Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2238 word(s) 2238 2021-11-03 10:06:57 |
2 Format correct Meta information modification 2238 2021-11-16 09:06:26 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Stefanov, S. Lipid Nanoparticulate Drug Delivery Systems and Skin Disorders. Encyclopedia. Available online: https://encyclopedia.pub/entry/16034 (accessed on 04 July 2024).
Stefanov S. Lipid Nanoparticulate Drug Delivery Systems and Skin Disorders. Encyclopedia. Available at: https://encyclopedia.pub/entry/16034. Accessed July 04, 2024.
Stefanov, Stefan. "Lipid Nanoparticulate Drug Delivery Systems and Skin Disorders" Encyclopedia, https://encyclopedia.pub/entry/16034 (accessed July 04, 2024).
Stefanov, S. (2021, November 16). Lipid Nanoparticulate Drug Delivery Systems and Skin Disorders. In Encyclopedia. https://encyclopedia.pub/entry/16034
Stefanov, Stefan. "Lipid Nanoparticulate Drug Delivery Systems and Skin Disorders." Encyclopedia. Web. 16 November, 2021.
Lipid Nanoparticulate Drug Delivery Systems and Skin Disorders
Edit

Lipid nanoparticles (LN) are recognized as promising drug delivery systems (DDS) in treating skin disorders. Solid lipid nanoparticles (SLN) together with nanostructured lipid carriers (NLC) exhibit excellent tolerability as these are produced from physiological and biodegradable lipids. Moreover, LN applied to the skin can improve stability, drug targeting, occlusion, penetration enhancement, and increased skin hydration compared with other drug nanocarriers. Furthermore, the features of LN can be enhanced by inclusion in suitable bases such as creams, ointments, gels (i.e., hydrogel, emulgel, bigel), lotions, etc. 

skin diseases lipid-based nanosystems solid lipid nanoparticles nanostructured lipid carriers

1. Introduction

1.1. Skin Disorders

The skin can be affected by various pathological changes, i.e., inflammatory, neoplastic, traumatic, hormonal, degenerative, and even hereditarily determined [1]. Infectious skin diseases such as bacterial, fungal or viral affect people and cause various dermatological problems. Chronic inflammatory skin diseases such as atopic dermatitis, allergic contact dermatitis, psoriasis, etc., are a consequence of infiltration of inflammatory T cells [2]. The schematic representation of various skin disorders is shown in Figure 1 [3][4][5][6][7][8][9][10][11][12][13][14].

Pharmaceuticals 14 01083 g003

Figure 1. Representation of the most common skin disorders [3][4][5][6][7][8][9][10][11][12][13][14].
In practice, most of the skin disorders are complicated, polygenic, and multifactorial [15]. This indicates that multiple factors, lifestyle, and the environment play a fundamental role in the clinical picture of the diseases [16].

1.2. Lipid Nanoparticulate Drug Delivery Systems

Lipid-based drug delivery systems (LBDDS) are formulations containing a dissolved or suspended drug substance in lipidic excipients [17]. LBDDS are a progressive strategy to formulate pharmaceuticals for topical delivery [18]. Liposomes, which are “pioneers” among lipid DDS, have been used to improve drug solubility and traditionally for topical and transdermal drug delivery.

Table 1 presents a brief overview of lipid-based drug delivery systems.

Table 1. Presentation of some lipid-based delivery systems.

Lipid-Based
Delivery System
Description Advantages Disadvantages
Nanovesicular carriers
Liposomes [19] Conventional single or multilayer vesicles. Formed by contact of biodegradable lipids with an aqueous medium. Widely used as drug carriers for hydrophilic and lipophilic molecules. Biocompatible and biodegradable lipids. Conventional production processes. Improved local delivery. Suitable for loading both hydrophobic and hydrophilic substances. Insufficient chemical and physical stability. Short half-life. Inadequate penetration into the viable epidermis and dermis. High production costs. Difficulties in scalability.
Transfersomes
[20][21][22]
Highly deformable, elastic or ultra-flexible liposomes. Vesicles, similar to conventional liposomes in terms of preparation and structure. Claimed to permeate as intact vesicles through the skin layers. Functionally deformed due to the presence of an edge activator. Smaller vesicle size, higher elasticity. Compared with conventional liposomes—better penetration through the skin. High membrane hydrophilicity and elasticity allow them to avoid aggregation and fusion under osmotic stress, unlike the conventional liposomes. Elasticity of these vesicles can be compromised by hydrophobic drug loading. Occlusive application and complete skin hydration limit transdermal delivery due to inhibition of transdermal hydration. Relatively high production costs. Absence of well-established regulatory guidance for skin delivery.
Ethosomes [23][24] Lipid vesicles are composed of phospholipids, ethanol, and water. Similar to liposomes in terms of their preparation techniques and structure. Concentration of ethanol 20–45%. Their size decreases with an increase in the ethanol concentration. Exhibit high encapsulation efficiency. Appropriate for both hydrophobic and hydrophilic drug loading. Enhanced skin delivery under both occlusive and nonocclusive conditions. Higher elasticity, smaller vesicle size, and higher entrapment efficiency than conventional liposomes. High ethanol content can lead to skin irritation and toxicity. Possible structural and chemical instability during long-term storage. Need to optimize the concentration of lipids and ethanol for improved physicochemical properties and stability of ethosomes.
Lipid nanoparticulate carriers
Solid lipid nanoparticles [25][26] Colloidal lipid nanoparticles are composed of a physiological biodegradable solid lipophilic matrix (solid at room temperature and body temperature), in which the drug molecules can be incorporated. Increased drug stability. High drug payload. Incorporation of lipophilic and hydrophilic drugs. Avoidance of organic solvents. Lack of biotoxicity of the carrier. Relatively cost-effective. SLN are incorporated into semisolid carriers such as ointments and gels due to the high water content. Potential expulsion of active compounds during storage. Cost-effective manufacturing process.
Nanostructure
Lipid Carriers [27][28]
Colloidal lipid nanoparticles composed of physiological mixing liquid lipid (oils) with the solid lipids, in which the liquid lipid is incorporated into the solid matrix or localized at the surface of solid particles Improved drug loading compared with SLN. Lower water content compared with SLN. Firmly incorporates the drug substance during storage. Biodegradable and biocompatible. Large-scale production is easily possible. Tendency to unpredictable gelation. Polymorphic transition. Low drug incorporation due to the crystalline structure of solid lipids. Lack of long-term stability data.
Lipospheres
[29][30][31]
Microspheres, composed of solid hydrophobic lipid core and stabilized by a monolayer of a phospholipid embedded on the surface. Improved drug stability, especially for photo-labile drugs. Possibility for controlled drug release. Controlled particle size. High drug loading. Biodegradable and biocompatible. Larger particle size and poor skin permeation compared with lipid-based vesicular carriers, SLN, and NLC. Poor drug loading for hydrophilic compounds.

2. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers

Solid lipid nanoparticles (SLN), as well as nanostructured lipid carriers (NLC), are extensively employed in cutaneous delivery systems. Since their creation in the nineties, lipid nanoparticles (SLN and NLC) are well known by the research and pharma technology community. Easily available raw materials, relatively simple production methods, biocompatibility, and non-toxicity as their advantages over other colloidal carriers can be mentioned as the main reasons for this [18].
Therefore, these two types of lipid nanoparticles—SLN and NLC, are classified according to their structure. First, SLN are developed with a composition of solid lipids only. Then, to upgrade to SLN, NLC were created by representing a mixture of solid and liquid lipids, with a predominant solid lipid [32].
The dermal use of SLN and NLC is proving to be one of the most convenient for therapeutic and cosmetic purposes, despite various applications to date. Lipid nanoparticles are aqueous dispersions with low viscosity for successful direct application to the skin, which implies their incorporation in semisolid systems based on SLN or NLC. One of the first successful administered and marketed products based on NLC is Cutanova® from Dr. Rimpler GmbH (Wedemark, Germany) and Nanobase® from Yamanouchi (Tokyo, Japan) [33].

2.1. Solid Lipid Nanoparticles

Generally speaking, SLN are nanometric colloidal carriers composed of a solid lipid core with an incorporated active pharmaceutical ingredient(s) (API) and a surfactant-stabilized shell [34][35][36]. SLN are composed in the 1990s and have unique properties such as biodegradability, impressive toxicity profile, protection of the API against degradation, high load capacity, sterilization ability, and scalability [37][38]. The interaction between the lipid core of SLN and the waxy lipids in SC leads to a significant permeation enhancement of the encapsulated drug into the skin, which determines their successful cutaneous application [39].

2.2. Nanostructured Lipid Carriers

The second generation of lipid nanoparticles—NLC, are composed of a mixture of solid lipids and liquid lipids in the nanocore, usually in a ratio of 7:3 to 9:1 [40]. This leads to a more significant disorder of the core of the lipid matrix, and accordingly decreases the melting point to stop the recrystallization of solid lipids [41]. NLC are considered to be an improved variety of SLN, holding the same unique properties, but with an optimized core composition, resulting in a higher drug loading capacity, better stability, and ability to act at lower temperatures. Of note is the fact that NLCs are still solid at body temperature [42].

2.3. Preparation of SLN and NLC

The literature describes a significant number of production methods and many different combinations of lipids to obtain SLN and NLC. Nevertheless, the most common technique used today is high-pressure homogenization (HPH). The procedure is divided into two stages:
  • Hot homogenization—the lipids are heated above their melting point;
  • Cold homogenization—takes place at low temperatures and is suitable for hydrophilic and temperature-sensitive API [43][44].
Other commonly used techniques are: Sonication/ultra-sonication [45][46], membrane contactor technique [47], phase inversion [48], solvent injection [49], emulsification [50], the microemulsion method [51], etc.

2.4. SLN and NLC in the Treatment of Skin Disorders

In dermal applications, SLN and NLC create a thin hydrophobic monolayer during skin contact, which has a pointed occlusive effect that settles the API penetration and prevents water loss from the skin [52].
When applied topically, the lipid nanoparticles interact with the sebum and specific skin lipids, provoking a change in the natural arrangement of corneocytes. As a result of this interaction, the encapsulated molecules are released, and their penetration into the lower layers of the epidermis and dermis is potentiated, depending on their lipophilicity, of course [53].

2.4.1. Antioxidant Effect

Okonogi and Riangjanapatee formulated NLC loaded with lycopene through a hot HPH. It has been found that the NLC with the highest concentration of lycopene had the slowest release rate and better antioxidant activity [54].
In another study, Shrotriya et al. reported the development of SLN loaded with resveratrol (entrapment efficiency of 86–89%) to treat irritant contact dermatitis (chronic skin disorder with eczematous injuries). The composition was realized by incorporation into a Carbopol gel and showed increased antioxidant activity compared with a conventional resveratrol gel [55].
Furthermore, Montenegro et al. designed a novel Idebenone (IDE)-loaded NLC containing tocopheryl acetate (VitE) as a liquid component to obtain a synergic effect between IDE and VitE [56].

2.4.2. Anti-Inflammatory Effect

Pivetta et al. formulated NLC with thymol for the local treatment of inflammatory skin diseases (entrapment efficiency of 89%). The NLC were incorporated into a gel and showed anti-inflammatory activity and healing of induced psoriasis in mice [57].
Gad et al. reported the encapsulation of chamomile oil in SLN for the local treatment of wounds. The composition contained stearic acid and chamomile oil and was prepared by the method of hot homogenization. Wound reduction was shown in the topical application in rats [58].

2.4.3. Antifungal Effect

Butani et al. developed a stable SLN system, containing amphotericin B with an enhancing antifungal activity (entrapment efficiency of 94%). The formulation indicated higher drug permeation and drug accumulation in the skin than the conventional gel in rats. A solvent diffusion technique was used for the preparation of the SLN [59].
NLC, for the treatment of candidiasis with Mediterranean essential oils and clotrimazole, were designed by Carbone et al. As a result, they are obtained as a stable NLC, without an initial burst effect and with prolonged release of clotrimazole, as well as an enhanced antifungal activity [60].

2.4.4. Anti-Acne Effect

Tretinoin-loaded NLC with anti-aging and anti-acne activities were reported by Ghate et al. The hot melt probe sonication and hot melt microemulsion methods were used to prepare the NLC. The tretinoin-loaded NLC in Carbopol gels showed no irritation or erythema after the application in rats [61].
Malik and Kaur developed the azelaic acid-loaded NLC, prepared by the melt emulsification and ultra-sonication method (entrapment efficiencies greater than 80%). NLC were incorporated into aloe-vera-based Carbopol hydrogels and demonstrated a deeper skin penetration than the commercial product (Aziderm 10%). Furthermore, the in vivo experiment in mice showed a higher effect of NLC incorporated into a gel than the plain drug suspended in the gel [62].
Table 2 presents the practical implementation of SLN and NLC in dermal drug delivery systems.
Table 2. Application of SLN and NLC DDS for the treatment of skin disorders.
LNP Type API/Drug Application Reference
SLN Doxorubicin Doxorubicin-loaded SLN for the treatment of skin cancer. [63]
SLN Adapalene Adapalene-loaded SLN in the gel for anti-acne treatment. [64]
SLN Triamcinolone acetonide Triamcinolone acetonide-loaded SLN for the topical treatment of psoriasis. [65]
SLN Resveratrol, vitamin E, and
epigallocatechin gallate
SLN containing resveratrol, vitamin E, and epigallocatechin gallate for antioxidant benefits. [66]
SLN Silybin Silybin-loaded SLN enriched gel for irritant contact dermatitis. [67]
SLN Fluconazole Fluconazole-loaded SLN topical gel for the treatment of pityriasis versicolor. [68]
SLN Tazarotene Tazarotene-loaded SLN for the treatment of psoriasis. [69]
SLN Miconazole nitrate Miconazole nitrate-loaded SLN for antifungal activity. [70]
SLN Adapalene Adapalene-loaded SLN for anti-acne therapy. [71]
SLN Isotretinoin and
α-tocopherol
SLN loaded with retinoic acid and lauric acid for the topical treatment of acne vulgaris. [72]
NLC Spironolactone Spironolactone-loaded NLC-based gel for the effective treatment of acne vulgaris. [73]
NLC Clobetasol propionate NLC-based topical gel of clobetasol propionate for the treatment of eczema. [74]
NLC Tacrolimus and tumor
necrosis factor α siRNA
NLC co-delivering tacrolimus and tumor necrosis factor α siRNA for the treatment of psoriasis. [75]
NLC Itraconazole Topical NLC containing itraconazole for the treatment of fungal infections. [76]
NLC Apremilast NLC for topical delivery of apremilast for the treatment of psoriasis. [77]
NLC Dithranol Dithranol-loaded NLC-based gel for the treatment of psoriasis. [78]
NLC Voriconazole Voriconazole-loaded NLC for antifungal applications. [79]
NLC Mometasone furoate NLC-based hydrogel of mometasone furoate for the treatment of psoriasis. [80]
NLC Antimicrobial peptide
nisin Z
Antimicrobial peptide nisin Z with conventional antibiotic-loaded NLC to enhance antimicrobial activity. [81]
NLC Adapalene and vitamin C Adapalene- and vitamin C-loaded NLC for acne treatment. [82]

3. Conclusions

Skin disorders represent a progressively emerging clinical public health problem. Treatment strategies based on conventional formulations are non-specific and can lead to considerable systemic toxicity. The progressive approach of the use of lipid nano-formulations as skin drug delivery systems can provide an incomparable prospect for the application of highly competent and safe treatments with the improved benefit-risk ratio.
The use of lipid nanoparticlulate DDS is favored recently due to the GRAS status of the excipients. Lipid nanocarriers can effectively protect the API from degradation on the skin’s surface, increase their concentration gradient in the upper skin layers, and enable gradual release. Lipid nanoparticles for topical application could be formulated with the high content of lipid matrix or dispersed in different foundations.
Lipid nanosystems provide a promising, flexible platform for the safe, effective, and biocompatible topical delivery of the API, as they do not cause cytotoxicity or morphological changes in the skin layers. The interest shown by pharmaceutical scientists, in the development of lipid nanoparticle delivery systems, may offer a future that provides sufficiently efficient lipid nanoparticle products for needy users.

References

  1. Tabassum, N.; Hamdani, M. Plants used to treat skin diseases. Pharmacogn. Rev. 2014, 8, 52–60.
  2. Ferreira, K.C.B.; Valle, A.B.C.d.S.; Paes, C.Q.; Tavares, G.D.; Pittella, F. Nanostructured Lipid Carriers for the Formulation of Topical Anti-Inflammatory Nanomedicines Based on Natural Substances. Pharmaceutics 2021, 13, 1454.
  3. Wolff, K.; Johnson, R.A.; Saavedra, A.P.; Roh, E.K. Fitzpatrick’s Color Atlas and Synopsis of Clinical Dermatology, 8th ed.; McGraw-Hill Education: New York, NY, USA, 2017; pp. 693–759. Available online: https://accessmedicine.mhmedical.com/content.aspx?bookid=2043&sectionid=154893575 (accessed on 19 October 2021).
  4. Urban, K.; Chu, S.; Scheufele, C.; Giesey, R.L.; Mehrmal, S.; Uppal, P.; Delost, G.R. The global, regional, and national burden of fungal skin diseases in 195 countries and territories: A cross-sectional analysis from the Global Burden of Disease Study 2017. JAAD Int. 2020, 2, 22–27.
  5. Craddock, L.N.; Schieke, S.M. Superficial fungal infection. In Fitzpatrick’s Dermatology, 9th ed.; Kang, S., Amagai, M., Bruckner, A.L., Enk, A.H., Margolis, D.J., McMichael, A.J., Orringer, J.S., Eds.; McGraw-Hill: New York, NY, USA, 2019; Chapter 160; pp. 1–48. Available online: https://accessmedicine.mhmedical.com/content.aspx?sectionid=210432218&bookid=2570&Resultclick=2 (accessed on 19 October 2021).
  6. Silverberg, B. A Structured Approach to Skin and Soft Tissue Infections (SSTIs) in an Ambulatory Setting. Clin. Pract. 2021, 11, 65–74.
  7. Pearson, D.R.; Margolis, D.J. Cellulitis and erysipelas. In Fitzpatrick’s Dermatology, 9th ed.; Kang, S., Amagai, M., Bruckner, A.L., Enk, A.H., Margolis, D.J., McMichael, A.J., Orringer, J.S., Eds.; McGraw-Hill: New York, NY, USA, 2019; Chapter 151; pp. 1–19. Available online: https://accessmedicine.mhmedical.com/content.aspx?bookid=2570&sectionid=210413306 (accessed on 19 October 2021).
  8. Sartelli, M.; Guirao, X.; Hardcastle, T.; Kluger, Y.; Boermeester, M.A.; Rasa, K.; Ansaloni, L.; Coccolini, F.; Montravers, P.; Abu-Zidan, F.M.; et al. 2018 WSES/SIS-E consensus conference: Recommendations for the management of skin and soft tissue infections. World J. Emerg. Surg. 2018, 13, 58.
  9. Falcone, M.; Concia, E.; Giusti, M.; Mazzone, A.; Santini, C.; Stefani, S.; Violi, F. Acute bacterial skin and skin structure infections in internal medicine wards: Old and new drugs. Inter. Emerg. Med. 2016, 11, 637–648.
  10. Khadr, L.; Harfouche, M.; Omori, R.; Schwarzer, G.; Chemaitelly, H.; Abu-Raddad, L.J. The Epidemiology of Herpes Simplex Virus Type 1 in Asia: Systematic Review, Meta-analyses, and Meta-regressions. Clin. Infect. Dis. 2019, 68, 757–772.
  11. Poole, C.L.; Kimberlin, D.W. Antiviral Approaches for the Treatment of Herpes Simplex Virus Infections in Newborn Infants. Annu. Rev. Virol. 2018, 5, 407–425.
  12. Mustafa, M.; Illzam, E.M.; Muniandy, R.K.; Sharifah, A.M.; Nang, M.K.; Ramesh, B. Herpes simplex virus infections, Pathophysiology and Management. IOSR J. Dent. Med. Sci. 2016, 15, 85–91.
  13. Schwingen, J.; Kaplan, M.; Kurschus, F.C. Review-Current Concepts in Inflammatory Skin Diseases Evolved by Transcriptome Analysis: In-Depth Analysis of Atopic Dermatitis and Psoriasis. Int. J. Mol. Sci. 2020, 21, 699.
  14. Ho, A.W.; Kupper, T.S. T cells and the skin: From protective immunity to inflammatory skin disorders. Nat. Rev. Immunol. 2019, 19, 490–502.
  15. Golan, Y. Current Treatment Options for Acute Skin and Skin-structure Infections. Clin. Infect. Dis. 2019, 68, 206–212.
  16. Aktas, E.; Esin, M.N. Skin disease symptoms and related risk factors among young workers in high-risk jobs. Contact Dermat. 2016, 75, 96–105.
  17. Filipczak, N.; Yalamarty, S.S.K.; Li, X.; Khan, M.M.; Parveen, F.; Torchilin, V. Lipid-Based Drug Delivery Systems in Regenerative Medicine. Materials 2021, 14, 5371.
  18. LePree, J.M. Lipid based delivery—Are lipid-based drug delivery systems in your formulation toolbox? Drug Dev. Deliv. 2017, 17, 20–25. Available online: https://drug-dev.com/lipid-based-delivery-are-lipid-based-drug-delivery-systems-in-your-formulation-toolbox/ (accessed on 19 October 2021).
  19. Carita, A.C.; Eloy, J.O.; Chorilli, M.; Lee, R.J.; Leonardi, G.R. Recent Advances and Perspectives in Liposomes for Cutaneous Drug Delivery. Curr. Med. Chem. 2018, 25, 606–635.
  20. Garg, V.; Singh, H.; Bimbrawh, S.; Singh, S.K.; Gulati, M.; Vaidya, Y.; Kaur, P. Ethosomes and Transfersomes: Principles, Perspectives and Practices. Curr. Drug. Deliv. 2017, 14, 613–633.
  21. Opatha, S.A.T.; Titapiwatanakun, V.; Chutoprapat, R. Transfersomes: A Promising Nanoencapsulation Technique for Transdermal Drug Delivery. Pharmaceutics 2020, 12, 855.
  22. Chaurasiya, P.; Ganju, E.; Upmanyu, N.; Ray, S.K.; Jain, P. Transfersomes: A novel technique for transdermal drug delivery. J. Drug Deliv. Ther. 2019, 9, 279–285.
  23. Nainwal, N.; Jawla, S.; Singh, R.; Saharan, V.A. Transdermal applications of ethosomes—A detailed review. J. Liposome Res. 2019, 29, 103–113.
  24. Natsheh, H.; Vettorato, E.; Touitou, E. Ethosomes for Dermal Administration of Natural Active Molecules. Curr. Pharm. Des. 2019, 25, 2338–2348.
  25. Paliwal, R.; Paliwal, S.R.; Kenwat, R.; Kurmi, B.D.; Sahu, M.K. Solid lipid nanoparticles: A review on recent perspectives and patents. Expert Opin. Ther. Pat. 2020, 30, 179–194.
  26. Liu, M.; Wen, J.; Sharma, M. Solid Lipid Nanoparticles for Topical Drug Delivery: Mechanisms, Dosage Form Perspectives, and Translational Status. Curr. Pharm. Des. 2020, 26, 3203–3217.
  27. Geng, Q.; Zhao, Y.; Wang, L.; Xu, L.; Chen, X.; Han, J. Development and Evaluation of Astaxanthin as Nanostructure Lipid Carriers in Topical Delivery. AAPS Pharm. Sci. Tech. 2020, 21, 318.
  28. Araujo, V.H.S.; Delello Di Filippo, L.; Duarte, J.L.; Spósito, L.; de Camargo, B.A.F.; da Silva, P.B.; Chorilli, M. Exploiting solid lipid nanoparticles and nanostructured lipid carriers for drug delivery against cutaneous fungal infections. Crit. Rev. Microbiol. 2020, 47, 79–90.
  29. Jain, A.; Pooladanda, V.; Bulbake, U.; Doppalapudi, S.; Rafeeqi, T.A.; Godugu, C.; Khan, W. Liposphere mediated topical delivery of thymoquinone in the treatment of psoriasis. Nanomedicine 2017, 13, 2251–2262.
  30. Esposito, E.; Sguizzato, M.; Bories, C.; Nastruzzi, C.; Cortesi, R. Production and Characterization of a Clotrimazole Liposphere Gel for Candidiasis Treatment. Polymers 2018, 10, 160.
  31. Waghule, T.; Gorantla, S.; Rapalli, V.K.; Shah, P.; Dubey, S.K.; Saha, R.N.; Singhvi, G. Emerging Trends in Topical Delivery of Curcumin Through Lipid Nanocarriers: Effectiveness in Skin Disorders. AAPS Pharm. Sci. Tech. 2020, 21, 284.
  32. Silva, A.C.; Amaral, M.H.; Lobo, J.M.; Lopes, C.M. Lipid nanoparticles for the delivery of biopharmaceuticals. Curr. Pharm. Biotechnol. 2015, 16, 291–302.
  33. Chella, N.; Shastri, N.R. Lipid Carriers: Role and Applications in Nano Drug Delivery. In Particulate Technology for Delivery of Therapeutics; Jana, S., Jana, S., Eds.; Springer: Singapore, 2017; pp. 253–289.
  34. Trombino, S.; Mellace, S.; Cassano, R. Solid lipid nanoparticles for antifungal drugs delivery for topical applications. Ther. Deliv. 2016, 7, 639–647.
  35. Xing, H.; Wang, H.; Wu, B.; Zhang, X. Lipid nanoparticles for the delivery of active natural medicines. Curr. Pharm. Des. 2017, 23, 6705–6713.
  36. De Souza, M.L.; Dos Santos, W.M.; de Sousa, A.L.M.D.; de Albuquerque Wanderley Sales, V.; Nóbrega, F.P.; de Oliveira, M.V.G.; Rolim-Neto, P.J. Lipid Nanoparticles as a Skin Wound Healing Drug Delivery System: Discoveries and Advances. Curr. Pharm. Des. 2020, 26, 4536–4550.
  37. Naseri, N.; Valizadeh, H.; Zakeri-Milani, P. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers: Structure, Preparation and Application. Adv. Pharm. Bull. 2015, 5, 305–313.
  38. Gratieri, T.; Krawczyk-Santos, A.P.; da Rocha, P.B.; Gelfuso, G.M.; Marreto, R.N.; Taveira, S.F. SLN- and NLC-Encapsulating Antifungal Agents: Skin Drug Delivery and their Unexplored Potential for Treating Onychomycosis. Curr. Pharm. Des. 2017, 23, 6684–6695.
  39. Pham, D.T.T.; Tran, P.H.L.; Tran, T.T.D. Development of solid dispersion lipid nanoparticles for improving skin delivery. Saudi Pharm. J. 2019, 27, 1019–1024.
  40. Waghule, T.; Rapalli, V.K.; Gorantla, S.; Saha, R.N.; Dubey, S.K.; Puri, A.; Singhvi, G. Nanostructured Lipid Carriers as Potential Drug Delivery Systems for Skin Disorders. Curr. Pharm. Des. 2020, 26, 4569–4579.
  41. Czajkowska-Kośnik, A.; Szekalska, M.; Winnicka, K. Nanostructured lipid carriers: A potential use for skin drug delivery systems. Pharmacol. Rep. 2019, 71, 156–166.
  42. Gordillo-Galeano, A.; Mora-Huertas, C.E. Solid lipid nanoparticles and nanostructured lipid carriers: A review emphasizing on particle structure and drug release. Eur. J. Pharm. Biopharm. 2018, 133, 285–308.
  43. Rajpoot, K. Solid Lipid Nanoparticles: A Promising Nanomaterial in Drug Delivery. Curr. Pharm. Des. 2019, 25, 3943–3959.
  44. Ganesan, P.; Narayanasamy, D. Lipid nanoparticles: Different preparation techniques, characterization, hurdles, and strategies for the production of solid lipid nanoparticles and nanostructured lipid carriers for oral drug delivery. Sustain. Chem. Pharm. 2017, 6, 37–56.
  45. Trivino, A.; Gumireddy, A.; Chauhan, H. Drug-Lipid-Surfactant Miscibility for the Development of Solid Lipid Nanoparticles. AAPS Pharm. Sci. Tech. 2019, 20, 46.
  46. Soleimanian, Y.; Goli, S.A.H.; Varshosaz, J.; Sahafi, S.M. Formulation and characterization of novel nanostructured lipid carriers made from beeswax, propolis wax and pomegranate seed oil. Food Chem. 2018, 244, 83–92.
  47. Sebaaly, C.; Charcosset, C.; Stainmesse, S.; Fessi, H.; Greige-Gerges, H. Clove essential oil-in-cyclodextrin-in-liposomes in the aqueous and lyophilized states: From laboratory to large scale using a membrane contactor. Carbohydr. Polym. 2016, 138, 75–85.
  48. Shah, M.K.; Khatri, P.; Vora, N.; Patel, N.K.; Jain, S.; Lin, S. Lipid nanocarriers: Preparation, characterization and absorption mechanism and applications to improve oral bioavailability of poorly water-soluble drugs. In Biomedical Applications of Nanoparticles; Grumezescu, A.M., Ed.; William Andrew Publishing: Norwich, NY, USA, 2019; pp. 117–147.
  49. Duong, V.-A.; Nguyen, T.-T.-L.; Maeng, H.-J. Preparation of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers for Drug Delivery and the Effects of Preparation Parameters of Solvent Injection Method. Molecules 2020, 25, 4781.
  50. Chaudhary, S.A.; Patel, D.M.; Patel, J.K.; Patel, D.H. Solvent Emulsification Evaporation and Solvent Emulsification Diffusion Techniques for Nanoparticles. In Emerging Technologies for Nanoparticle Manufacturing; Patel, J.K., Pathak, Y.V., Eds.; Springer: Cham, Switzerland, 2021; pp. 287–300.
  51. Singh, D.; Tiwary, A.K.; Bedi, N. Self-microemulsifying Drug Delivery System for Problematic Molecules: An Update. Recent Pat. Nanotech. 2019, 13, 92–113.
  52. Desai, P.; Patlolla, R.R.; Singh, M. Interaction of nanoparticles and cell-penetrating peptides with skin for transdermal drug delivery. Mol. Membr. Biol. 2010, 27, 247–259.
  53. Jensen, L.B.; Petersson, K.; Nielsen, H.M. In vitro penetration properties of solid lipid nanoparticles in intact and barrier-impaired skin. Eur. J. Pharm. Biopharm. 2011, 79, 68–75.
  54. Okonogi, S.; Riangjanapatee, P. Physicochemical characterization of lycopene-loaded nanostructured lipid carrier formula-tions for topical administration. Int. J. Pharm. 2015, 478, 726–735.
  55. Shrotriya, S.N.; Ranpise, N.S.; Vidhate, B.V. Skin targeting of resveratrol utilizing solid lipid nanoparticle-engrossed gel for chemically induced irritant contact dermatitis. Drug Deliv. Transl. Res. 2017, 7, 37–52.
  56. Montenegro, L.; Messina, C.M.; Manuguerra, S.; Santagati, L.M.; Pasquinucci, L.; Turnaturi, R.; Parenti, C.; Arena, R.; Santulli, A. In Vitro Antioxidant Activity and In Vivo Topical Efficacy of Lipid Nanoparticles Co-Loading Idebenone and Tocopheryl Acetate. Appl. Sci. 2019, 9, 845.
  57. Pivetta, T.P.; Simões, S.; Araújo, M.M.; Carvalho, T.; Arruda, C.; Marcato, P.D. Development of nanoparticles from natural lipids for topical delivery of thymol: Investigation of its anti-inflammatory properties. Colloids Surf. B Biointerfaces 2018, 164, 281–290.
  58. Gad, H.A.; El-Rahman, F.A.A.; Hamdy, G.M. Chamomile oil loaded solid lipid nanoparticles: A naturally formulated remedy to enhance the wound healing. J. Drug Deliv. Sci. Technol. 2019, 50, 329–338.
  59. Butani, D.; Yewale, C.; Misra, A. Topical Amphotericin B solid lipid nanoparticles: Design and development. Colloids Surf. B Biointerfaces 2016, 139, 17–24.
  60. Carbone, C.; do Céu Teixeira, M.; do Céu Sousa, M.; Martins-Gomes, C.; Silva, A.M.; Souto, E.M.B.; Musumeci, T. Clotrima- zole-Loaded Mediterranean Essential Oils NLC: A Synergic Treatment of Candida Skin Infections. Pharmaceutics 2019, 11, 231.
  61. Ghate, V.M.; Lewis, S.A.; Prabhu, P.; Dubey, A.; Patel, N. Nanostructured lipid carriers for the topical delivery of tretinoin. Eur. J. Pharm. Biopharm. 2016, 108, 253–261.
  62. Malik, D.S.; Kaur, G. Exploring therapeutic potential of azelaic acid loaded NLCs for the treatment of acne vulgaris. J. Drug Deliv. Sci. Technol. 2020, 55, 101418.
  63. Tupal, A.; Sabzichi, M.; Ramezani, F.; Kouhsoltani, M.; Hamishehkar, H. Dermal delivery of doxorubicin-loaded solid lipid nanoparticles for the treatment of skin cancer. J. Microencapsul. 2016, 33, 372–380.
  64. Harde, H.; Agrawal, A.K.; Katariya, M.; Kale, D.; Jain, S. Development of a topical adapalene-solid lipid nanoparticle load-ed gel with enhanced efficacy and improved skin tolerability. RSC Adv. 2015, 5, 43917–43929.
  65. Pradhan, M.; Singh, D.; Singh, M.R. Influence of selected variables on fabrication of Triamcinolone acetonide loaded solid lipid nanoparticles for topical treatment of dermal disorders. Artif. Cells Nanomed. Biotechnol. 2016, 44, 392–400.
  66. Wang, W.; Chen, L.; Huang, X.; Shao, A. Preparation and Characterization of Minoxidil Loaded Nanostructured Lipid Car-riers. AAPS Pharm. Sci. Tech. 2017, 18, 509–516.
  67. Shrotriya, S.N.; Vidhate, B.V.; Shukla, M.S. Formulation and development of Silybin loaded solid lipid nanoparticle enriched gel for irritant contact dermatitis. J. Drug Deliv. Sci. Technol. 2017, 41, 164–173.
  68. El-Housiny, S.; Shams Eldeen, M.A.; El-Attar, Y.A.; Salem, H.A.; Attia, D.; Bendas, E.R.; El-Nabarawi, M.A. Flucona-zole-loaded solid lipid nanoparticles topical gel for treatment of pityriasis versicolor: Formulation and clinical study. Drug Deliv. 2018, 25, 78–90.
  69. Aland, R.; Ganesan, M.; Rajeswara, R.P. Development and optimization of tazarotene loaded solid lipid nanoparticles for topical delivery. Asian J. Pharm. Clin. Res. 2019, 12, 63–77.
  70. Al-Maghrabi, P.M.; Khafagy, E.-S.; Ghorab, M.M.; Gad, S. Influence of formulation variables on miconazole nitrate-loaded lipid based nanocarrier for topical delivery. Colloids Surf. B Biointerfaces 2020, 193, 111046.
  71. Bhalekar, M.; Upadhaya, P.; Madgulkar, A. Formulation and evaluation of Adapalene-loaded nanoparticulates for epidermal localization. Drug Deliv. Transl. Res. 2015, 5, 585–595.
  72. Gupta, S.; Wairkar, S.; Bhatt, L.K. Isotretinoin and α-tocopherol acetate-loaded solid lipid nanoparticle topical gel for the treatment of acne. J. Microencapsul. 2020, 37, 1–9.
  73. Kelidari, H.R.; Saeedi, M.; Hajheydari, Z.; Akbari, J.; Morteza-Semnani, K.; Akhtari, J.; Valizadeh, H.; Asare-Addo, K.; Nokhodchi, A. Spironolactone loaded nanostructured lipid carrier gel for effective treatment of mild and moderate ac-ne vulgaris: A randomized, double-blind, prospective trial. Colloids Surf. B Biointerfaces 2016, 146, 47–53.
  74. Nagaich, U.; Gulati, N. Nanostructured lipid carriers (NLC) based controlled release topical gel of clobetasol propionate: Design and in vivo characterization. Drug Deliv. Transl. Res. 2016, 6, 289–298.
  75. Viegas, J.S.R.; Praça, F.G.; Caron, A.L.; Suzuki, I.; Silvestrini, A.V.P.; Medina, W.S.G.; Del Ciampo, J.O.; Kravicz, M.; Bentley, M.V.L.B. Nanostructured lipid carrier co-delivering tacrolimus and TNF-α siRNA as an innovate approach to psoriasis. Drug Deliv. Transl. Res. 2020, 10, 646–660.
  76. Passos, J.S.; De Martino, L.C.; Dartora, V.F.C.; De Araujo, G.L.B.; Ishida, K.; Lopes, L.B. Development, skin targeting and antifungal efficacy of topical lipid nanoparticles containing itraconazole. Eur. J. Pharm. Sci. 2020, 149, 105296.
  77. Madan, J.R.; Khobaragade, S.; Dua, K.; Awasthi, R. Formulation, optimization, and in vitro evaluation of nanostructured lipid carriers for topical delivery of Apremilast. Dermatol. Ther. 2020, 33, e13370.
  78. Sathe, P.; Saka, R.; Kommineni, N.; Raza, K.; Khan, W. Dithranol-loaded nanostructured lipid carrier-based gel ameliorate psoriasis in imiquimod-induced mice psoriatic plaque model. Drug Dev. Ind. Pharm. 2019, 45, 826–838.
  79. Waghule, T.; Rapalli, V.K.; Singhvi, G.; Manchanda, P.; Hans, N.; Dubey, S.K.; Hasnain, M.S.; Nayak, A.K. Voriconazole loaded nanostructured lipid carriers based topical delivery system: QbD based designing, characterization, in-vitro and ex-vivo evaluation. J. Drug Deliv. Sci. Technol. 2019, 52, 303–315.
  80. Kaur, N.; Sharma, K.; Bedi, N. Topical Nanostructured Lipid Carrier Based Hydrogel of Mometasone Furoate for the Treatment of Psoriasis. Pharm. Nanotechnol. 2018, 6, 133–143.
  81. Lewies, A.; Wentzel, J.F.; Jordaan, A.; Bezuidenhout, C.; Du Plessis, L.H. Interactions of the antimicrobial peptide nisin Z with conventional antibiotics and the use of nanostructured lipid carriers to enhance antimicrobial activity. Int. J. Pharm. 2017, 526, 244–253.
  82. Jain, A.; Garg, N.K.; Jain, A.; Kesharwani, P.; Jain, A.K.; Nirbhavane, P.; Tyagi, R.K. A synergistic approach of adapalene-loaded nanostructured lipid carriers, and vitamin C co-administration for treating acne. Drug Dev. Ind. Pharm. 2016, 42, 897–905.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 636
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 16 Nov 2021
1000/1000
Video Production Service