Exploring Nanocarriers as Treatment Modalities for Skin Cancer
Abstract
:1. Introduction
2. Molecular Pathways
3. Nanoformulations-Based Delivery of Various Therapeutic Agents for Skin Cancer Rationale for Using Nanocarriers
4. Vesicular Nanoformulations for Skin Cancer
4.1. Liposomes
4.2. Niosomes
4.3. Transferosomes
4.4. Ethosomes
4.5. Transethosomes
4.6. Solid Lipid Nanoparticles (SLNs)
4.7. Nanostructured Lipid Carriers (NLCs)
5. Polymeric Micelles and Nanoparticles (NPs)
5.1. Polymeric Micelles
5.2. Polymeric NPs
5.3. Dendrimers
6. Nanofibers
7. Metallic NPs
8. Nanogels
9. Nanoemulsions (NEs)
10. Conclusions and Future Prospects
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Hausman, D.M. What Is Cancer? Perspect. Biol. Med. Perspect. Biol. Med. 2019, 62, 778–784. [Google Scholar] [CrossRef] [PubMed]
- Hajdu, S.I. A note from history: Landmarks in history of cancer, part 1. Cancer 2011, 117, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
- Kalhori, M.R.; Arefian, E.; Atanaki, F.F.; Kavousi, K.; Soleimani, M. miR-548x and miR-4698 controlled cell proliferation by affecting the PI3K/AKT signaling pathway in Glioblastoma cell lines. Sci. Rep. 2020, 10, 1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Falto-Aizpurua, L.A.; Griffith, R.D.; Nouri, K. Josef Jadassohn. JAMA Dermatol. 2015, 151, 41. [Google Scholar] [CrossRef]
- Kentsis, A. Why do young people get cancer? Pediatr. Blood Cancer 2020, 67, e28335. [Google Scholar] [CrossRef] [PubMed]
- Hill, B.T. Etiology of Cancer. In Clinical Ophthalmic Oncology: Basic Principles; Singh, A.D., Damato, B.E., Eds.; Springer International Publishing: Cham, Switzerland, 2019; pp. 11–17. [Google Scholar] [CrossRef]
- Yokota, J. Tumor progression and metastasis. Carcinogenesis 2000, 21, 497–503. [Google Scholar] [CrossRef] [PubMed]
- Trager, M.H.; Geskin, L.J.; Samie, F.H.; Liu, L. Biomarkers in melanoma and non-melanoma skin cancer prevention and risk stratification. Exp. Dermatol. 2022, 31, 4–12. [Google Scholar] [CrossRef]
- Guy, G.P., Jr.; Thomas, C.C.; Thompson, T.; Watson, M.; Massetti, G.M.; Richardson, L.C. Vital signs: Melanoma incidence and mortality trends and projections—United States, 1982–2030. Morb. Mortal. Wkly. Rep. 2015, 64, 591–596. [Google Scholar]
- Sabir, F.; Barani, M.; Rahdar, A.; Bilal, A.; Zafar, M.; Bungau, S.; Kyzas, G. How to Face Skin Cancer with Nanomaterials: A Review. Biointerface Res. Appl. Chem. 2020, 11, 11931–11955. [Google Scholar]
- Lazar, M.; Davis, A.; Diamandis, C. Skin Cancer Prevention: Which Strategy Is the Most Promising? Zenodo Publishing: Geneva, Switzerland, 2021. [Google Scholar] [CrossRef]
- Muntyanu, A.; Ghazawi, M.; Nedjar, H.; Rahme, E.; Alakel, A.; Zubarev, A.; Netchiporouk, E.; Litvinov, V. Non-Melanoma Skin Cancer Distribution in the Russian Federation. Dermatology 2020, 237, 1007–1015. [Google Scholar] [CrossRef]
- Parker, E.R. The influence of climate change on skin cancer incidence—A review of the evidence. Int. J. Women’s Dermatol. 2021, 7, 17–27. [Google Scholar] [CrossRef]
- Labani, S.; Asthana, S.; Rathore, K.; Sardana, K. Incidence of melanoma and nonmelanoma skin cancers in Indian and the global regions. J. Cancer Res. Ther. 2021, 17, 906–911. [Google Scholar] [CrossRef]
- Davis, D.S.; Robinson, C.; Callender, V.D. Skin cancer in women of color: Epidemiology, pathogenesis and clinical manifestations. Int. J. Women’s Dermatol. 2021, 7, 127–134. [Google Scholar] [CrossRef]
- Carr, S.; Smith, C.; Wernberg, J. Epidemiology and Risk Factors of Melanoma. Surg. Clin. N. Am. 2020, 100, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Urban, K.; Mehrmal, S.; Uppal, P.; Giesey, R.L.; Delost, G.R. The global burden of skin cancer: A longitudinal analysis from the Global Burden of Disease Study, 1990–2017. JAAD Int. 2021, 2, 98–108. [Google Scholar] [CrossRef]
- Niino, M.; Matsuda, T. Age-specific skin cancer incidence rate in the world. Jpn. J. Clin. Oncol. 2021, 51, 848–849. [Google Scholar] [CrossRef] [PubMed]
- Griffin, L.L.; Ali, F.R.; Lear, J.T. Non-melanoma skin cancer. Clin. Med. 2016, 16, 62–65. [Google Scholar] [CrossRef]
- Linares, M.A.; Zakaria, A.; Nizran, P. Skin Cancer. Prim. Care Clin. Off. Pract. 2015, 42, 645–659. [Google Scholar] [CrossRef]
- De Gruijl, F.R.; van Kranen, H.J.; Mullenders, L.H. UV-induced DNA damage, repair, mutations and oncogenic pathways in skin cancer. J. Photochem. Photobiol. B Biol. 2001, 63, 19–27. [Google Scholar] [CrossRef] [PubMed]
- Chamcheu, J.C.; Roy, T.; Uddin, M.B.; Banang-Mbeumi, S.; Chamcheu, R.-C.N.; Walker, A.L.; Liu, Y.-Y.; Huang, S. Role and Therapeutic Targeting of the PI3K/Akt/mTOR Signaling Pathway in Skin Cancer: A Review of Current Status and Future Trends on Natural and Synthetic Agents Therapy. Cells 2019, 8, 803. [Google Scholar] [CrossRef] [Green Version]
- Swann, G. Editorial. J. Vis. Commun. Med. 2010, 33, 148–149. [Google Scholar] [CrossRef]
- Busco, G.; Robert, E.; Chettouh-Hammas, N.; Pouvesle, J.-M.; Grillon, C. The emerging potential of cold atmospheric plasma in skin biology. Free. Radic. Biol. Med. 2020, 161, 290–304. [Google Scholar] [CrossRef]
- Gour, V.; Agrawal, P.; Pandey, V.; Kanwar, I.L.; Haider, T.; Tiwari, R.; Soni, V. Chapter 10—Nanoparticles and skin cancer. In Nano Drug Delivery Strategies for the Treatment of Cancers; Yadav, A.K., Gupta, U., Sharma, R., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 245–273. [Google Scholar] [CrossRef]
- Ohbayashi, N.; Fukuda, M. Recent advances in understanding the molecular basis of melanogenesis in melanocytes. F1000Research 2020, 9, 608. [Google Scholar] [CrossRef] [PubMed]
- Wysong, A.; Higgins, S.; Blalock, T.W.; Ricci, D.; Nichols, R.; Smith, F.L.; Kossintseva, I. Defining skin cancer local recurrence. J. Am. Acad. Dermatol. 2019, 81, 581–599. [Google Scholar] [CrossRef] [PubMed]
- Cives, M.; Mannavola, F.; Lospalluti, L.; Sergi, M.C.; Cazzato, G.; Filoni, E.; Cavallo, F.; Giudice, G.; Stucci, L.S.; Porta, C.; et al. Non-Melanoma Skin Cancers: Biological and Clinical Features. Int. J. Mol. Sci. 2020, 21, 5394. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2016. CA Cancer J. Clin. 2016, 66, 7–30. [Google Scholar] [CrossRef] [Green Version]
- Khan, N.H.; Mir, M.; Qian, L.; Baloch, M.; Khan, M.F.A.; Rehman, A.-U.; Ngowi, E.E.; Wu, D.-D.; Ji, X.-Y. Skin cancer biology and barriers to treatment: Recent applications of polymeric micro/nanostructures. J. Adv. Res. 2022, 36, 223–247. [Google Scholar] [CrossRef] [PubMed]
- Ouhtit, A.; Nakazawa, H.; Yamasaki, H.; Armstrong, B.K.; Kricker, A.; Tan, E.; English, D.R. UV-Radiation-Specific p53 Mutation Frequency in Normal Skin as a Predictor of Risk of Basal Cell Carcinoma. Gynecol. Oncol. 1998, 90, 523–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mota, A.H.; Rijo, P.; Molpeceres, J.; Reis, C.P. Broad overview of engineering of functional nanosystems for skin delivery. Int. J. Pharm. 2017, 532, 710–728. [Google Scholar] [CrossRef]
- Nuccitelli, R.A. Nano-Pulse Stimulation (NPS) in Seborrheic Keratosis Optimization Study. 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04249115 (accessed on 19 March 2023).
- Lang, J.E. Study of Topical SOR007 Ointment for Cutaneous Metastases. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03101358 (accessed on 19 March 2023).
- Adnan, M.; Haider, F.; Naseem, N.; Haider, T. Transethosomes: A Promising Challenge for Topical Delivery Short Title: Transethosomes for Topical Delivery. Drug Res. 2023, 73, 200–212. [Google Scholar] [CrossRef]
- Adnan, M.; Afzal, O.; Altamimi, A.S.A.; Alamri, M.A.; Haider, T.; Haider, F. Development and optimization of transethosomal gel of apigenin for topical delivery: In-vitro, ex-vivo and cell line assessment. Int. J. Pharm. 2023, 631, 122506. [Google Scholar] [CrossRef] [PubMed]
- Zou, Y.; Ge, M.; Wang, X. Targeting PI3K-AKT-mTOR by LY3023414 inhibits human skin squamous cell carcinoma cell growth in vitro and in vivo. Biochem. Biophys. Res. Commun. 2017, 490, 385–392. [Google Scholar] [CrossRef]
- Zhang, X.; Lan, D.; Ning, S.; Ruan, L. Anticancer action of lactucopicrin in SKMEL-5 human skin cancer cells is mediated via apoptosis induction, G2/M cell cycle arrest and downregulation of m=TOR/PI3K/AKT signalling pathway. J. Balk. Union Oncol. 2018, 23, 224–228. [Google Scholar]
- López-Camarillo, C.; Ocampo, E.A.; Casamichana, M.L.; Pérez-Plasencia, C.; Álvarez-Sánchez, E.; Marchat, L.A. Protein Kinases and Transcription Factors Activation in Response to UV-Radiation of Skin: Implications for Carcinogenesis. Int. J. Mol. Sci. 2012, 13, 142–172. [Google Scholar] [CrossRef] [PubMed]
- Brinkhuizen, T.; Weijzen, C.A.H.; Eben, J.; Thissen, M.R.; van Marion, A.M.; Lohman, B.G.; Winnepenninckx, V.J.L.; Nelemans, P.J.; van Steensel, M.A.M. Immunohistochemical Analysis of the Mechanistic Target of Rapamycin and Hypoxia Signalling Pathways in Basal Cell Carcinoma and Trichoepithelioma. PLoS ONE 2014, 9, e106427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haar, E.V.; Lee, S.-I.; Bandhakavi, S.; Griffin, T.J.; Kim, D.-H. Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat. Cell Biol. 2007, 9, 316–323. [Google Scholar] [CrossRef] [PubMed]
- Strozyk, E.; Kulms, D. The Role of AKT/mTOR Pathway in Stress Response to UV-Irradiation: Implication in Skin Carcinogenesis by Regulation of Apoptosis, Autophagy and Senescence. Int. J. Mol. Sci. 2013, 14, 15260–15285. [Google Scholar] [CrossRef] [Green Version]
- Bermudez, Y.; Stratton, S.P.; Curiel-Lewandrowski, C.; Warneke, J.; Hu, C.; Bowden, G.T.; Dickinson, S.E.; Dong, Z.; Bode, A.M.; Saboda, K.; et al. Activation of the PI3K/Akt/mTOR and MAPK Signaling Pathways in Response to Acute Solar-Simulated Light Exposure of Human Skin. Cancer Prev. Res. 2015, 8, 720–728. [Google Scholar] [CrossRef] [Green Version]
- Taylor, J.S. Unraveling the Molecular Pathway from Sunlight to Skin Cancer. Acc. Chem. Res. 1994, 27, 76–82. [Google Scholar] [CrossRef]
- Simanshu, D.K.; Nissley, D.V.; McCormick, F. RAS Proteins and Their Regulators in Human Disease. Cell 2017, 170, 17–33. [Google Scholar] [CrossRef] [Green Version]
- Hwang, S.-Y.; Chae, J.-I.; Kwak, A.-W.; Lee, M.-H.; Shim, J.-H. Alternative Options for Skin Cancer Therapy via Regulation of AKT and Related Signaling Pathways. Int. J. Mol. Sci. 2020, 21, 6869. [Google Scholar] [CrossRef]
- Cazzola, M.; Malcovati, L. Diagnosis and treatment of sideroblastic anemias: From defective heme synthesis to abnormal RNA splicing. Hematology 2015, 2015, 19–25. [Google Scholar] [CrossRef] [Green Version]
- Fargnoli, M.C.; Chimenti, S.; Peris, K.; Keller, G.; Soyer, H.P.; Pozzo, V.D.; Höfler, H. CDKN2a/p16INK4a Mutations and Lack of p19ARF Involvement in Familial Melanoma Kindreds. J. Investig. Dermatol. 1998, 111, 1202–1206. [Google Scholar] [CrossRef]
- Chow, H.Y.; Jubb, A.M.; Koch, J.N.; Jaffer, Z.M.; Stepanova, D.; Campbell, D.A.; Duron, S.G.; O’Farrell, M.; Cai, K.Q.; Klein-Szanto, A.J.; et al. p21-Activated Kinase 1 Is Required for Efficient Tumor Formation and Progression in a Ras-Mediated Skin Cancer Model. Cancer Res. 2012, 72, 5966–5975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, Y.-H.; Zhang, Y.-Q.; Jin, M.-H.; Jin, Y.-H.; Qiu, M.-Y.; Li, W.-L.; He, C.; Yu, L.-Y.; Hyun, J.W.; Lee, J.; et al. Peroxiredoxin I deficiency increases keratinocyte apoptosis in a skin tumor model via the ROS-p38 MAPK pathway. Biochem. Biophys. Res. Commun. 2020, 529, 635–641. [Google Scholar] [CrossRef] [PubMed]
- Wellbrock, C.; Arozarena, I. The Complexity of the ERK/MAP-Kinase Pathway and the Treatment of Melanoma Skin Cancer. Front. Cell Dev. Biol. 2016, 4, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mancebo, S.E.; Wang, S.Q. Skin cancer: Role of ultraviolet radiation in carcinogenesis. Rev. Environ. Health 2014, 29, 265–273. [Google Scholar] [CrossRef]
- Altaylouni, T. Prostate Tumor Heterogeneity and Aggressiveness Revealed via the Androgen Receptor Expression and Activation by Tyrosine Phosphorylation; McGill University: Montreal, QC, Canada, 2018. [Google Scholar]
- Kalal, B.S.; Upadhya, D.; Pai, V.R. Chemotherapy Resistance Mechanisms in Advanced Skin Cancer. Oncol. Rev. 2017, 11, 326. [Google Scholar] [CrossRef] [Green Version]
- Kirby, J.S.; Miller, C.J. Intralesional chemotherapy for nonmelanoma skin cancer: A practical review. J. Am. Acad. Dermatol. 2010, 63, 689–702. [Google Scholar] [CrossRef]
- Busic, V.; Mesic, H.; Begic, A.; Tindholdt, T.; Solberg, U.; Tonseth, K.; Gulbrandsen, P. Breast reconstruction with deepithelialized DIEP flap after recurrent mastitis. Plast. Reconstr. Surg. 2004, 113, 782–784. [Google Scholar] [CrossRef]
- Zhang, L.; Ji, Z.; Zhang, J.; Yang, S. Photodynamic therapy enhances skin cancer chemotherapy effects through autophagy regulation. Photodiagnosis Photodyn. Ther. 2019, 28, 159–165. [Google Scholar] [CrossRef]
- Iii, S.D.A. Treatment of skin cancer using multiple modalities. J. Am. Acad. Dermatol. 1982, 7, 143–171. [Google Scholar]
- Alam, T.; Khan, S.; Gaba, B.; Haider, M.F.; Baboota, S.; Ali, J. Nanocarriers as treatment modalities for hypertension. Drug Deliv. 2017, 24, 358–369. [Google Scholar] [CrossRef] [Green Version]
- Moorthi, C.; Krishnan, K.; Manavalan, R.; Kathiresan, K. Preparation and characterization of curcumin–piperine dual drug loaded nanoparticles. Asian Pac. J. Trop. Biomed. 2012, 2, 841–848. [Google Scholar] [CrossRef] [Green Version]
- Shaikh, J.; Ankola, D.D.; Beniwal, V.; Singh, D.; Kumar, M.N.V.R. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur. J. Pharm. Sci. 2009, 37, 223–230. [Google Scholar] [CrossRef]
- Mulik, R.S.; Mönkkönen, J.; Juvonen, R.O.; Mahadik, K.R.; Paradkar, A.R. Transferrin mediated solid lipid nanoparticles containing curcumin: Enhanced in vitro anticancer activity by induction of apoptosis. Int. J. Pharm. 2010, 398, 190–203. [Google Scholar] [CrossRef] [PubMed]
- Torchilin, V.P. Micellar Nanocarriers: Pharmaceutical Perspectives. Pharm. Res. 2007, 24, 1–16. [Google Scholar] [CrossRef]
- Danhier, F.; Vroman, B.; Lecouturier, N.; Crokart, N.; Pourcelle, V.; Freichels, H.; Jérôme, C.; Marchand-Brynaert, J.; Feron, O.; Préat, V. Targeting of tumor endothelium by RGD-grafted PLGA-nanoparticles loaded with Paclitaxel. J. Control. Release 2009, 140, 166–173. [Google Scholar] [CrossRef] [PubMed]
- Shaji, J.; Bajaj, R. Formulation development of 5-fluorouracil transethosomes for skin cancer therapy. Int. J. Pharm. Pharm. Res. 2017, 11, 454–464. [Google Scholar]
- Shaji, J. Optimization and Characterization of 5-Fluorouracil Transethosomes for Skin Cancer Therapy Using Response Surface Methodology. Int. J. Adv. Res. 2018, 6, 1225–1233. [Google Scholar] [CrossRef]
- Moolakkadath, T.; Aqil, M.; Ahad, A.; Imam, S.S.; Praveen, A.; Sultana, Y.; Mujeeb, M.; Iqbal, Z. Fisetin loaded binary ethosomes for management of skin cancer by dermal application on UV exposed mice. Int. J. Pharm. 2019, 560, 78–91. [Google Scholar] [CrossRef]
- Mbah, C.C.; Builders, P.F.; Attama, A.A. Nanovesicular carriers as alternative drug delivery systems: Ethosomes in focus. Expert Opin. Drug Deliv. 2014, 11, 45–59. [Google Scholar] [CrossRef]
- Aini, P.N.; Muhammad, D.; Eko, P.G.; Rachmat, M.; Fahmi, E. Formulation, Characterization and Antioxidant Myricetin Nanophytosome for Topical Delivery. Asian J. Pharm. Res. Dev. 2020, 8, 9–13. [Google Scholar] [CrossRef]
- Al-Musawi, S.; Ibraheem, S.; Mahdi, S.A.; Albukhaty, S.; Haider, A.J.; Kadhim, A.A.; Kadhim, K.A.; Kadhim, H.A.; Al-Karagoly, H. Smart Nanoformulation Based on Polymeric Magnetic Nanoparticles and Vincristine Drug: A Novel Therapy for Apoptotic Gene Expression in Tumors. Life 2021, 11, 71. [Google Scholar] [CrossRef]
- Calienni, M.N.; Vega, D.M.; Temprana, C.F.; Izquierdo, M.C.; Ybarra, D.E.; Bernabeu, E.; Moretton, M.; Alvira, F.C.; Chiappetta, D.; Alonso, S.d.V.; et al. The Topical Nanodelivery of Vismodegib Enhances Its Skin Penetration and Performance In Vitro While Reducing Its Toxicity In Vivo. Pharmaceutics 2021, 13, 186. [Google Scholar] [CrossRef] [PubMed]
- Calienni, M.N.; Febres-Molina, C.; Llovera, R.E.; Zevallos-Delgado, C.; Tuttolomondo, M.E.; Paolino, D.; Fresta, M.; Barazorda-Ccahuana, H.L.; Gómez, B.; Alonso, S.D.V.; et al. Nanoformulation for potential topical delivery of Vismodegib in skin cancer treatment. Int. J. Pharm. 2019, 565, 108–122. [Google Scholar] [CrossRef]
- Gamal, F.A.; Sayed, O.M.; El-Ela, F.I.A.; Kharshoum, R.M.; Salem, H.F. Treatment of Basal Cell Carcinoma via Binary Ethosomes of Vismodegib: In Vitro and In Vivo Studies. AAPS PharmSciTech 2020, 21, 51. [Google Scholar] [CrossRef]
- Krishnan, V.; Mitragotri, S. Nanoparticles for topical drug delivery: Potential for skin cancer treatment. Adv. Drug Deliv. Rev. 2020, 153, 87–108. [Google Scholar] [CrossRef] [PubMed]
- Jadhav, S.M.; Morey, P.; Karpe, M.M.; Kadam, V. Novel vesicular system: An overview. J. Appl. Pharm. Sci. 2012, 2, 193–202. [Google Scholar]
- Yu, H.; Li, J.; Shi, K.; Huang, Q. Structure of modified ε-polylysine micelles and their application in improving cellular antioxidant activity of curcuminoids. Food Funct. 2011, 2, 373–380. [Google Scholar] [CrossRef] [Green Version]
- Nishiyama, N.; Kataoka, K. Current state, achievements, and future prospects of polymeric micelles as nanocarriers for drug and gene delivery. Pharmacol. Ther. 2006, 112, 630–648. [Google Scholar] [CrossRef] [PubMed]
- Barrera, M.F.; Enriquez, M.F.J.; Nicolaza, P.; De Leon, G.R.D.; Olivares, R.J.L. Recent Progress in Antitumoral Nanotechnology. Int. J. Nanopart. Nanotechnol. 2015, 1, 1. [Google Scholar] [CrossRef]
- Lohumi, A. A novel drug delivery system: Niosomes review. J. Drug Deliv. Ther. 2012, 2, 129–135. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, N.; Kumar, K.; Pant, N.C. An updated review on transfersomes: A novel vesicular system for transdermal drug delivery. Univers. J. Pharm. Res. 2017, 2, 42–45. [Google Scholar] [CrossRef]
- Opatha, S.A.T.; Titapiwatanakun, V.; Chutoprapat, R. Transfersomes: A Promising Nanoencapsulation Technique for Transdermal Drug Delivery. Pharmaceutics 2020, 12, 855. [Google Scholar] [CrossRef] [PubMed]
- Manosroi, A.; Jantrawut, P.; Khositsuntiwong, N.; Manosroi, W.; Manosroi, J. Novel Elastic Nanovesicles for Cosmeceutical and Pharmaceutical Applications. Chiang Mai J. Sci. 2009, 36, 166–178. [Google Scholar]
- Gamal, A.; Saeed, H.; El-Ela, F.I.A.; Salem, H.F. Improving the Antitumor Activity and Bioavailability of Sonidegib for the Treatment of Skin Cancer. Pharmaceutics 2021, 13, 1560. [Google Scholar] [CrossRef]
- Imran, M.; Iqubal, M.K.; Imtiyaz, K.; Saleem, S.; Mittal, S.; Rizvi, M.M.A.; Ali, J.; Baboota, S. 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. 2020, 587, 119705. [Google Scholar] [CrossRef]
- Gu, Y.; Yang, M.; Tang, X.; Wang, T.; Yang, D.; Zhai, G.; Liu, J. Lipid nanoparticles loading triptolide for transdermal delivery: Mechanisms of penetration enhancement and transport properties. J. Nanobiotechnol. 2018, 16, 68. [Google Scholar] [CrossRef] [Green Version]
- Sahu, S.; Saraf, S.; Kaur, C.D.; Saraf, S. Biocompatible Nanoparticles for Sustained Topical Delivery of Anticancer Phytoconstituent Quercetin. Pak. J. Biol. Sci. 2013, 16, 601–609. [Google Scholar] [CrossRef] [Green Version]
- Kabanov, A.V.; Vinogradov, S.V. Nanogels as Pharmaceutical Carriers: Finite Networks of Infinite Capabilities. Angew. Chem. Int. Ed. 2009, 48, 5418–5429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghorab, M.M.; Alsaid, M.S.; El-Gaby, M.S.A.; Elaasser, M.M.; Nissan, Y.M. Antimicrobial and anticancer activity of some novel fluorinated thiourea derivatives carrying sulfonamide moieties: Synthesis, biological evaluation and molecular docking. Chem. Cent. J. 2017, 11, 32. [Google Scholar] [CrossRef] [Green Version]
- Fadeel, D.A.A.; Kamel, R.; Fadel, M. PEGylated lipid nanocarrier for enhancing photodynamic therapy of skin carcinoma using curcumin: In-vitro/in-vivo studies and histopathological examination. Sci. Rep. 2020, 10, 10435. [Google Scholar] [CrossRef] [PubMed]
- Shakeel, F.; Haq, N.; Al-Dhfyan, A.; Alanazi, F.K.; Alsarra, I.A. Chemoprevention of skin cancer using low HLB surfactant nanoemulsion of 5-fluorouracil: A preliminary study. Drug Deliv. 2015, 22, 573–580. [Google Scholar] [CrossRef] [PubMed]
- Ekambaram, R.; Saravanan, S.; Selvam, N.; Dharmalingam, S. Statistical optimization of novel acemannan polysaccharides assisted TiO2 nanorods based nanofibers for skin cancer application. Carbohydr. Polym. Technol. Appl. 2021, 2, 100048. [Google Scholar] [CrossRef]
- Saber, M.M.; Mirtajani, S.B.; Karimzadeh, K. Green synthesis of silver nanoparticles using Trapa natans extract and their anticancer activity against A431 human skin cancer cells. J. Drug Deliv. Sci. Technol. 2018, 47, 375–379. [Google Scholar] [CrossRef]
- Sharma, A.; Sharma, U.S. Liposomes in drug delivery: Progress and limitations. Int. J. Pharm. 1997, 154, 123–140. [Google Scholar] [CrossRef]
- Dua, J.S.; Rana, P.A.C.; Bhandari, D.A.K. Liposome: Methods of preparation and applications. Int. J. Pharm. Stud. Res. 2012, 3, 14–20. [Google Scholar]
- Alavi, M.; Karimi, N.; Safaei, M. Application of Various Types of Liposomes in Drug Delivery Systems. Adv. Pharm. Bull. 2017, 7, 3–9. [Google Scholar] [CrossRef]
- Barenholz, Y. Liposome application: Problems and prospects. Curr. Opin. Colloid Interface Sci. 2001, 6, 66–77. [Google Scholar] [CrossRef]
- Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef] [Green Version]
- Schaeffer, H.E.; Krohn, D.L. Liposomes in topical drug delivery. Investig. Ophthalmol. Vis. Sci. 1982, 22, 220–227. [Google Scholar]
- Park, J.W. Liposome-based drug delivery in breast cancer treatment. Breast Cancer Res. 2002, 4, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haider, T.; Dubey, S.; Kanwar, I.L.; Pandey, V.; Soni, V. Preparation, Optimization and in vitro Studies of Spectrin Decorated Liposomes: A Promising Strategy for Cancer Treatment: Spectrin anchored liposomes for treatment of cancer. Trends Pept. Protein Sci. 2021, 6, e6. [Google Scholar] [CrossRef]
- Petrilli, R.; Eloy, J.O.; Saggioro, F.P.; Chesca, D.L.; de Souza, M.C.; Dias, M.V.; Luis, L.; Lee, R.J.; Lopez, R.F. Skin cancer treatment effectiveness is improved by iontophoresis of EGFR-targeted liposomes containing 5-FU compared with subcutaneous injection. J. Control. Release 2018, 283, 151–162. [Google Scholar] [CrossRef]
- Adler-Moore, J.P.; Chiang, S.-M.; Satorius, A.; Guerra, D.; McAndrews, B.; McManus, E.J.; Proffitt, R.T. Treatment of murine candidosis and cryptococcosis with a unilamellar liposomal amphotericin B formulation (AmBisome). J. Antimicrob. Chemother. 1991, 28, 63–71. [Google Scholar] [CrossRef] [PubMed]
- Yarosh, D.; Alas, L.G.; Yee, V.; Oberyszyn, A.; Kibitel, J.T.; Mitchell, D.; Rosenstein, R.; Spinowitz, A.; Citron, M. Pyrimidine dimer removal enhanced by DNA repair liposomes reduces the incidence of UV skin cancer in mice. Cancer Res. 1992, 52, 4227–4231. [Google Scholar]
- Bayoumi, M.; Arafa, M.G.; Nasr, M.; Sammour, O.A. Nobiletin-loaded composite penetration enhancer vesicles restore the normal miRNA expression and the chief defence antioxidant levels in skin cancer. Sci. Rep. 2021, 11, 20197. [Google Scholar] [CrossRef] [PubMed]
- Singh, S. Liposome encapsulation of doxorubicin and celecoxib in combination inhibits progression of human skin cancer cells. Int. J. Nanomed. 2018, 13 (Suppl. S1), 11–13. [Google Scholar] [CrossRef] [Green Version]
- Chen, B.; Le, W.; Wang, Y.; Li, Z.; Wang, D.; Ren, L.; Lin, L.; Cui, S.; Hu, J.J.; Hu, Y.; et al. Targeting Negative Surface Charges of Cancer Cells by Multifunctional Nanoprobes. Theranostics 2016, 6, 1887–1898. [Google Scholar] [CrossRef]
- Haider, T.; Soni, V. Response surface methodology and artificial neural network-based modeling and optimization of phosphatidylserine targeted nanocarriers for effective treatment of cancer: In vitro and in silico studies. J. Drug Deliv. Sci. Technol. 2022, 75, 103663. [Google Scholar] [CrossRef]
- Jose, A.; Labala, S.; Ninave, K.M.; Gade, S.K.; Venuganti, V.V.K. Effective Skin Cancer Treatment by Topical Co-delivery of Curcumin and STAT3 siRNA Using Cationic Liposomes. AAPS PharmSciTech 2018, 19, 166–175. [Google Scholar] [CrossRef]
- Haider, T.; Tiwari, R.; Vyas, S.P.; Soni, V. Molecular determinants as therapeutic targets in cancer chemotherapy: An update. Pharmacol. Ther. 2019, 200, 85–109. [Google Scholar] [CrossRef]
- Haider, T.; Sandha, K.K.; Soni, V.; Gupta, P.N. Recent advances in tumor microenvironment associated therapeutic strategies and evaluation models. Mater. Sci. Eng. C 2020, 116, 111229. [Google Scholar] [CrossRef]
- Lee, C.; Na, K. Anthocyanin-Loaded Liposomes Prepared by the pH-Gradient Loading Method to Enhance the Anthocyanin Stability, Antioxidation Effect and Skin Permeability. Macromol. Res. 2020, 28, 289–297. [Google Scholar] [CrossRef]
- Castañeda-Reyes, E.D.; Perea-Flores, M.D.J.; Davila-Ortiz, G.; Lee, Y.; de Mejia, E.G. Development, Characterization and Use of Liposomes as Amphipathic Transporters of Bioactive Compounds for Melanoma Treatment and Reduction of Skin Inflammation: A Review. Int. J. Nanomed. 2020, 15, 7627–7650. [Google Scholar] [CrossRef]
- Marwah, M.; Perrie, Y.; Badhan, R.K.S.; Lowry, D. Intracellular uptake of EGCG-loaded deformable controlled release liposomes for skin cancer. J. Liposome Res. 2020, 30, 136–149. [Google Scholar] [CrossRef] [PubMed]
- El-Kayal, M.; Nasr, M.; Elkheshen, S.; Mortada, N. Colloidal (-)-epigallocatechin-3-gallate vesicular systems for prevention and treatment of skin cancer: A comprehensive experimental study with preclinical investigation. Eur. J. Pharm. Sci. 2019, 137, 104972. [Google Scholar] [CrossRef] [PubMed]
- Sharma, R.; Mody, N.; Kushwah, V.; Jain, S.; Vyas, S.P. C-Type lectin receptor(s)-targeted nanoliposomes: An intelligent approach for effective cancer immunotherapy. Nanomedicine 2017, 12, 1945–1959. [Google Scholar] [CrossRef]
- Haider, M.; Kanoujia, J.; Tripathi, C.B.; Arya, M.; Kaithwas, G.; Saraf, S.A. Pioglitazone loaded vesicular carriers for anti-diabetic activity: Development and optimization as per central composite design. J. Pharm. Sci. Pharmacol. 2015, 2, 11–20. [Google Scholar] [CrossRef]
- Gharbavi, M.; Amani, J.; Kheiri-Manjili, H.; Danafar, H.; Sharafi, A. Niosome: A Promising Nanocarrier for Natural Drug Delivery through Blood-Brain Barrier. Adv. Pharmacol. Sci. 2018, 2018, 6847971. [Google Scholar] [CrossRef]
- Sarker, A.; Shimu, I.J.; Alam, S.A.A. Niosome: As dermal drug delivery tool. IOSR J. Pharm. Biol. Sci. 2015, 10, 73–79. [Google Scholar]
- Bhardwaj, P.; Tripathi, P.; Gupta, R.; Pandey, S. Niosomes: A review on niosomal research in the last decade. J. Drug Deliv. Sci. Technol. 2020, 56, 101581. [Google Scholar] [CrossRef]
- Hamishehkar, H.; Rahimpour, Y.; Kouhsoltani, M. Niosomes as a propitious carrier for topical drug delivery. Expert Opin. Drug Deliv. 2013, 10, 261–272. [Google Scholar] [CrossRef]
- Chen, S.; Hanning, S.; Falconer, J.; Locke, M.; Wen, J. Recent advances in non-ionic surfactant vesicles (niosomes): Fabrication, characterization, pharmaceutical and cosmetic applications. Eur. J. Pharm. Biopharm. 2019, 144, 18–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jothy, M.A.; Shanmuganathan, S. An overview on niosome as carrier in dermal drug delivery. J. Pharm. Sci. Res. 2015, 7, 923. [Google Scholar]
- Moghassemi, S.; Hadjizadeh, A. Nano-niosomes as nanoscale drug delivery systems: An illustrated review. J. Control. Release 2014, 185, 22–36. [Google Scholar] [CrossRef]
- Kazi, K.M.; Mandal, A.S.; Biswas, N.; Guha, A.; Chatterjee, S.; Behera, M.; Kuotsu, K. Niosome: A future of targeted drug delivery systems. J. Adv. Pharm. Technol. Res. 2010, 1, 374–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Y.-Q.; Chen, W.-R.; Tsosie, J.K.; Xie, X.; Li, P.; Wan, J.-B.; He, C.-W.; Chen, M.-W. Niosome Encapsulation of Curcumin: Characterization and Cytotoxic Effect on Ovarian Cancer Cells. J. Nanomater. 2016, 2016, 6365295. [Google Scholar] [CrossRef] [Green Version]
- Yeo, P.L.; Lim, C.L.; Chye, S.M.; Ling, A.P.K.; Koh, R.Y. Niosomes: A review of their structure, properties, methods of preparation, and medical applications. Asian Biomed. 2017, 11, 301–314. [Google Scholar] [CrossRef] [Green Version]
- Shinu, P.; Nair, A.B.; Kumari, B.; Jacob, S.; Kumar, M.; Tiwari, A.; Tiwari, V.; Venugopala, K.N.; Attimarad, M.; Nagaraja, S. Recent Advances and Appropriate use of Niosomes for the Treatment of Skin Cancer. Indian J. Pharm. Educ. Res. 2022, 56, 924–937. [Google Scholar] [CrossRef]
- Sharma, P.K.S.P.; Saxena, P.; Jaswanth, A.; Chalamaiah, M.; Tekade, K.R.; Balasubramaniam, A. Novel Encapsulation of Lycopene in Niosomes and Assessment of its Anticancer Activity. J. Bioequiv. Bioavailab. 2016, 8, 224–232. [Google Scholar] [CrossRef]
- Chermahini, S.H.; Najafi, R.B. Niosome encapsulated fluorouracil as drug delivery system to basal-cell skin Cancer. J. Nanosci. Nanomed 2019, 3, 1–4. [Google Scholar]
- Paolino, D.; Cosco, D.; Muzzalupo, R.; Trapasso, E.; Picci, N.; Fresta, M. Innovative bola-surfactant niosomes as topical delivery systems of 5-fluorouracil for the treatment of skin cancer. Int. J. Pharm. 2008, 353, 233–242. [Google Scholar] [CrossRef] [PubMed]
- Pawar, S.; Shevalkar, G.; Vavia, P. Glucosamine-anchored doxorubicin-loaded targeted nano-niosomes: Pharmacokinetic, toxicity and pharmacodynamic evaluation. J. Drug Target. 2016, 24, 730–743. [Google Scholar] [CrossRef] [PubMed]
- Shah, H.S.; Gotecha, A.; Jetha, D.; Rajput, A.; Bariya, A.; Panchal, S.; Butani, S. Gamma oryzanol niosomal gel for skin cancer: Formulation and optimization using quality by design (QbD) approach. AAPS Open 2021, 7, 9. [Google Scholar] [CrossRef]
- Chauhan, P.; Tyagi, B.K. Herbal novel drug delivery systems and transfersomes. J. Drug Deliv. Ther. 2018, 8, 162–168. [Google Scholar] [CrossRef]
- Benson, H.A.E. Transfersomes for transdermal drug delivery. Expert Opin. Drug Deliv. 2006, 3, 727–737. [Google Scholar] [CrossRef]
- Kumar, N.; Dubey, A.; Mishra, A.; Tiwari, P. Ethosomes: A Novel Approach in Transdermal Drug Delivery System. Int. J. Pharm. Life Sci. 2020, 11, 6598–6608. [Google Scholar]
- Jangdey, M.S.; Gupta, A.; Saraf, S.; Saraf, S. Development and optimization of apigenin-loaded transfersomal system for skin cancer delivery: In vitro evaluation. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1452–1462. [Google Scholar] [CrossRef] [Green Version]
- Raahulan, S.; Sanapalli, B.K.R.; Karri, V.V.S.R. Veera Venkata Satyanarayana Reddy, Paclitaxel loaded transfersomal vesicular drug delivery for the treatment of melanoma skin cancers. Int. J. Res. Pharm. Sci. 2019, 10, 2891–2897. [Google Scholar] [CrossRef]
- Jangdey, M.S.; Kaur, C.D.; Saraf, S. Efficacy of Concanavalin-A conjugated nanotransfersomal gel of apigenin for enhanced targeted delivery of UV induced skin malignant melanoma. Artif. Cells Nanomed. Biotechnol. 2019, 47, 904–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aute, P.P.; Kamble, M.S.; Chaudhari, P.D.; Bhosale, A.V. A comprehensive review on ethosomes. Int. J. Res. Dev. Pharm. Life Sci. 2012, 2, 218–224. [Google Scholar]
- Parashar, T.; Sachan, R.; Singh, V.; Singh, G.; Tyagi, S.; Patel, C.; Gupta, A. Ethosomes: A recent vesicle of transdermal drug delivery system. Int. J. Res. Dev. Pharm. Life Sci. 2013, 2, 285–292. [Google Scholar]
- Hariharanb, S.; Justinc, A. Topical delivery of drugs using ethosomes: A review. Indian Drugs 2019, 56, 7. [Google Scholar]
- Zhang, Z.; Wo, Y.; Zhang, Y.; Wang, D.; He, R.; Chen, H.; Cui, D. In vitro study of ethosome penetration in human skin and hypertrophic scar tissue. Nanomed. Nanotechnol. Biol. Med. 2012, 8, 1026–1033. [Google Scholar] [CrossRef]
- Zhang, J.-P.; Wei, Y.-H.; Zhou, Y.; Li, Y.-Q.; Wu, X.-A. Ethosomes, binary ethosomes and transfersomes of terbinafine hydrochloride: A comparative study. Arch. Pharmacal Res. 2012, 35, 109–117. [Google Scholar] [CrossRef]
- Ismail, T.A.; Shehata, T.M.; Mohamed, D.I.; Elsewedy, H.S.; Soliman, W.E. Quality by Design for Development, Optimization and Characterization of Brucine Ethosomal Gel for Skin Cancer Delivery. Molecules 2021, 26, 3454. [Google Scholar] [CrossRef]
- Nainwal, N.; Jawla, S.; Singh, R.; Saharan, V.A. Transdermal applications of ethosomes—A detailed review. J. Liposome Res. 2019, 29, 103–113. [Google Scholar] [CrossRef]
- Cristiano, M.C.; Froiio, F.; Spaccapelo, R.; Mancuso, A.; Nisticò, S.P.; Udongo, B.P.; Fresta, M.; Paolino, D. Sulforaphane-Loaded Ultradeformable Vesicles as A Potential Natural Nanomedicine for the Treatment of Skin Cancer Diseases. Pharmaceutics 2020, 12, 6. [Google Scholar] [CrossRef] [Green Version]
- Peram, M.R.; Jalalpure, S.; Kumbar, V.; Patil, S.; Joshi, S.; Bhat, K.; Diwan, P. Factorial design based curcumin ethosomal nanocarriers for the skin cancer delivery: In Vitro evaluation. J. Liposome Res. 2019, 29, 291–311. [Google Scholar] [CrossRef]
- Moolakkadath, T.; Aqil, M.; Ahad, A.; Imam, S.S.; Iqbal, B.; Sultana, Y.; Mujeeb, M.; Iqbal, Z. Development of transethosomes formulation for dermal fisetin delivery: Box–Behnken design, optimization, in vitro skin penetration, vesicles–skin interaction and dermatokinetic studies. Artif. Cells Nanomed. Biotechnol. 2018, 46 (Suppl. S2), 755–765. [Google Scholar] [CrossRef] [Green Version]
- Mousa, I.A.; Hammady, T.M.; Gad, S.; Zaitone, S.A.; El-Sherbiny, M.; Sayed, O.M. Formulation and Characterization of Metformin-Loaded Ethosomes for Topical Application to Experimentally Induced Skin Cancer in Mice. Pharmaceuticals 2022, 15, 657. [Google Scholar] [CrossRef]
- Garg, V.; Singh, H.; Bhatia, A.; Raza, K.; Singh, S.K.; Singh, B.; Beg, S. Systematic Development of Transethosomal Gel System of Piroxicam: Formulation Optimization, In Vitro Evaluation, and Ex Vivo Assessment. AAPS PharmSciTech 2017, 18, 58–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdulbaqi, I.M.; Darwis, Y.; Assi, R.A.; Khan, N.A.K. Transethosomal gels as carriers for the transdermal delivery of colchicine: Statistical optimization, characterization, and ex vivo evaluation. Drug Des. Dev. Ther. 2018, 12, 795–813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Šturm, L.; Ulrih, N.P. Basic Methods for Preparation of Liposomes and Studying Their Interactions with Different Compounds, with the Emphasis on Polyphenols. Int. J. Mol. Sci. 2021, 22, 6547. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, T.; Singh, J.; Kaur, S.; Sandhu, S.; Singh, G.; Kaur, I.P. Enhancing Bioavailability and Stability of Curcumin Using Solid Lipid Nanoparticles (CLEN): A Covenant for Its Effectiveness. Front. Bioeng. Biotechnol. 2020, 8, 879. [Google Scholar] [CrossRef]
- Rohit, B.; Pal, K.I. A method to prepare solid lipid nanoparticles with improved entrapment efficiency of hydrophilic drugs. Curr. Nanosci. 2013, 9, 211–220. [Google Scholar] [CrossRef]
- Sabir, F.; Qindeel, M.; Rehman, A.U.; Ahmad, N.M.; Khan, G.M.; Csoka, I.; Ahmed, N. An efficient approach for development and optimisation of curcumin-loaded solid lipid nanoparticles’ patch for transdermal delivery. J. Microencapsul. 2021, 38, 233–248. [Google Scholar] [CrossRef]
- Banerjee, I.; De, M.; Dey, G.; Bharti, R.; Chattopadhyay, S.; Ali, N.; Chakrabarti, P.; Reis, R.L.; Kundu, S.C.; Mandal, M. A peptide-modified solid lipid nanoparticle formulation of paclitaxel modulates immunity and outperforms dacarbazine in a murine melanoma model. Biomater. Sci. 2019, 7, 1161–1178. [Google Scholar] [CrossRef]
- Müller, R.H.; Petersen, R.D.; Hommoss, A.; Pardeike, J. Nanostructured lipid carriers (NLC) in cosmetic dermal products. Adv. Drug Deliv. Rev. 2007, 59, 522–530. [Google Scholar] [CrossRef]
- Müller, R.H.; Radtke, M.; Wissing, S.A. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. Adv. Drug Deliv. Rev. 2002, 1 (Suppl. S54), S131–S155. [Google Scholar] [CrossRef]
- Agrawal, R.D.; Tatode, A.A.; Rarokar, N.R.; Umekar, M.J. Polymeric micelle as a nanocarrier for delivery of therapeutic agents: A comprehensive review. J. Drug Deliv. Ther. 2020, 10, 191–195. [Google Scholar] [CrossRef] [Green Version]
- Moradi, A.; Farboud, E.S.; Nasrollahi, S.A.; Kashani, M.N.; Dinarvand, R. Tretinoin loaded nanostructured lipid carrier (NLC): Safe and effective drug delivery system. Nanosci. Nanotechnol.-Asia 2017, 7, 221–229. [Google Scholar] [CrossRef]
- Iqbal, B.; Ali, J.; Ganguli, M.; Mishra, S.; Baboota, S. Silymarin-loaded nanostructured lipid carrier gel for the treatment of skin cancer. Nanomedicine 2019, 14, 1077–1093. [Google Scholar] [CrossRef] [PubMed]
- Gundogdu, E.; Demir, E.-S.; Ekinci, M.; Ozgenc, E.; Ilem-Ozdemir, D.; Senyigit, Z.; Gonzalez-Alvarez, I.; Bermejo, M. An Innovative Formulation Based on Nanostructured Lipid Carriers for Imatinib Delivery: Pre-Formulation, Cellular Uptake and Cytotoxicity Studies. Nanomaterials 2022, 12, 250. [Google Scholar] [CrossRef] [PubMed]
- Mehan, N.; Kumar, M.; Bhatt, S.; Shankar, R.; Kumari, B.; Pahwa, R.; Kaushik, D. Self-Assembly polymeric nano micelles for the futuristic treatment of skin cancer and phototoxicity: Therapeutic and clinical advancement. Crit. Rev.™ Ther. Drug Carr. Syst. 2022, 39, 79–95. [Google Scholar] [CrossRef] [PubMed]
- Quiñones, O.G.; Pierre, M.B.R. Cutaneous Application of Celecoxib for Inflammatory and Cancer Diseases. Curr. Cancer Drug Targets 2019, 19, 5–16. [Google Scholar] [CrossRef]
- Lapteva, M.; Santer, V.; Mondon, K.; Patmanidis, I.; Chiriano, G.; Scapozza, L.; Gurny, R.; Möller, M.; Kalia, Y.N. Targeted cutaneous delivery of ciclosporin A using micellar nanocarriers and the possible role of inter-cluster regions as molecular transport pathways. J. Control. Release 2014, 196, 9–18. [Google Scholar] [CrossRef]
- Pandey, V.; Haider, T.; Chandak, A.R.; Chakraborty, A.; Banerjee, S.; Soni, V. Technetium labeled doxorubicin loaded silk fibroin nanoparticles: Optimization, characterization and in vitro evaluation. J. Drug Deliv. Sci. Technol. 2020, 56, 101539. [Google Scholar] [CrossRef]
- Haider, T.; Pandey, V.; Behera, C.; Kumar, P.; Gupta, P.N.; Soni, V. Nisin and nisin-loaded nanoparticles: A cytotoxicity investigation. Drug Dev. Ind. Pharm. 2022, 48, 310–321. [Google Scholar] [CrossRef] [PubMed]
- Pandey, V.; Haider, T.; Agrawal, P.; Soni, S.; Soni, V. Advances in Natural Polymeric Nanoparticles for the Drug Delivery. In Advanced Drug Delivery Methods; IntechOpen: London, UK, 2022. [Google Scholar] [CrossRef]
- Akram, W.; Garud, N. Design expert as a statistical tool for optimization of 5-ASA-loaded biopolymer-based nanoparticles using Box Behnken factorial design. Future J. Pharm. Sci. 2021, 7, 146. [Google Scholar] [CrossRef]
- Zhang, Z.; Tsai, P.-C.; Ramezanli, T.; Michniak-Kohn, B.B. Polymeric nanoparticles-based topical delivery systems for the treatment of dermatological diseases. WIREs Nanomed. Nanobiotechnol. 2013, 5, 205–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, L.; Liu, Z.; Wang, L.; Cun, D.; Tong, H.H.Y.; Yan, R.; Chen, X.; Wang, R.; Zheng, Y. Enhanced topical penetration, system exposure and anti-psoriasis activity of two particle-sized, curcumin-loaded PLGA nanoparticles in hydrogel. J. Control. Release 2017, 254, 44–54. [Google Scholar] [CrossRef]
- Valizadeh, A.; Khaleghi, A.A.; Alipanah, H.; Zarenezhad, E.; Osanloo, M. Anticarcinogenic Effect of Chitosan Nanoparticles Containing Syzygium aromaticum Essential Oil or Eugenol Toward Breast and Skin Cancer Cell Lines. BioNanoScience 2021, 11, 678–686. [Google Scholar] [CrossRef]
- Neelakandan, M.; Manoharan, S.; Muralinaidu, R.; Thara, J.M. Tumor preventive and antioxidant efficacy of chlorogenic acid–loaded chitosan nanoparticles in experimental skin carcinogenesis. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 533–546. [Google Scholar] [CrossRef]
- Ramezanli, T.; Kilfoyle, B.E.; Zhang, Z.; Michniak-Kohn, B.B. Polymeric nanospheres for topical delivery of vitamin D3. Int. J. Pharm. 2017, 516, 196–203. [Google Scholar] [CrossRef] [Green Version]
- Das, S.; Das, J.; Samadder, A.; Paul, A.; Khuda-Bukhsh, A.R. Efficacy of PLGA-loaded apigenin nanoparticles in Benzo[a]pyrene and ultraviolet-B induced skin cancer of mice: Mitochondria mediated apoptotic signalling cascades. Food Chem. Toxicol. 2013, 62, 670–680. [Google Scholar] [CrossRef]
- Yaman, S.; Ramachandramoorthy, H.; Oter, G.; Zhukova, D.; Nguyen, T.; Sabnani, M.K.; Weidanz, J.A.; Nguyen, K.T. Melanoma Peptide MHC Specific TCR Expressing T-Cell Membrane Camouflaged PLGA Nanoparticles for Treatment of Melanoma Skin Cancer. Front. Bioeng. Biotechnol. 2020, 8, 943. [Google Scholar] [CrossRef]
- Pandey, V.; Haider, T.; Chandak, A.R.; Chakraborty, A.; Banerjee, S.; Soni, V. Surface modified silk fibroin nanoparticles for improved delivery of doxorubicin: Development, characterization, in-vitro studies. Int. J. Biol. Macromol. 2020, 164, 2018–2027. [Google Scholar] [CrossRef]
- Nayak, D.; Thathapudi, N.C.; Ashe, S.; Nayak, B. Bioengineered ethosomes encapsulating AgNPs and Tasar silk sericin proteins for non melanoma skin carcinoma (NMSC) as an alternative therapeutics. Int. J. Pharm. 2021, 596, 120265. [Google Scholar] [CrossRef]
- Balashanmugam, P.; Sucharithra, G.; Agnes Mary, S.; Tamil Selvi, A. Efficacy of biopolymeric PVA-AuNPs and PCL-Curcumin loaded electrospun nanofibers and their anticancer activity against A431 skin cancer cell line. Mater. Today Commun. 2020, 25, 101276. [Google Scholar] [CrossRef]
- Hong, W.; Guo, F.; Yu, N.; Ying, S.; Lou, B.; Wu, J.; Gao, Y.; Ji, X.; Wang, H.; Li, A.; et al. A Novel Folic Acid Receptor-Targeted Drug Delivery System Based on Curcumin-Loaded β-Cyclodextrin Nanoparticles for Cancer Treatment, Drug Design. Dev. Ther. 2021, 15, 2843–2855. [Google Scholar] [CrossRef]
- Cao, X.; Tan, T.; Zhu, D.; Yu, H.; Liu, Y.; Zhou, H.; Jin, Y.; Xia, Q. Paclitaxel-Loaded Macrophage Membrane Camouflaged Albumin Nanoparticles for Targeted Cancer Therapy. Int. J. Nanomed. 2020, 15, 1915–1928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scott, R.W.J.; Wilson, O.M.; Crooks, R.M. Synthesis, Characterization, and Applications of Dendrimer-Encapsulated Nanoparticles. J. Phys. Chem. B 2005, 109, 692–704. [Google Scholar] [CrossRef]
- Svenson, S. Dendrimers as versatile platform in drug delivery applications. Eur. J. Pharm. Biopharm. 2009, 71, 445–462. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Zhao, W.; Ma, Q.; Gao, Y.; Wang, W.; Zhang, X.; Dong, Y.; Zhang, T.; Liang, Y.; Han, S. Functional nano-systems for transdermal drug delivery and skin therapy. Nanoscale Adv. 2023, 5, 1527–1558. [Google Scholar] [CrossRef] [PubMed]
- Majoros, I.J.; Myc, A.; Thomas, T.; Mehta, C.B.; Baker, J.R. PAMAM Dendrimer-Based Multifunctional Conjugate for Cancer Therapy: Synthesis, Characterization, and Functionality. Biomacromolecules 2006, 7, 572–579. [Google Scholar] [CrossRef]
- Yiyun, C.; Na, M.; Tongwen, X.; Rongqiang, F.; Xueyuan, W.; Xiaomin, W.; Longping, W. Transdermal Delivery of Nonsteroidal Anti-Inflammatory Drugs Mediated by Polyamidoamine (PAMAM) Dendrimers. J. Pharm. Sci. 2007, 96, 595–602. [Google Scholar] [CrossRef]
- Abbasi, E.; Aval, S.F.; Akbarzadeh, A.; Milani, M.; Nasrabadi, H.T.; Joo, S.W.; Hanifehpour, Y.; Nejati-Koshki, K.; Pashaei-Asl, R. Dendrimers: Synthesis, applications, and properties. Nanoscale Res. Lett. 2014, 9, 247. [Google Scholar] [CrossRef] [Green Version]
- Ybarra, D.E.; Calienni, M.N.; Ramirez, L.F.B.; Frias, E.T.A.; Lillo, C.; Alonso, S.D.V.; Montanari, J.; Alvira, F.C. Vismodegib in PAMAM-dendrimers for potential theragnosis in skin cancer. OpenNano 2022, 7, 100053. [Google Scholar] [CrossRef]
- Thuy, L.T.; Kang, N.; Choi, M.; Lee, M.; Choi, J.S. Dendrimeric micelles composed of polyamidoamine dendrimer-peptide-cholesterol conjugates as drug carriers for the treatment of melanoma and bacterial infection. J. Ind. Eng. Chem. 2022, 114, 361–376. [Google Scholar] [CrossRef]
- Tan, E.; Lim, C. Mechanical characterization of nanofibers–a review. Compos. Sci. Technol. 2006, 66, 1102–1111. [Google Scholar] [CrossRef]
- Verreck, G.; Chun, I.; Rosenblatt, J.; Peeters, J.; Van Dijck, A.; Mensch, J.; Noppe, M.; Brewster, M.E. Incorporation of drugs in an amorphous state into electrospun nanofibers composed of a water-insoluble, nonbiodegradable polymer. J. Control. Release 2003, 92, 349–360. [Google Scholar] [CrossRef]
- Chronakis, I.S. Novel nanocomposites and nanoceramics based on polymer nanofibers using electrospinning process—A review. J. Mater. Process. Technol. 2005, 167, 283–293. [Google Scholar] [CrossRef]
- Goyal, R.; Macri, L.K.; Kaplan, H.M.; Kohn, J. Nanoparticles and nanofibers for topical drug delivery. J. Control. Release 2016, 240, 77–92. [Google Scholar] [CrossRef]
- Patel, G.; Yadav, B.K. Formulation, Characterization and In vitro Cytotoxicity of 5-Fluorouracil Loaded Polymeric Electrospun Nanofibers for the Treatment of Skin Cancer. Recent Pat. Nanotechnol. 2019, 13, 114–128. [Google Scholar] [CrossRef] [PubMed]
- Rengifo, A.F.C.; Stefanes, N.M.; Toigo, J.; Mendes, C.; Argenta, D.F.; Dotto, M.E.R.; da Silva, M.C.S.; Nunes, R.J.; Caon, T.; Parize, A.L.; et al. PEO-chitosan nanofibers containing carboxymethyl-hexanoyl chitosan/dodecyl sulfate nanoparticles loaded with pyrazoline for skin cancer treatment. Eur. Polym. J. 2019, 119, 335–343. [Google Scholar] [CrossRef]
- Balan, P.; Indrakumar, J.; Murali, P.; Korrapati, P.S. Bi-faceted delivery of phytochemicals through chitosan nanoparticles impregnated nanofibers for cancer therapeutics. Int. J. Biol. Macromol. 2020, 142, 201–211. [Google Scholar] [CrossRef]
- Niu, J.; Chu, Y.; Huang, Y.-F.; Chong, Y.-S.; Jiang, Z.-H.; Mao, Z.-W.; Peng, L.-H.; Gao, J.-Q. Transdermal gene delivery by functional peptide-conjugated cationic gold nanoparticle reverses the progression and metastasis of cutaneous melanoma. ACS Appl. Mater. Interfaces 2017, 9, 9388–9401. [Google Scholar] [CrossRef] [PubMed]
- Kraeling, M.E.; Topping, V.D.; Keltner, Z.M.; Belgrave, K.R.; Bailey, K.D.; Gao, X.; Yourick, J.J. In vitro percutaneous penetration of silver nanoparticles in pig and human skin. Regul. Toxicol. Pharmacol. 2018, 95, 314–322. [Google Scholar] [CrossRef] [PubMed]
- Baroli, B.; Ennas, M.G.; Loffredo, F.; Isola, M.; Pinna, R.; López-Quintela, M.A. Penetration of Metallic Nanoparticles in Human Full-Thickness Skin. J. Investig. Dermatol. 2007, 127, 1701–1712. [Google Scholar] [CrossRef]
- Chen, Y.; Wu, Y.; Gao, J.; Zhang, Z.; Wang, L.; Chen, X.; Mi, J.; Yao, Y.; Guan, D.; Chen, B.; et al. Transdermal Vascular Endothelial Growth Factor Delivery with Surface Engineered Gold Nanoparticles. ACS Appl. Mater. Interfaces 2017, 9, 5173–5180. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Lai, X.; Shao, L.; Li, L. Evaluation of immunoresponses and cytotoxicity from skin exposure to metallic nanoparticles. Int. J. Nanomed. 2018, 13, 4445–4459. [Google Scholar] [CrossRef] [Green Version]
- Safwat, M.A.; Soliman, G.M.; Sayed, D.; Attia, M.A. Fluorouracil-Loaded Gold Nanoparticles for the Treatment of Skin Cancer: Development, in Vitro Characterization, and in Vivo Evaluation in a Mouse Skin Cancer Xenograft Model. Mol. Pharm. 2018, 15, 2194–2205. [Google Scholar] [CrossRef]
- Agostinis, P.; Berg, K.; Cengel, K.A.; Foster, T.H.; Girotti, A.W.; Gollnick, S.O.; Hahn, S.M.; Hamblin, M.R.; Juzeniene, A.; Kessel, D.; et al. Photodynamic therapy of cancer: An update. CA Cancer J. Clin. 2011, 61, 250–281. [Google Scholar] [CrossRef]
- Chen, L.; Huang, J.; Li, X.; Huang, M.; Zeng, S.; Zheng, J.; Peng, S.; Li, S. Progress of Nanomaterials in Photodynamic Therapy Against Tumor. Front. Bioeng. Biotechnol. 2022, 10, 920162. [Google Scholar] [CrossRef]
- Li, Y.; He, G.; Fu, L.-H.; Younis, M.R.; He, T.; Chen, Y.; Lin, J.; Li, Z.; Huang, P. A Microneedle Patch with Self-Oxygenation and Glutathione Depletion for Repeatable Photodynamic Therapy. ACS Nano 2022, 16, 17298–17312. [Google Scholar] [CrossRef]
- Wang, X.; Luo, D.; Basilion, J.P. Photodynamic Therapy: Targeting Cancer Biomarkers for the Treatment of Cancers. Cancers 2021, 13, 2992. [Google Scholar] [CrossRef] [PubMed]
- Zhang, D.-Y.; Liang, Y.; Wang, M.; Younis, M.R.; Yi, H.; Zhao, X.; Chang, J.; Zheng, Y.; Guo, W.; Yu, X. Self-Assembled Carrier-Free Nanodrugs for Starvation Therapy-Amplified Photodynamic Therapy of Cancer. Adv. Healthc. Mater. 2023, 2203177. [Google Scholar] [CrossRef]
- Abrahamse, H.; Hamblin, M.R. New photosensitizers for photodynamic therapy. Biochem. J. 2016, 473, 347–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, Y.; Zhou, Y.; Liu, L.; Xu, Y.; Chen, Q.; Wang, Y.; Wu, S.; Deng, Y.; Zhang, J.; Shao, A. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front. Mol. Biosci. 2020, 7, 193. [Google Scholar] [CrossRef] [PubMed]
- Tian, H.; Zhang, T.; Qin, S.; Huang, Z.; Zhou, L.; Shi, J.; Nice, E.C.; Na Xie, N.; Huang, C.; Shen, Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J. Hematol. Oncol. 2022, 15, 132. [Google Scholar] [CrossRef] [PubMed]
- Shang, L.; Zhou, X.; Zhang, J.; Shi, Y.; Zhong, L. Metal Nanoparticles for Photodynamic Therapy: A Potential Treatment for Breast Cancer. Molecules 2021, 26, 6532. [Google Scholar] [CrossRef]
- Gunaydin, G.; Gedik, M.E.; Ayan, S. Photodynamic Therapy—Current Limitations and Novel Approaches. Front. Chem. 2021, 9, 691697. [Google Scholar] [CrossRef]
- Sargazi, S.; Er, S.; Gelen, S.S.; Rahdar, A.; Bilal, M.; Arshad, R.; Ajalli, N.; Khan, M.F.A.; Pandey, S. Application of titanium dioxide nanoparticles in photothermal and photodynamic therapy of cancer: An updated and comprehensive review. J. Drug Deliv. Sci. Technol. 2022, 75, 103605. [Google Scholar] [CrossRef]
- Reis, S.R.R.D.; Helal-Neto, E.; Barros, A.O.D.S.D.; Pinto, S.R.; Portilho, F.L.; Siqueira, L.B.d.O.; Alencar, L.M.R.; Dahoumane, S.A.; Alexis, F.; Ricci-Junior, E.; et al. Dual Encapsulated Dacarbazine and Zinc Phthalocyanine Polymeric Nanoparticle for Photodynamic Therapy of Melanoma. Pharm. Res. 2021, 38, 335–346. [Google Scholar] [CrossRef]
- Li, T.-F.; Xu, H.-Z.; Xu, Y.-H.; Yu, T.-T.; Tang, J.-M.; Li, K.; Wang, C.; Peng, X.-C.; Li, Q.-R.; Sang, X.-Y.; et al. Efficient Delivery of Chlorin e6 by Polyglycerol-Coated Iron Oxide Nanoparticles with Conjugated Doxorubicin for Enhanced Photodynamic Therapy of Melanoma. Mol. Pharm. 2021, 18, 3601–3615. [Google Scholar] [CrossRef]
- Alotaibi, G.; Alharthi, S.; Basu, B.; Ash, D.; Dutta, S.; Singh, S.; Prajapati, B.G.; Bhattacharya, S.; Chidrawar, V.R.; Chitme, H. Nano-Gels: Recent Advancement in Fabrication Methods for Mitigation of Skin Cancer. Gels 2023, 9, 331. [Google Scholar] [CrossRef] [PubMed]
- Sahu, P.; Kashaw, S.K.; Jain, S.; Sau, S.; Iyer, A.K. Assessment of penetration potential of pH responsive double walled biodegradable nanogels coated with eucalyptus oil for the controlled delivery of 5-fluorouracil: In vitro and ex vivo studies. J. Control. Release 2017, 253, 122–136. [Google Scholar] [CrossRef] [PubMed]
- Levang, A.K.; Zhao, K.; Singh, J. Effect of ethanol/propylene glycol on the in vitro percutaneous absorption of aspirin, biophysical changes and macroscopic barrier properties of the skin. Int. J. Pharm. 1999, 181, 255–263. [Google Scholar] [CrossRef] [PubMed]
- Witting, M.; Molina, M.; Obst, K.; Plank, R.; Eckl, K.M.; Hennies, H.C.; Calderón, M.; Frieß, W.; Hedtrich, S. Thermosensitive dendritic polyglycerol-based nanogels for cutaneous delivery of biomacromolecules. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1179–1187. [Google Scholar] [CrossRef]
- Badalkhani, O.; Pires, P.C.; Mohammadi, M.; Babaie, S.; Paiva-Santos, A.C.; Hamishehkar, H. Nanogel Containing Gamma-Oryzanol-Loaded Nanostructured Lipid Carriers and TiO2/MBBT: A Synergistic Nanotechnological Approach of Potent Natural Antioxidants and Nanosized UV Filters for Skin Protection. Pharmaceuticals 2023, 16, 670. [Google Scholar] [CrossRef]
- McClements, D.J.; Rao, J. Food-Grade Nanoemulsions: Formulation, Fabrication, Properties, Performance, Biological Fate, and Potential Toxicity. Crit. Rev. Food Sci. Nutr. 2011, 51, 285–330. [Google Scholar] [CrossRef]
- Shakeel, F.; Baboota, S.; Ahuja, A.; Ali, J.; Aqil, M.; Shafiq, S. Nanoemulsions as vehicles for transdermal delivery of aceclofenac. AAPS PharmSciTech 2007, 8, 104. [Google Scholar] [CrossRef] [Green Version]
- Tadros, T.; Izquierdo, P.; Esquena, J.; Solans, C. Formation and stability of nano-emulsions. Adv. Colloid Interface Sci. 2004, 108–109, 303–318. [Google Scholar] [CrossRef]
- Yukuyama, M.N.; Ghisleni, D.D.M.; Pinto, T.J.A.; Bou-Chacra, N.A. Nanoemulsion: Process selection and application in cosmetics—A review. Int. J. Cosmet. Sci. 2016, 38, 13–24. [Google Scholar] [CrossRef] [Green Version]
- Solans, C.; Izquierdo, P.; Nolla, J.; Azemar, N.; Garcia-Celma, M.J. Nano-emulsions. Curr. Opin. Colloid Interface Sci. 2005, 10, 102–110. [Google Scholar] [CrossRef]
- Martínez-Razo, G.; Pires, P.C.; Domínguez-López, M.L.; Veiga, F.; Vega-López, A.; Paiva-Santos, A.C. Norcantharidin Nanoemulsion Development, Characterization, and In Vitro Antiproliferation Effect on B16F1 Melanoma Cells. Pharmaceuticals 2023, 16, 501. [Google Scholar] [CrossRef] [PubMed]
- Ranjbar, R.; Zarenezhad, E.; Abdollahi, A.; Nasrizadeh, M.; Firooziyan, S.; Namdar, N.; Osanloo, M. Nanoemulsion and Nanogel Containing Cuminum cyminum L Essential Oil: Antioxidant, Anticancer, Antibacterial, and Antilarval Properties. J. Trop. Med. 2023, 2023, 5075581. [Google Scholar] [CrossRef] [PubMed]
Carrier System | Therapeutic Agents | Drawbacks of Drugs in the Treatment of Skin Cancer | Particle Size (nm) | Zeta Potential (mV) | Remarks | Ref. |
---|---|---|---|---|---|---|
Liposomes | Vincristine | Possess a rapid clearance rate, have a large volume of distribution in the body, and dose-related neurotoxicity | 103.6 ± 0.6 | −2.3 ± 0.1 | Enhanced stability, superior antitumor efficacy, and reduction in toxicity | [79] |
Niosomes | Lycopene | Lycopene is sensitive to light and heat and undergoes oxidation when stored. It limits the activity of lycopene | 223 ± 8 | −2.1 ± 1.2 | With enhanced penetration, the activity of lycopene was prevented, and bioavailability increased. | [80,81] |
Transferosomes | Paclitaxel | Low solubility, low permeability, and upon exceeding the dose, causes hypersensitivity reaction | 200.0 | −26.0 | Increased permeability and stability, the better release of drug | [82] |
Ethosomes | Sulforaphane | Poor physiochemical properties and skin permeation | 227 ± 3 | −26 ± 1 | Enhanced skin permeation | [83] |
Transethosomes | 5-Fluorouracil | Low bioavailability and rapid degradation when given orally | 57.0 | −46.19 ± 15.3 | Elevated efficacy and controlled release | [67,68] |
Solid Lipid NPs | Doxorubicin (Dox) | Nonspecific distribution-related side effects are cardiotoxicity, oral ulceration alopecia, and myelosuppression | 92 ± 2.0 | 0.044 ± 0.007 | Maximized efficacy, enhanced stability, and absence of cytotoxicity in untargeted organs | [84] |
Nanostructured lipid carriers | Resveratrol | Lowers blood sugar level on chronic use, physiochemical instability | 191 ± 5.20 | −10.00 ± 0.30 | Enhanced epidermal deposition and site-specific release of drug | [85] |
Natural NPs | Quercetin | Lower stability, Low solubility, conventional formulation requires a higher dose (1500 mg) for an acceptable therapeutic level, and when given orally, it shows low absorption | 228.77 ± 2.0 | −16.7 | Enhanced localized action, dose requirement reduced to 100 mg, stability and solubility improved. | [86] |
Synthetic NPs | Paclitaxel | BCS class Ⅳ drug that shows low permeability and low solubility; side effect includes hypotension, lethargy, neurotoxicity, and nephrotoxicity | 146 ± 2.0 | 0.12 ± 3.6 | Antitumor activity of paclitaxel improved, and endothelium targeting of the tumor was achieved. | [66] |
Dendrimers | Fluoroisothiocynate | Conventional intravenous administration causes difficulty breathing, cardiac arrhythmia, dizziness, severe pain in the arm, and sweating. | 14.45 ± 0.8 | 13.94 ± 1.5 | Dendrimers localize at the targeted site. After iontophoretic delivery of dendrimer, the amount of dendrimer in the epidermis was 3-fold high, and degradation of the enzyme was prevented. | [87,88] |
Nanogels | Curcumin | Low stability and low aqueous solubility limit its clinical application. | -- | −21.6 | Better penetration across the skin and higher cytotoxic activity when compared to conventional pure curcumin. | |
PEG-NPs | Curcumin | Low stability and low aqueous solubility limit its clinical application | 167.60 ± 15.12 | −26.91 | Higher drug release when compared to free curcumin suspension, MTT assays of nanoformulations showed higher efficacy when compared to conventional curcumin suspension | [89] |
Nanoemulsions | 5-fluorouracil | Rapid G.I. degradation when given orally and inadequate bioavailability | 68.20 ± 2.65 | −25.92 | The nanoemulsion was found to be much more productive than free 5-fluorouracil formulation and IC50 the value reported as 398 µM | [90] |
Nanofibres | Resveratrol | Physical instability and chemical instability | 15.9 ± 10.0 | Porosity 90.69% | Permeation across the skin enhanced, a percentage of cell viability of about 37.2% at 500 µg/mL was observed, and increased cytotoxicity activity found | [91] |
Metallic NPs | Trapa natans extract | Physiochemical instability | 30–90 | ----------- | 100 µg/mL concentration of formulation reduces the cell viability of A431 skin cancer cells to 24.3% | [92] |
Niosomes | Liposomes |
---|---|
Less expensive than liposomes | More expensive than niosomes |
Nonionic surfactants are stable | Phospholipids may undergo oxiditive degradation |
The surface charge may present on niosomes | The neutral charge may be due to phospholipid |
The particular method requires the purification, storage, and handling of phospholipids | Comparatively, no particular method requires |
Ingredients | Role | Example |
---|---|---|
Phospholipid | Vesicle forming unit | Phosphatidylcholine, dipalmitoyl phosphatidyl choline |
Edge activators (surface active agents) | Enhance flexibility | Tween 20, span 80, sodium deoxycholate, sodium cholate |
Alcohol | Solvents | Methanol, ethanol |
Buffers | Hydration medium | Phosphate saline (pH 6.4) |
Class | Concentration (%) | Example | Uses |
---|---|---|---|
Phospholipids | 0.5–10 | Phospholipon 90G, 90H, 80H, Lipoid S100, S75, S75–3, E80 Dipalmityl phosphatidylcholine, Distearyl phosphatidylcholine | Vesicle forming unit |
Edge activators/surfactant or permeation enhancer | 10–50 of the totals phospholipid concentration | Tween 60, 80, 20 Span 80, 60, 40, 20 Cremophor RH-40 SPACE (skin penetrating and cell entering peptide) Oleic acid, Sodium cholate, Deoxy sodium cholate, Dimethyl sulfoxide | Increase the skin permeability or act as a penetration enhancer |
Alcohol | 20–50 | Ethanol, Isopropyl alcohol | For providing the softness for vesicle membrane As a penetration enhancer |
Glycol | Propylene glycol (p.G.) Transcutol RTM | Permeation enhancer | |
Cholesterol | 0.1–1 | Cholesterol | Gives stability and rigidity to vesicle |
Dye | q.s. | Rhodamine-123 Rhodamine red Fluorescence Isothiocyanate (FITC) 6-Carboxy fluorescence | Characterization study |
Vehicle | q.s. | Carbopol, etc. | Ac as gel former |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Adnan, M.; Akhter, M.H.; Afzal, O.; Altamimi, A.S.A.; Ahmad, I.; Alossaimi, M.A.; Jaremko, M.; Emwas, A.-H.; Haider, T.; Haider, M.F. Exploring Nanocarriers as Treatment Modalities for Skin Cancer. Molecules 2023, 28, 5905. https://doi.org/10.3390/molecules28155905
Adnan M, Akhter MH, Afzal O, Altamimi ASA, Ahmad I, Alossaimi MA, Jaremko M, Emwas A-H, Haider T, Haider MF. Exploring Nanocarriers as Treatment Modalities for Skin Cancer. Molecules. 2023; 28(15):5905. https://doi.org/10.3390/molecules28155905
Chicago/Turabian StyleAdnan, Mohammad, Md. Habban Akhter, Obaid Afzal, Abdulmalik S. A. Altamimi, Irfan Ahmad, Manal A. Alossaimi, Mariusz Jaremko, Abdul-Hamid Emwas, Tanweer Haider, and Md. Faheem Haider. 2023. "Exploring Nanocarriers as Treatment Modalities for Skin Cancer" Molecules 28, no. 15: 5905. https://doi.org/10.3390/molecules28155905
APA StyleAdnan, M., Akhter, M. H., Afzal, O., Altamimi, A. S. A., Ahmad, I., Alossaimi, M. A., Jaremko, M., Emwas, A. -H., Haider, T., & Haider, M. F. (2023). Exploring Nanocarriers as Treatment Modalities for Skin Cancer. Molecules, 28(15), 5905. https://doi.org/10.3390/molecules28155905