Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Nanocarrier Based Topical Drug Delivery- A Promising Strategy for Treatment of Skin Cancer

Author(s): Rupesh Jain, Ila Sarode, Gautam Singhvi and Sunil Kumar Dubey*

Volume 26, Issue 36, 2020

Page: [4615 - 4623] Pages: 9

DOI: 10.2174/1381612826666200826140448

Price: $65

Abstract

Skin cancers are one of the most widespread and complex forms of the disease, resulting in very high mortality rates across the world. The current treatments available for skin cancer include chemotherapy, surgery, radiotherapy, etc. The selected treatment options for skin cancer are usually decided based on the condition of a patient and the type of skin cancer. The effectiveness of skin cancer therapy is still limited because of poor penetrability of the drug into stratum corneum or lesions, low efficacy, required higher concentration of the active pharmaceutical ingredients to reach a therapeutic effect. Besides, low bioavailability at the site of action, the requirement of high dose, causes skin irritation, which significantly hinders the drug absorption through the stratum corneum. Thus, nanocarriers have been used to bypass the problems associated with conventional anti-cancer drug delivery systems. In the current scenario, nanotechnology-based therapy has shown great potential in the management of skin cancer, and these can be used for a more efficient drug delivery system to treat cancers. In this review article, the information on different nanocarrier systems for skin cancer has been elucidated. Moreover, the various nanoparticulate strategies and their effectiveness to treat skin cancer have been discussed.

Keywords: Nanocarriers, skin, cancer, targeted drug delivery, topical drug delivery, dermatology.

[1]
Brown MB, Martin GP, Jones SA, Akomeah FK. Dermal and transdermal drug delivery systems: current and future prospects. Drug Deliv 2006; 13(3): 175-87.
[http://dx.doi.org/10.1080/10717540500455975 ] [PMID: 16556569]
[2]
Singh Malik D, Mital N, Kaur G. Topical drug delivery systems: A patent review. In: Expert Opinion on Therapeutic Patents. Taylor and Francis Ltd 2016; 26: 213-8.
[3]
Gupta M, Agrawal U, Vyas SP. Nanocarrier-based topical drug delivery for the treatment of skin diseases. Expert Opin Drug Deliv 2012; 9(7): 783-804.
[http://dx.doi.org/10.1517/17425247.2012.686490 ] [PMID: 22559240]
[4]
Das P, Deshmukh N, Badore N, Ghulaxe C, Patel P. A Review Article on Melanoma. J Pharm Sci 2016; 8(2): 112-7.
[5]
Smith V, Walton S. Treatment of facial basal cell carcinoma : A review. J Skin Cancer 2011; 2011 : 380371.
[http://dx.doi.org/10.1155/2011/380371]
[6]
Matei C, Tampa M, Poteca T, et al. Photodynamic therapy in the treatment of basal cell carcinoma. J Med Life 2013; 8(2): 112-7.
[7]
Vances AD, Translational IN. Nanoemulsions and nanoparticles for non-melanoma skin cancer : effects of lipid materials. Clin Transl Oncol 2013; 15(6): 417-24.
[8]
Dianzani C, Zara GP, Maina G, et al. Drug Delivery Nanoparticles in Skin Cancers. Biomed Res J 2014; 2014 895986
[http://dx.doi.org/10.1155/2014/895986]
[9]
Akhter MH, Rizwanullah M, Ahmad J, Ahsan MJ, Mujtaba MA, Amin S. Nanocarriers in advanced drug targeting: setting novel paradigm in cancer therapeutics. Artif Cells Nanomed Biotechnol 2018; 46(5): 873-84.
[http://dx.doi.org/10.1080/21691401.2017.1366333 ] [PMID: 28830262]
[10]
Hejmady S, Singhvi G, Saha RN, Dubey SK. Regulatory aspects in process development and scale-up of nanopharmaceuticals. Ther Deliv 2020; 11(6): 341-3.
[http://dx.doi.org/10.4155/tde-2020-0034 ] [PMID: 32349643]
[11]
Dubey SK, Pradyuth SK, Saha RN, et al. Application of photodynamic therapy drugs for management of glioma. J Porphyrins Pthalocyanins 2019; 23: 121-28.
[http://dx.doi.org/10.1142/S1088424619300192]
[12]
Orthaber K, Pristovnik M, Skok K, Perić B, Maver U. Skin cancer and its treatment: novel treatment approaches with emphasis on nanotechnology. J Nanomater 2017; 2017.
[13]
Simões MCF, Sousa JJS, Pais AACC. Skin cancer and new treatment perspectives: A review. Cancer Lett 2015; 357(1): 8-42.
[http://dx.doi.org/10.1155/2014/895986]
[14]
Zeb A, Arif ST, Malik M, et al. Potential of nanoparticulate carriers for improved drug delivery via skin. Int J Pharm 2019; 397(1-2): 164-72.
[http://dx.doi.org/10.1007/s40005-018-00418-8]
[15]
Naves LB, Dhand C, Venugopal JR, Rajamani L, Ramakrishna S, Almeida L. Nanotechnology for the treatment of melanoma skin cancer. Prog Biomater 2017; 6(1-2): 13-26.
[http://dx.doi.org/10.1007/s40204-017-0064-z ] [PMID: 28303522]
[16]
Bei D, Meng J, Youan BBC. Engineering nanomedicines for improved melanoma therapy: progress and promises. Nanomedicine (Lond) 2010; 5(9): 1385-99.
[http://dx.doi.org/10.2217/nnm.10.117 ] [PMID: 21128721]
[17]
Simões MCF, Sousa JJS, Pais AACC. Skin cancer and new treatment perspectives: a review. Cancer Lett 2015; 357(1): 8-42.
[http://dx.doi.org/10.1016/j.canlet.2014.11.001 ] [PMID: 25444899]
[18]
Tran MA, Watts RJ, Robertson GP. Use of liposomes as drug delivery vehicles for treatment of melanoma. Pigment Cell Melanoma Res 2009; 22(4): 388-99.
[http://dx.doi.org/10.1111/j.1755-148X.2009.00581.x ] [PMID: 19493316]
[19]
de Leeuw J, de Vijlder HC, Bjerring P, Neumann HAM. Liposomes in dermatology today. J Eur Acad Dermatol Venereol 2009; 23(5): 505-16.
[http://dx.doi.org/10.1111/j.1468-3083.2009.03100.x ] [PMID: 19175703]
[20]
El Maghraby GM, Barry BW, Williams AC. Liposomes and skin: from drug delivery to model membranes. Eur J Pharm Sci 2008; 34(4-5): 203-22.
[http://dx.doi.org/10.1016/j.ejps.2008.05.002 ] [PMID: 18572392]
[21]
Slingerland M, Guchelaar HJ, Gelderblom H. Liposomal drug formulations in cancer therapy: 15 years along the road. Drug Discov Today 2012; 17(3-4): 160-6.
[http://dx.doi.org/10.1016/j.drudis.2011.09.015 ] [PMID: 21983329]
[22]
Mohan A, Narayanan S, Sethuraman S, Krishnan UM. Novel resveratrol and 5-fluorouracil coencapsulated in PEGylated nanoliposomes improve chemotherapeutic efficacy of combination against head and neck squamous cell carcinoma. Biomed Res Int 2014; 2014: 424239
[23]
Singh S. Liposome encapsulation of doxorubicin and celecoxib in combination inhibits progression of human skin cancer cells. Int J Nanomedicine 2018; 13(T-NANO 2014 Abstracts): 11-3.
[http://dx.doi.org/10.2147/IJN.S124701] [PMID: 29593389]
[24]
Hussain A, Samad A, Ramzan M, Ahsan MN, Ur Rehman Z, Ahmad FJ. Elastic liposome-based gel for topical delivery of 5-fluorouracil: in vitro and in vivo investigation. Drug Deliv 2016; 23(4): 1115-29.
[http://dx.doi.org/10.3109/10717544.2014.976891 ] [PMID: 25379805]
[25]
Rai S, Pandey V, Rai G. Transfersomes as versatile and flexible nano-vesicular carriers in skin cancer therapy: the state of the art. Nano Rev Exp 2017; 8(1): 1325708
[http://dx.doi.org/10.1080/20022727.2017.1325708 ] [PMID: 30410704]
[26]
Benson HA. Transfersomes for transdermal drug delivery. Expert Opin Drug Deliv 2006; 3(6): 727-37.
[http://dx.doi.org/10.1517/17425247.3.6.727 ] [PMID: 17076595]
[27]
Jiang T, Wang T, Li T, et al. Enhanced transdermal drug delivery by transfersome-embedded oligopeptide hydrogel for topical chemotherapy of melanoma. ACS Nano 2018; 12(10): 9693-701.
[http://dx.doi.org/10.1021/acsnano.8b03800 ] [PMID: 30183253]
[28]
El Zaafarany GM, Awad GAS, Holayel SM, Mortada ND. Role of edge activators and surface charge in developing ultradeformable vesicles with enhanced skin delivery. Int J Pharm 2010; 397(1-2): 164-72.
[http://dx.doi.org/10.1016/j.ijpharm.2010.06.034 ] [PMID: 20599487]
[29]
Zhang N, Yin Y, Xu SJ, Chen WS. 5-Fluorouracil: mechanisms of resistance and reversal strategies. Molecules 2008; 13(8): 1551-69.
[http://dx.doi.org/10.3390/molecules13081551 ] [PMID: 18794772]
[30]
Khan MA, Pandit J, Sultana Y, et al. Novel carbopol-based transfersomal gel of 5-fluorouracil for skin cancer treatment: in vitro characterization and in vivo study. Drug Deliv 2015; 22(6): 795-802.
[http://dx.doi.org/10.3109/10717544.2014.902146 ] [PMID: 24735246]
[31]
Alvi IA, Madan J, Kaushik D, Sardana S, Pandey RS, Ali A. Comparative study of transfersomes, liposomes, and niosomes for topical delivery of 5-fluorouracil to skin cancer cells: preparation, characterization, in-vitro release, and cytotoxicity analysis. Anticancer Drugs 2011; 22(8): 774-82.
[http://dx.doi.org/10.1097/CAD.0b013e328346c7d6 ] [PMID: 21799471]
[32]
Elsayed MMA, Abdallah OY, Naggar VF, Khalafallah NM. Deformable liposomes and ethosomes: mechanism of enhanced skin delivery. Int J Pharm 2006; 322(1-2): 60-6.
[http://dx.doi.org/10.1016/j.ijpharm.2006.05.027 ] [PMID: 16806755]
[33]
Oh EK, Jin SE, Kim JK, Park JS, Park Y, Kim CK. Retained topical delivery of 5-aminolevulinic acid using cationic ultradeformable liposomes for photodynamic therapy. Eur J Pharm Sci 2011; 44(1-2): 149-57.
[http://dx.doi.org/10.1016/j.ejps.2011.07.003 ] [PMID: 21782942]
[34]
Raahulan S, Sanapalli BKR, Karri VVSR. Paclitaxel loaded transfersomal vesicular drug delivery for the treatment of melanoma skin cancers. Int J Res Pharm Sci 2019; 10(4): 2891-7.
[35]
Teskač K, Kristl J. The evidence for solid lipid nanoparticles mediated cell uptake of resveratrol. Int J Pharm 2010; 390(1): 61-9.
[http://dx.doi.org/10.1016/j.ijpharm.2009.10.011 ] [PMID: 19833178]
[36]
Shah KA, Date AA, Joshi MD, Patravale VB. Solid lipid nanoparticles (SLN) of tretinoin: potential in topical delivery. Int J Pharm 2007; 345(1-2): 163-71.
[http://dx.doi.org/10.1016/j.ijpharm.2007.05.061 ] [PMID: 17644288]
[37]
Ourique AF, Melero A, de Bona da Silva C, et al. Improved photostability and reduced skin permeation of tretinoin: development of a semisolid nanomedicine. Eur J Pharm Biopharm 2011; 79(1): 95-101.
[http://dx.doi.org/10.1016/j.ejpb.2011.03.008 ] [PMID: 21402157]
[38]
Severino P, Fangueiro JF, Ferreira SV, et al. Nanoemulsions and nanoparticles for non-melanoma skin cancer: effects of lipid materials. Clin Transl Oncol 2013; 15(6): 417-24.
[http://dx.doi.org/10.1007/s12094-012-0982-0 ] [PMID: 23344664]
[39]
Tran MA, Watts RJ, Robertson GP. Use of liposomes as drug delivery vehicles for treatment of melanoma. Pigment Cell and Melanoma Research. NIH Public Access 22: 388-99.
[40]
Kogan A, Garti N. Microemulsions as transdermal drug delivery vehicles. Adv Colloid Interface Sci 2006; 123-126: 369-85.
[http://dx.doi.org/10.1016/j.cis.2006.05.014 ] [PMID: 16843424]
[41]
Pinheiro AC, Coimbra MA, Vicente AA. In vitro behaviour of curcumin nanoemulsions stabilized by biopolymer emulsifiers – Effect of interfacial composition. Food Hydrocoll 2016; 52: 460-7.
[http://dx.doi.org/10.1016/j.foodhyd.2015.07.025]
[42]
Gurdag S, Khandare J, Stapels S, Matherly LH, Kannan RM. Activity of dendrimer-methotrexate conjugates on methotrexate-sensitive and -resistant cell lines. Bioconjug Chem 2006; 17(2): 275-83.
[http://dx.doi.org/10.1021/bc0501855 ] [PMID: 16536456]
[43]
Coimbra M, Rijcken CJF, Stigter M, Hennink WE, Storm G, Schiffelers RM. Antitumor efficacy of dexamethasone-loaded core-crosslinked polymeric micelles. J Control Release 2012; 163(3): 361-7.
[http://dx.doi.org/10.1016/j.jconrel.2012.09.014 ] [PMID: 23041274]
[44]
Küchler S, Herrmann W, Panek-Minkin G, et al. SLN for topical application in skin diseases--characterization of drug-carrier and carrier-target interactions. Int J Pharm 2010; 390(2): 225-33.
[http://dx.doi.org/10.1016/j.ijpharm.2010.02.004 ] [PMID: 20153414]
[45]
Agrawal M, Saraf S, Saraf S, et al. Recent strategies and advances in the fabrication of nano lipid carriers and their application towards brain targeting. J Controlled Rel 2020; 321: 372-415.
[http://dx.doi.org/10.1016/j.jconrel.2020.02.020]
[46]
Desai P, Patlolla RR, Singh M. Interaction of nanoparticles and cell-penetrating peptides with skin for transdermal drug delivery. Mol Membr Biol 2010; 27(7): 247-59.
[http://dx.doi.org/10.3109/09687688.2010.522203 ] [PMID: 21028936]
[47]
Keck CM, Anantaworasakul P, Patel M, et al. A new concept for the treatment of atopic dermatitis: silver-nanolipid complex (sNLC). Int J Pharm 2014; 462(1-2): 44-51.
[http://dx.doi.org/10.1016/j.ijpharm.2013.12.044 ] [PMID: 24378329]
[48]
Zhai H, Maibach HI. Effects of skin occlusion on percutaneous absorption: an overview. Skin Pharmacol Appl Skin Physiol 2001; 14(1): 1-10.
[http://dx.doi.org/10.1159/000056328 ] [PMID: 11174085]
[49]
Iqbal MA, Md S, Sahni JK, Baboota S, Dang S, Ali J. Nanostructured lipid carriers system: recent advances in drug delivery. J Drug Target 2012; 20(10): 813-30.
[http://dx.doi.org/10.3109/1061186X.2012.716845 ] [PMID: 22931500]
[50]
Schneider M, Stracke F, Hansen S, Schaefer UF. Nanoparticles and their interactions with the dermal barrier. Dermatoendocrinol 2009; 1(4): 197-206.
[http://dx.doi.org/10.4161/derm.1.4.9501 ] [PMID: 20592791]
[51]
Loo Ch, Basri M, Ismail R, et al. Effect of compositions in nanostructured lipid carriers (NLC) on skin hydration and occlusion. Int J Nanomedicine 2013; 8: 13-22.
[PMID: 23293516]
[52]
N. D. Nanostructure lipid carriers: A promising tool for the drug delivery in the treatment of skin cancer. Asian J Pharm Clin Res 2019; 12(5): 15-26.
[53]
Bharadwaj R, Das PJ, Pal P, Mazumder B. Topical delivery of paclitaxel for treatment of skin cancer. Drug Dev Ind Pharm 2016; 42(9): 1482-94.
[http://dx.doi.org/10.3109/03639045.2016.1151028 ] [PMID: 26850463]
[54]
Khallaf RA, Salem HF, Abdelbary A. 5-Fluorouracil shell-enriched solid lipid nanoparticles (SLN) for effective skin carcinoma treatment. Drug Deliv 2016; 23(9): 3452-60.
[http://dx.doi.org/10.1080/10717544.2016.1194498 ] [PMID: 27240935]
[55]
Geetha T, Kapila M, Prakash O, Deol PK, Kakkar V, Kaur IP. Sesamol-loaded solid lipid nanoparticles for treatment of skin cancer. J Drug Target 2015; 23(2): 159-69.
[http://dx.doi.org/10.3109/1061186X.2014.965717 ] [PMID: 25268273]
[56]
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(4): 372-80.
[http://dx.doi.org/10.1080/02652048.2016.1200150 ] [PMID: 27338131]
[57]
Rajinikanth PS, Chellian J. Development and evaluation of nanostructured lipid carrier-based hydrogel for topical delivery of 5-fluorouracil. Int J Nanomedicine 2016; 11: 5067-77.
[http://dx.doi.org/10.2147/IJN.S117511 ] [PMID: 27785014]
[58]
Qidwai A, Khan S, Md S, et al. Nanostructured lipid carrier in photodynamic therapy for the treatment of basal-cell carcinoma. Drug Deliv 2016; 23(4): 1476-85.
[http://dx.doi.org/10.3109/10717544.2016.1165310 ] [PMID: 26978275]
[59]
Nastiti CMRR, Ponto T, Abd E, Grice JE, Benson HAE, Roberts MS. Topical nano and microemulsions for skin delivery. Pharmaceutics 2017; 9(4): 1-25.
[http://dx.doi.org/10.3390/pharmaceutics9040037 ] [PMID: 28934172]
[60]
Shaker DS, Ishak RAH, Ghoneim A, Elhuoni MA. Nanoemulsion: a review on mechanisms for the transdermal delivery of hydrophobic and hydrophilic drugs. Sci Pharm 2019; 87(3): 17.
[http://dx.doi.org/10.3390/scipharm87030017]
[61]
Gorain B, Choudhury H, Nair AB, Dubey SK, Kesharwani P. Theranostic application of nanoemulsions in chemotherapy Drug Discovery Today. Elsevier Ltd 2020.
[62]
Rai VK, Mishra N, Yadav KS, Yadav NP. Nanoemulsion as pharmaceutical carrier for dermal and transdermal drug delivery: Formulation development, stability issues, basic considerations and applications. J Controlled Rel 2018; 270: 203-5.
[63]
Singh Y, Meher JG, Raval K, et al. Nanoemulsion: Concepts, development and applications in drug delivery. J Controlled Rel 2017; 252: 28-49.
[64]
Shakeel F, Haq N, Al-Dhfyan A, Alanazi FK, Alsarra IA. Chemoprevention of skin cancer using low HLB surfactant nanoemulsion of 5-fluorouracil: a preliminary study. Drug Deliv 2015; 22(4): 573-80.
[http://dx.doi.org/10.3109/10717544.2013.868557 ] [PMID: 24350612]
[65]
Salmivuori M, Grönroos M, Tani T, et al. Hexyl aminolevulinate, 5-aminolevulinic acid nanoemulsion, and methyl aminolevulinate in photodynamic therapy of non-aggressive basal cell carcinomas: A non-sponsored, randomized, prospective and double-blinded trial. J Eur Acad Dermatol Venereol 2020. Online ahead of print.
[http://dx.doi.org/10.1111/jdv.16357] [PMID: 32196772]
[66]
Molaei MJ. Carbon quantum dots and their biomedical and therapeutic applications: A review. RSC Advances 2019; 9(12): 6460-81.
[http://dx.doi.org/10.1039/C8RA08088G]
[67]
Wang Y, Zhu Y, Yu S, Jiang C. Fluorescent carbon dots: Rational synthesis, tunable optical properties and analytical applications. RSC Advances 2017; 7(65): 40973-89.
[http://dx.doi.org/10.1039/C7RA07573A]
[68]
Singh S, Mishra A, Kumari R, Sinha KK, Singh MK, Das P. Carbon dots assisted formation of DNA hydrogel for sustained release of drug. Carbon N Y 2017; 114: 169-76.
[http://dx.doi.org/10.1016/j.carbon.2016.12.020]
[69]
Sharma A, Das J. Small molecules derived carbon dots: synthesis and applications in sensing, catalysis, imaging, and biomedicine. J Nanobiotechnology 2019; 17(1): 92.
[http://dx.doi.org/10.1186/s12951-019-0525-8 ] [PMID: 31451110]
[70]
Beack S, Kong WH, Jung HS, et al. Photodynamic therapy of melanoma skin cancer using carbon dot - chlorin e6 - hyaluronate conjugate. Acta Biomater 2015; 26: 295-305.
[http://dx.doi.org/10.1016/j.actbio.2015.08.027 ] [PMID: 26297888]
[71]
Xu N, Du J, Yao Q, et al. Precise photodynamic therapy: Penetrating the nuclear envelope with photosensitive carbon dots. Carbon N Y 2020; 159: 74-82.
[http://dx.doi.org/10.1016/j.carbon.2019.12.002]
[72]
Li Q, Zhou R, Xie Y, Li Y, Chen Y, Cai X. Sulphur-doped carbon dots as a highly efficient nano-photodynamic agent against oral squamous cell carcinoma. Cell Prolif 2020; 53(4) e12786
[http://dx.doi.org/10.1111/cpr.12786 ] [PMID: 32301195]
[73]
Aguilar Cosme JR, Bryant HE, Claeyssens F. Carbon dot-protoporphyrin IX conjugates for improved drug delivery and bioimaging. PLoS One 2019; 14(7) e0220210
[http://dx.doi.org/10.1371/journal.pone.0220210 ] [PMID: 31344086]
[74]
Li S, Luo J, Yang X, Wan Y, Liu C. A novel immunosensor for squamous cell carcinoma antigen determination based on CdTe@Carbon dots nanocomposite electrochemiluminescence resonance energy transfer. Sens Actuators B Chem 2014; 197: 43-9.
[http://dx.doi.org/10.1016/j.snb.2014.02.066]
[75]
Kuche K, Maheshwari R, Tambe V, et al. Carbon nanotubes (CNTs) based advanced dermal therapeutics: Current trends and future potential. In: Nanoscale 2018; 8911-37.
[76]
Shin SR, Jung SM, Zalabany M, et al. Carbon-nanotube-embedded hydrogel sheets for engineering cardiac constructs and bioactuators. ACS Nano 2013; 7(3): 2369-80.
[http://dx.doi.org/10.1021/nn305559j ] [PMID: 23363247]
[77]
Shi J, Ma R, Wang L, et al. The application of hyaluronic acid-derivatized carbon nanotubes in hematoporphyrin monomethyl ether-based photodynamic therapy for in vivo and in vitro cancer treatment. Int J Nanomedicine 2013; 8: 2361-73.
[http://dx.doi.org/10.2147/IJN.S45407 ] [PMID: 23843694]
[78]
Siu KS, Chen D, Zheng X, et al. Non-covalently functionalized single-walled carbon nanotube for topical siRNA delivery into melanoma. Biomaterials 2014; 35(10): 3435-42.
[http://dx.doi.org/10.1016/j.biomaterials.2013.12.079 ] [PMID: 24424208]
[79]
Albert K, Hsu HY. Carbon-based materials for photo-triggered theranostic applications. Molecules 2016; 21(11): 1-28.
[http://dx.doi.org/10.3390/molecules21111585 ] [PMID: 27879628]
[80]
Samanta PN, Das KK. 5-Aminolevulinic acid functionalized boron nitride and carbon nanotubes as drug delivery vehicles for skin anticancer drugs: A theoretical study. RSC Advances 2016; 6(95): 92547-59.
[http://dx.doi.org/10.1039/C6RA14693G]
[81]
Dave K, Krishna Venuganti VV. Dendritic polymers for dermal drug delivery. Ther Deliv 2017; 8(12): 1077-96.
[http://dx.doi.org/10.4155/tde-2017-0091 ] [PMID: 29125060]
[82]
Shukla R, Singh A, Pardhi V, et al. Dendrimer-based nanoparticulate delivery system for cancer therapy. In: Polymeric Nanoparticles as a Promising Tool for Anti-cancer Therapeutics.Elsevier Inc. 2019; 233-55.
[http://dx.doi.org/10.1016/B978-0-12-816963-6.00011-X]
[83]
Ioannidis JPA. Reproducible pharmacokinetics. J Pharmacokinet Pharmacodyn 2019; 46(2): 111-6.
[http://dx.doi.org/10.1007/s10928-019-09621-y ] [PMID: 31004315]
[84]
Noriega-Luna B, Godínez LA, Rodríguez FJ, et al. Applications of dendrimers in drug delivery agents, diagnosis, therapy, and detection. J Nanomater 2014; 2014.
[85]
Dubey SK, Salunkhe S, Agrawal M, et al. Understanding the pharmaceutical aspects of dendrimers for the delivery of anticancer drugs. Curr Drug Targets 2020; 21(6): 528-40.
[http://dx.doi.org/10.2174/1389450120666191031092259 ] [PMID: 31670619]
[86]
Sun M, Fan A, Wang Z, Zhao Y. Dendrimer-mediated drug delivery to the skin. Soft Matter 2012; 8(16): 4301-5.
[http://dx.doi.org/10.1039/c2sm07280g]
[87]
Parhi R, Swain S. Transdermal evaporation drug delivery system: Concept to commercial products. Adv Pharm Bull 2018; 8(4): 535-50.
[http://dx.doi.org/10.15171/apb.2018.063 ] [PMID: 30607327]
[88]
Chauhan AS, Sridevi S, Chalasani KB, et al. Dendrimer-mediated transdermal delivery: enhanced bioavailability of indomethacin. J Control Release 2003; 90(3): 335-43.
[http://dx.doi.org/10.1016/S0168-3659(03)00200-1 ] [PMID: 12880700]
[89]
Rodriguez L, Vallecorsa P, Battah S, et al. Aminolevulinic acid dendrimers in photodynamic treatment of cancer and atheromatous disease. Photochem Photobiol Sci 2015; 14(9): 1617-27.
[http://dx.doi.org/10.1039/C5PP00126A ] [PMID: 26066768]
[90]
Cline EN, Li MH, Choi SK, et al. Paclitaxel-conjugated PAMAM dendrimers adversely affect microtubule structure through two independent modes of action. Biomacromolecules 2013; 14(3): 654-64.
[http://dx.doi.org/10.1021/bm301719b ] [PMID: 23391096]
[91]
Mutalik S, Shetty PK, Kumar A, Kalra R, Parekh HS. Enhancement in deposition and permeation of 5-fluorouracil through human epidermis assisted by peptide dendrimers. Drug Deliv 2014; 21(1): 44-54.
[http://dx.doi.org/10.3109/10717544.2013.845861 ] [PMID: 24134794]
[92]
Venuganti VVK, Saraswathy M, Dwivedi C, Kaushik RS, Perumal OP. Topical gene silencing by iontophoretic delivery of an antisense oligonucleotide-dendrimer nanocomplex: the proof of concept in a skin cancer mouse model. Nanoscale 2015; 7(9): 3903-14.
[http://dx.doi.org/10.1039/C4NR05241B ] [PMID: 25436837]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy