. 41(1), 22–26; 28, 30; quiz 31 (1995).
    • 28. Witte MB, Barbul A. General principles of wound healing. Surg. Clin. North Am. 77(3), 509–528 (1997).
    • 29. Okonkwo UA, DiPietro LA. Diabetes and wound angiogenesis. Int. J. Mol. Sci. 18(7), 1419 (2017).
    • 30. Zhao R, Liang H, Clarke E et al. Inflammation in chronic wounds. Int. J. Mol. Sci. 17(12), 2085 (2016).
    • 31. Brem H, Tomic-Canic M. Cellular and molecular basis of wound healing in diabetes. J. Clin. Invest. 117(5), 1219–1222 (2007).
    • 32. Hassan WU, Greiser U, Wang W. Role of adipose-derived stem cells in wound healing. Wound Repair Regen. 22(3), 313–325 (2014).
    • 33. Kato J, Kamiya H, Himeno T et al. Mesenchymal stem cells ameliorate impaired wound healing through enhancing keratinocyte functions in diabetic foot ulcerations on the plantar skin of rats. J. Diabetes Complications 28(5), 588–595 (2014).
    • 34. Kota SK, Meher LK, Jammula S et al. Aberrant angiogenesis: the gateway to diabetic complications. Indian J. Endocrinol. Metab. 16(6), 918 (2012).
    • 35. Ellis S, Lin EJ, Tartar D. Immunology of wound healing. Curr. Dermatol. Rep. 7(4), 350–358 (2018).
    • 36. Cañedo-Dorantes L, Cañedo-Ayala M. Skin acute wound healing: a comprehensive review. Int. J. Inflamm. 2019, 3706315 (2019).
    • 37. Raziyeva K, Kim Y, Zharkinbekov Z et al. Immunology of acute and chronic wound healing. Biomolecules 11(5), 700 (2021).
    • 38. Larouche J, Sheoran S, Maruyama K, Martino MM. Immune regulation of skin wound healing: mechanisms and novel therapeutic targets. Adv. Wound Care 7(7), 209–231 (2018).
    • 39. Han G, Ceilley R. Chronic wound healing: a review of current management and treatments. Adv. Ther. 34(3), 599–610 (2017).
    • 40. Badhwar R, Mangla B, Neupane YR et al. Quercetin loaded silver nanoparticles in hydrogel matrices for diabetic wound healing. Nanotechnology 32(50), 1–13 (2021).
    • 41. Ayyanar M, Ignacimuthu S. Herbal medicines for wound healing among tribal people in southern India: ethnobotanical and scientific evidences. Int. J. Appl. Res. Nat. Prod. 2(3), 29–42 (2009).
    • 42. Maver T, Maver U, Stana Kleinschek K et al. A review of herbal medicines in wound healing. Int. J. Dermatol. 54(7), 740–751 (2015).
    • 43. Kwakman PH, Zaat SA. Antibacterial components of honey. IUBMB Life 64(1), 48–55 (2012).
    • 44. Leyden JJ, Kligman AM. Contact dermatitis to neomycin sulfate. JAMA 242(12), 1276–1278 (1979).
    • 45. Storm-Versloot MN, Vos CG, Ubbink DT, Vermeulen H. Topical silver for preventing wound infection. Cochrane Database Syst. Rev. (3), CD006478 (2010).
    • 46. Alexiadou K, Doupis J. Management of diabetic foot ulcers. Diabetes Ther. 3(1), 1–15 (2012).
    • 47. Boateng JS, Matthews KH, Stevens HN, Eccleston GM. Wound healing dressings and drug delivery systems: a review. J. Pharm. Sci. 97(8), 2892–2923 (2008).
    • 48. Tibbles PM, Edelsberg JS. Hyperbaric-oxygen therapy. New Engl. J. Med. 334(25), 1642–1648 (1996).
    • 49. Caldwell MD. Bacteria and antibiotics in wound healing. Surg. Clin. 100(4), 757–776 (2020).
    • 50. Brett D. A review of collagen and collagen-based wound dressings. Wounds 20(12), 347–356 (2008).
    • 51. Singh O, Gupta SS, Soni M et al. Collagen dressing versus conventional dressings in burn and chronic wounds: a retrospective study. J. Cutan. Aesthet. Surg. 4(1), 12 (2011).
    • 52. Mi Y, Zhong L, Lu S et al. Quercetin promotes cutaneous wound healing in mice through Wnt/beta-catenin signaling pathway. J. Ethnopharmacol. 290, 115066 (2022).
    • 53. Hara M, Verkman A. Glycerol replacement corrects defective skin hydration, elasticity, and barrier function in aquaporin-3-deficient mice. Proc. Natl Acad. Sci. 100(12), 7360–7365 (2003).
    • 54. Hatahet T, Morille M, Hommoss A et al. Quercetin topical application, from conventional dosage forms to nanodosage forms. Eur. J. Pharm. Biopharm. 108, 41–53 (2016).
    • 55. Lee HN, Shin SA, Choo GS et al. Anti-inflammatory effect of quercetin and galangin in LPS-stimulated RAW264. 7 macrophages and DNCB-induced atopic dermatitis animal models. Int. J. Mol. Med. 41(2), 888–898 (2018).
    • 56. Terao J, Piskula M, Yao Q. Protective effect of epicatechin, epicatechin gallate, and quercetin on lipid peroxidation in phospholipid bilayers. Arch. Biochem. Biophys. 308(1), 278–284 (1994).
    • 57. Park YR, Sultan MT, Park HJ et al. NF-kappaB signaling is key in the wound healing processes of silk fibroin. Acta Biomater. 67, 183–195 (2018).
    • 58. Yang HL, Tsai YC, Korivi M et al. Lucidone promotes the cutaneous wound healing process via activation of the PI(3)K/AKT, Wnt/beta-catenin and NF-kappaB signaling pathways. Biochim. Biophys. Acta Mol. Cell. Res. 1864(1), 151–168 (2017).
    • 59. Xuan Y, Chi L, Tian H et al. The activation of the NF-kappaB-JNK pathway is independent of the PI3K-Rac1-JNK pathway involved in the bFGF-regulated human fibroblast cell migration. J. Dermatol. Sci. 82(1), 28–37 (2016).
    • 60. Liu T, Zhang L, Joo D, Sun SC. NF-kappaB signaling in inflammation. Signal Transduct. Target Ther. 2, 17023 (2017).
    • 61. Ambrozova N, Ulrichova J, Galandakova A. Models for the study of skin wound healing. The role of Nrf2 and NF-kappaB. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc. Czech Repub. 161(1), 1–13 (2017).
    • 62. Self DJ. The use of animals in medical education and research. Theoret. Med. 10(1), 9–19 (1989).
    • 63. Majtan J, Bohova J, Garcia-Villalba R et al. Fir honeydew honey flavonoids inhibit TNF-α-induced MMP-9 expression in human keratinocytes: a new action of honey in wound healing. Arch. Dermatol. Res. 305(7), 619–627 (2013).
    • 64. Rothwell JA, Day AJ, Morgan MR. Experimental determination of octanol–water partition coefficients of quercetin and related flavonoids. J. Agric. Food Chem. 53(11), 4355–4360 (2005).
    • 65. Monika P, Chandraprabha MN, Rangarajan A et al. Challenges in healing wound: role of complementary and alternative medicine. Front. Nutr. 8, 791899 (2021).
    • 66. Hamdy A, Ibrahem M. Management of aphthous ulceration with topical quercetin: a randomized clinical trial. J. Contemp. Dent. Pract. 11(4), E009–016 (2010).
    • 67. Taskan MM, Yuce HB, Karatas O, Gevrek F. Topical quercetin gel application improved wound healing in Wistar rats. Ann. Med. Res. 26(10), 2397–2404 (2019).
    • 68. Yin G, Wang Z, Wang Z, Wang X. Topical application of quercetin improves wound healing in pressure ulcer lesions. Exp. Dermatol. 27(7), 779–786 (2018).
    • 69. Bhatia N, Kaur G, Soni V et al. Evaluation of the wound healing potential of isoquercetin-based cream on scald burn injury in rats. Burns Trauma 4, 7 (2016).
    • 70. Doersch KM, Newell-Rogers MK. The impact of quercetin on wound healing relates to changes in αV and β1 integrin expression. Exp. Biol. Med. 242(14), 1424–1431 (2017).
    • 71. Ahmad A, Imran M, Sharma N. Precision nanotoxicology in drug development: current trends and challenges in safety and toxicity implications of customized multifunctional nanocarriers for drug-delivery applications. Pharmaceutics 14(11), 2463 (2022).
    • 72. Tapfumaneyi P, Imran M, Alavi SE, Mohammed Y. Science of, and insights into, thermodynamic principles for dermal formulations. Drug Discov. Today 28(5), 103521 (2023).
    • 73. Jin X, Imran M, Mohammed Y. Topical semisolid products-understanding the impact of metamorphosis on skin penetration and physicochemical properties. Pharmaceutics 14(11), 2487 (2022).
    • 74. Roberts MS, Cheruvu HS, Mangion SE et al. Topical drug delivery: history, percutaneous absorption, and product development. Adv. Drug Deliv. Rev. 177, 113929 (2021).
    • 75. Jangde R, Srivastava S, Singh MR, Singh D. In vitro and in vivo characterization of quercetin loaded multiphase hydrogel for wound healing application. Int. J. Biol. Macromol. 115, 1211–1217 (2018).
    • 76. Jee J-P, Pangeni R, Jha SK et al. Preparation and in vivo evaluation of a topical hydrogel system incorporating highly skin-permeable growth factors, quercetin, and oxygen carriers for enhanced diabetic wound-healing therapy. Int. J. Nanomed. 14, 5449 (2019).
    • 77. Ajmal G, Bonde GV, Mittal P et al. Biomimetic PCL-gelatin based nanofibers loaded with ciprofloxacin hydrochloride and quercetin: a potential antibacterial and anti-oxidant dressing material for accelerated healing of a full thickness wound. Int. J. Pharm. 567, 118480 (2019).
    • 78. Choudhary A, Kant V, Jangir BL, Joshi V. Quercetin loaded chitosan tripolyphosphate nanoparticles accelerated cutaneous wound healing in Wistar rats. Eur. J. Pharm. 880, 173172 (2020).
    • 79. Chen Y-B, Zhang Y-B, Wang Y-L et al. A novel inhalable quercetin-alginate nanogel as a promising therapy for acute lung injury. J. Nanobiotechnol. 20(1), 1–17 (2022).
    • 80. Jangde R, Singh D. Preparation and optimization of quercetin-loaded liposomes for wound healing, using response surface methodology. Artificial Cells Nanomed. Biotechnol. 44(2), 635–641 (2016).
    • 81. Gupta A. Nanoemulsions. Nanoparticles for Biomedical Applications. Elsevier, VA, USA, 371–384 (2020).
    • 82. Solans C, Solé I. Nano-emulsions: formation by low-energy methods. Curr. Opin. Colloid Interface Sci. 17(5), 246–254 (2012).
    • 83. Thakur N, Garg G, Sharma P, Kumar N. Nanoemulsions: a review on various pharmaceutical application. Global J. Pharmacol. 6(3), 222–225 (2012).
    • 84. Bonferoni MC, Rossi S, Sandri G et al. Nanoemulsions for “nose-to-brain” drug delivery. Pharmaceutics 11(2), 84 (2019).
    • 85. Sahoo S, Parveen S, Panda J. The present and future of nanotechnology in human health care. Nanomed. Nanotechnol. Biol. Med. 3(1), 20–31 (2007).
    • 86. Zorzi GK, Caregnato F, Moreira JCF et al. Antioxidant effect of nanoemulsions containing extract of Achyrocline satureioides (Lam) DC – Asteraceae. AAPS PharmSciTech. 17(4), 844–850 (2016).
    • 87. Singh R, Lillard JW Jr. Nanoparticle-based targeted drug delivery. Exp. Mol. Pathol. 86(3), 215–223 (2009).
    • 88. Wang W, Lu KJ, Yu CH et al. Nano-drug delivery systems in wound treatment and skin regeneration. J. Nanobiotechnol. 17(1), 82 (2019).
    • 89. Ribeiro MC, Correa VLR, Silva F et al. Wound healing treatment using insulin within polymeric nanoparticles in the diabetes animal model. Eur. J. Pharm. Sci. 150, 105330 (2020).
    • 90. Mohammed Y, Holmes A, Kwok PCL et al. Advances and future perspectives in epithelial drug delivery. Adv. Drug Deliv. Rev. 186, 114293 (2022).
    • 91. Bairagi U, Mittal P, Singh J, Mishra B. Preparation, characterization, and in vivo evaluation of nano formulations of ferulic acid in diabetic wound healing. Drug Dev. Ind. Pharm. 44(11), 1783–1796 (2018).
    • 92. Robson A-L, Dastoor PC, Flynn J et al. Advantages and limitations of current imaging techniques for characterizing liposome morphology. Front. Pharmacol. 9, 80 (2018).
    • 93. Sahoo SK, Labhasetwar V. Nanotech approaches to drug delivery and imaging. Drug Discov. Today 8(24), 1112–1120 (2003).
    • 94. Akbarzadeh A, Rezaei-Sadabady R, Davaran S et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 8(1), 1–9 (2013).
    • 95. Manca ML, Matricardi P, Cencetti C et al. Combination of argan oil and phospholipids for the development of an effective liposome-like formulation able to improve skin hydration and allantoin dermal delivery. Int. J. Pharm. 505(1–2), 204–211 (2016).
    • 96. Xu HL, Chen PP, ZhuGe DL et al. Liposomes with silk fibroin hydrogel core to stabilize bfgf and promote the wound healing of mice with deep second-degree scald. Adv. Healthc. Mater. 6(19), 1700344 (2017).
    • 97. Imran M, Iqubal MK, Imtiyaz K et al. Topical nanostructured lipid carrier gel of quercetin and resveratrol: formulation, optimization, in vitro and ex vivo study for the treatment of skin cancer. Int. J. Pharm. 587, 119705 (2020).
    • 98. Imran M, Jha SK, Hasan N et al. Overcoming multidrug resistance of antibiotics via nanodelivery systems. Pharmaceutics 14(3), 586 (2022).
    • 99. Hasan N, Imran M, Nadeem M et al. Formulation and development of novel lipid-based combinatorial advanced nanoformulation for effective treatment of non-melanoma skin cancer. Int. J. Pharm. 632, 122580 (2023).
    • 100. Liu Q, Yang X, Sun J et al. Size-dependent biological effects of quercetin nanocrystals. Molecules 24(7), 1438 (2019).
    • 101. Kakran M, Shegokar R, Sahoo NG et al. Long-term stability of quercetin nanocrystals prepared by different methods. J. Pharm. Pharmacol. 64(10), 1394–1402 (2012).
    • 102. Müller RH, Mäder K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery – a review of the state of the art. Eur. J. Pharm. Biopharm. 50(1), 161–177 (2000).
    • 103. Khanna K, Sharma N, Rawat S et al. Intranasal solid lipid nanoparticles for management of pain: a full factorial design approach, characterization & gamma scintigraphy. Chem. Phys. Lipids 236, 105060 (2021).
    • 104. Hasan N, Imran M, Kesharwani P et al. Intranasal delivery of Naloxone-loaded solid lipid nanoparticles as a promising simple and non-invasive approach for the management of opioid overdose. Int. J. Pharm. 599, 120428 (2021).
    • 105. Imran M, Iqubal MK, Imtiyaz K et al. Topical nanostructured lipid carrier gel of quercetin and resveratrol: formulation, optimization, in vitro and ex vivo study for the treatment of skin cancer. Int. J. Pharm. 587, 119705 (2020).
    • 106. Makwana V, Jain R, Patel K et al. Solid lipid nanoparticles (SLN) of efavirenz as lymph targeting drug delivery system: elucidation of mechanism of uptake using chylomicron flow blocking approach. Int. J. Pharm. 495(1), 439–446 (2015).
    • 107. Sukhotnik I, Moati D, Shaoul R et al. Quercetin prevents small intestinal damage and enhances intestinal recovery during methotrexate-induced intestinal mucositis of rats. Food Nutr. Res. 62, 10.29219/fnr.v62.1327 (2018).
    • 108. Varrica C, Carvalheiro M, Faria-Silva C et al. Topical allopurinol-loaded nanostructured lipid carriers: a novel approach for wound healing management. Bioengineering (Basel) 8(12), 192 (2021).
    • 109. Lee HJ, Jeong M, Na YG et al. An EGF- and curcumin-co-encapsulated nanostructured lipid carrier accelerates chronic-wound healing in diabetic rats. Molecules. 25(20), 4610 (2020).
    • 110. Elmowafy M, Al-Sanea MM. Nanostructured lipid carriers (NLCs) as drug delivery platform: advances in formulation and delivery strategies. Saudi Pharm. J. 29(9), 999–1012 (2021).
    • 111. Chen H, Wang Y, Zhai Y et al. Development of a ropivacaine-loaded nanostructured lipid carrier formulation for transdermal delivery. Colloids Surf. A Physicochem. Engin. Aspects 465, 130–136 (2015).
    • 112. Liu L, Tang Y, Gao C et al. Characterization and biodistribution in vivo of quercetin-loaded cationic nanostructured lipid carriers. Colloids Surf. B Biointerfaces 115, 125–131 (2014).
    • 113. Ge X, Wei M, He S, Yuan W-E. Advances of non-ionic surfactant vesicles (niosomes) and their application in drug delivery. Pharmaceutics 11(2), 55 (2019).
    • 114. Khatoon M, Shah KU, Din FU et al. Proniosomes derived niosomes: recent advancements in drug delivery and targeting. Drug Deliv. 24(2), 56–69 (2017).
    • 115. Tseng Y-C, Mozumdar S, Huang L. Lipid-based systemic delivery of siRNA. Adv. Drug Deliv. Rev. 61(9), 721–731 (2009).
    • 116. Abdelkader H, Alani AW, Alany RG. Recent advances in non-ionic surfactant vesicles (niosomes), self-assembly, fabrication, characterization, drug delivery applications and limitations. Drug Deliv. 21(2), 87–100 (2014).
    • 117. Jadon PS, Gajbhiye V, Jadon RS et al. Enhanced oral bioavailability of griseofulvin via niosomes. AAPS Pharmscitech. 10(4), 1186–1192 (2009).
    • 118. Gharbavi M, Amani J, Kheiri-Manjili H et al. Niosome: a promising nanocarrier for natural drug delivery through blood–brain barrier. Adv. Pharm. Sci. 2018, 6847971 (2018).
    • 119. Mohapatra A, Uthaman S, Park I-K. Polyethylene glycol nanoparticles as promising tools for anticancer therapeutics. In: Polymeric Nanoparticles as a Promising Tool for Anti-Cancer Therapeutics. Academic Press, CA, USA, 205–231 (2019).
    • 120. Imran M, Saleem S, Chaudhuri A et al. Docetaxel: an update on its molecular mechanisms, therapeutic trajectory and nanotechnology in the treatment of breast, lung and prostate cancer. J. Drug Deliv. Sci. Technol. 60, 101959 (2020).
    • 121. Qiao Z-Y, Zhang R, Du F-S et al. Multi-responsive nanogels containing motifs of ortho ester, oligo (ethylene glycol) and disulfide linkage as carriers of hydrophobic anti-cancer drugs. J. Control. Rel. 152(1), 57–66 (2011).
    • 122. Neamtu I, Rusu AG, Diaconu A et al. Basic concepts and recent advances in nanogels as carriers for medical applications. Drug Deliv. 24(1), 539–557 (2017).
    • 123. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mat. 12(11), 991–1003 (2013).
    • 124. Zarekar NS, Lingayat VJ, Pande VV. Nanogel as a novel platform for smart drug delivery system. Nanosci. Nanotechnol. 4(1), 25–31 (2017).
    • 125. Hajialyani M, Tewari D, Sobarzo-Sanchez E et al. Natural product-based nanomedicines for wound healing purposes: therapeutic targets and drug delivery systems. Int. J. Nanomed. 13, 5023 (2018).
    • 126. Anumolu SS, Menjoge AR, Deshmukh M et al. Doxycycline hydrogels with reversible disulfide crosslinks for dermal wound healing of mustard injuries. Biomaterials 32(4), 1204–1217 (2011).
    • 127. Li M, Li H, Li X et al. A bioinspired alginate-gum arabic hydrogel with micro-/nanoscale structures for controlled drug release in chronic wound healing. ACS Appl. Mater. Interfaces 9(27), 22160–22175 (2017).
    • 128. Mahmoudi M, Gould LJ. Opportunities and challenges of the management of chronic wounds: a multidisciplinary viewpoint. Chron. Wound Care Manage. Res. 7, 27 (2020).
    • 129. Pierpont YN, Dinh TP, Salas RE et al. Obesity and surgical wound healing: a current review. Int. Scholar. Res. Notices 2014, 638936 (2014).
    • 130. Landén NX, Li D, Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell. Mol. Life Sci. 73(20), 3861–3885 (2016).
    • 131. Yip WL. Influence of oxygen on wound healing. Int. Wound J. 12(6), 620–624 (2015).