Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

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

Mini-Review Article

Colloidal Nanocarriers as Versatile Targeted Delivery Systems for Cervical Cancer

Author(s): Abimanyu Sugumaran* and Vishali Mathialagan

Volume 26, Issue 40, 2020

Page: [5174 - 5187] Pages: 14

DOI: 10.2174/1381612826666200625110950

Price: $65

conference banner
Abstract

Background: The second most common malignant cancer of the uterus is cervical cancer, which is present worldwide, has a rising death rate and is predominant in developing countries. Different classes of anticancer agents are used to treat cervical carcinoma. The use of these agents results in severe untoward side-effects, toxicity, and multidrug resistance (MDR) with higher chances of recurrence and spread beyond the pelvic region. Moreover, the resulting clinical outcome remains very poor even after surgical procedures and treatment with conventional chemotherapy. Because of the nonspecificity of their use, the agents wipe out both cancerous and normal tissues. Colloidal nano dispersions have now been focusing on site-specific delivery for cervical cancer, and there has been much advancement.

Methods: This review aims to highlight the problems in the current treatment of cervical cancer and explore the potential of colloidal nanocarriers for selective delivery of anticancer drugs using available literature.

Results: In this study, we surveyed the role and potential of different colloidal nanocarriers in cervical cancer, such as nanoemulsion, nanodispersions, polymeric nanoparticles, and metallic nanoparticles and photothermal and photodynamic therapy. We found significant advancement in colloidal nanocarrier-based cervical cancer treatment.

Conclusion: Cervical cancer-targeted treatment with colloidal nanocarriers would hopefully result in minimal toxic side effects, reduced dosage frequency, and lower MDR incidence and enhance the patient survival rates. The future direction of the study should be focused more on the regulatory barrier of nanocarriers based on clinical outcomes for cervical cancer targeting with cost-effective analysis.

Keywords: Colloidal nanocarrier, cervical cancer targeting, nanoparticles, drug delivery system, polymeric nanoparticles, nanoencapsulation.

[1]
Ali CI, Makata NE. Cervical cancer: a health limiting condition. Gynecol Obstet (Sunnyvale) 2016; 61000378
[2]
Ren G, Zhao YP, Yang L, Fu CX. Anti-proliferative effect of clitocine from the mushroom Leucopaxillus giganteus on human cervical cancer HeLa cells by inducing apoptosis. Cancer Lett 2008; 262(2): 190-200.
[http://dx.doi.org/10.1016/j.canlet.2007.12.013] [PMID: 18222036]
[3]
Clarke MA, Wentzensen N, Mirabello L, et al. Human papillomavirus DNA methylation as a potential biomarker for cervical cancer. Cancer Epidemiol Biomarkers Prev 2012; 21(12): 2125-37.
[http://dx.doi.org/10.1158/1055-9965.EPI-12-0905] [PMID: 23035178]
[4]
Alsbeih G. HPV infection in cervical and other cancers in Saudi Arabia: implication for prevention and vaccination. Front Oncol 2014; 4: 65.
[http://dx.doi.org/10.3389/fonc.2014.00065] [PMID: 24744990]
[5]
Roden R, Wu TC. How will HPV vaccines affect cervical cancer? Nat Rev Cancer 2006; 6(10): 753-63.
[http://dx.doi.org/10.1038/nrc1973] [PMID: 16990853]
[6]
Rodríguez AC, Schiffman M, Herrero R, et al. Rapid clearance of human papillomavirus and implications for clinical focus on persistent infections. J Natl Cancer Inst 2008; 100(7): 513-7.
[http://dx.doi.org/10.1093/jnci/djn044] [PMID: 18364507]
[7]
Jaisamrarn U, Castellsagué X, Garland SM, et al. Natural history of progression of HPV infection to cervical lesion or clearance: Analysis of the control arm of the large, randomised PATRICIA study. PLoS One 2013; 8(11)e79260
[http://dx.doi.org/10.1371/journal.pone.0079260] [PMID: 24260180]
[8]
Bergman H, Buckley BS, Villanueva G, et al. Comparison of different human papillomavirus (HPV) vaccine types and dose schedules for prevention of HPV-related disease in females and males. Cochrane Database Syst Rev 2019; 2019(11)CD013479
[http://dx.doi.org/10.1002/14651858.CD013479] [PMID: 31755549]
[9]
Lorusso D, Petrelli F, Coinu A, Raspagliesi F, Barni S. A systematic review comparing cisplatin and carboplatin plus paclitaxel-based chemotherapy for recurrent or metastatic cervical cancer. Gynecol Oncol 2014; 133(1): 117-23.
[http://dx.doi.org/10.1016/j.ygyno.2014.01.042] [PMID: 24486604]
[10]
Kang YJ, O’Connell DL, Tan J, et al. Optimal uptake rates for initial treatments for cervical cancer in concordance with guidelines in Australia and Canada: Results from two large cancer facilities. Cancer Epidemiol 2015; 39(4): 600-11.
[http://dx.doi.org/10.1016/j.canep.2015.04.009] [PMID: 26004990]
[11]
Yadav N, Parveen S, Banerjee M. Potential of nano-phytochemicals in cervical cancer therapy. Clin Chim Acta 2020; 505: 60-72.
[http://dx.doi.org/10.1016/j.cca.2020.01.035] [PMID: 32017926]
[12]
Sims LB, Curry KC, Parupalli S, Horner G, Frieboes HB. Steinbach-rankins jm. efficacy of surface-modified plga nanoparticles as a function of cervical cancer type. Pharm Res 2019; 36(5): 66.
[http://dx.doi.org/10.1007/s11095-019-2602-y] [PMID: 30868271]
[13]
Vamanu E, Ene M, Biță B, Ionescu C, Crăciun L, Sârbu I. In Vitro Human microbiota response to exposure to silver nanoparticles biosynthesized with mushroom extract. Nutrients 2018; 10(5): 607.
[http://dx.doi.org/10.3390/nu10050607] [PMID: 29757931]
[14]
Yuan YG, Zhang S, Hwang JY, Kong IK. Silver nanoparticles potentiates cytotoxicity and apoptotic potential of camptothecin in human cervical cancer cells. Oxid Med Cell Longev 2018; 20186121328
[http://dx.doi.org/10.1155/2018/6121328] [PMID: 30647812]
[15]
Arayne MS, Sultana N. Review: nanoparticles in drug delivery for the treatment of cancer. Pak J Pharm Sci 2006; 19(3): 258-68.
[PMID: 16935836]
[16]
Bulbake U, Doppalapudi S, Kommineni N, Khan W. Liposomal formulations in clinical use: An updated review. Pharmaceutics 2017; 9(2): 12.
[http://dx.doi.org/10.3390/pharmaceutics9020012] [PMID: 28346375]
[17]
Riggio C, Pagni E, Raffa V, Cuschieri A. Nano-oncology: Clinical application for cancer therapy and future perspectives. J Nanomater 2011; 2011164506
[http://dx.doi.org/10.1155/2011/164506]
[18]
Gupta S, Gupta MK. Possible role of nanocarriers in drug delivery against cervical cancer. Nano Rev Exp 2017; 8(1)1335567
[http://dx.doi.org/10.1080/20022727.2017.1335567] [PMID: 30410707]
[19]
Ordikhani F, Erdem Arslan M, Marcelo R, et al. Drug delivery approaches for the treatment of cervical cancer. Pharmaceutics 2016; 8(3): 23.
[http://dx.doi.org/10.3390/pharmaceutics8030023] [PMID: 27447664]
[20]
de la Harpe KM, Kondiah PPD, Choonara YE, Marimuthu T, du Toit LC, Pillay V. The hemocompatibility of nanoparticles: A review of cell-nanoparticle interactions and hemostasis. Cells 2019; 8(10): 1209.
[http://dx.doi.org/10.3390/cells8101209] [PMID: 31591302]
[21]
Ma Y, Zheng Y, Liu K, et al. Nanoparticles of Poly(lactide-coglycolide)-d-a-tocopheryl polyethylene glycol 1000 succinate randomcopolymer for cancer treatment. Nanoscale Res Lett 2010; 5(7): 1161-9.
[http://dx.doi.org/10.1007/s11671-010-9620-3] [PMID: 20596457]
[22]
Seyyedi N, Farjadian F, Farhadi A, et al. A high yield gold nanoparticle-based DNA isolation method for human papillomaviruses genotypes from cervical cancer tissue samples. Beilstein Arch 2019; 1: 106.
[23]
Murugesan K, Koroth J, Srinivasan PP, et al. Effects of green synthesised silver nanoparticles (ST06-AgNPs) using curcumin derivative (ST06) on human cervical cancer cells (HeLa) in vitro and EAC tumor bearing mice models. Int J Nanomedicine 2019; 14: 5257-70.
[http://dx.doi.org/10.2147/IJN.S202404] [PMID: 31409988]
[24]
Sarkar S, Kotteeswaran V. Green synthesis of silver nanoparticles from aqueous leaf extract of Pomegranate (Punica granatum) and their anticancer activity on human cervical cancer cells. Advances in Natural Sciences: Nanoscience and Nanotechnology 2018; 9(2)025014
[http://dx.doi.org/10.1088/2043-6254/aac590]
[25]
Bhatnagar I, Venkatesan J, Kiml SK. Polymer functionalized single walled carbon nanotubes mediated drug delivery of gliotoxin in cancer cells. J Biomed Nanotechnol 2014; 10(1): 120-30.
[http://dx.doi.org/10.1166/jbn.2014.1677] [PMID: 24724504]
[26]
Botasini S, Méndez E. Silver nanoparticle aggregation not triggered by an ionic strength mechanism. J Nanopart Res 2013; 15: 1526.
[http://dx.doi.org/10.1007/s11051-013-1526-4]
[27]
Banerjee SL, Khamrai M, Sarkar K, Singha NK, Kundu PP. Modified chitosan encapsulated core-shell Ag Nps for superior antimicrobial and anticancer activity. Int J Biol Macromol 2016; 85: 157-67.
[http://dx.doi.org/10.1016/j.ijbiomac.2015.12.068] [PMID: 26724687]
[28]
Tran TH, Nguyen CT, Gonzalez-Fajardo L, et al. Long circulating self-assembled nanoparticles from cholesterol-containing brush-like block copolymers for improved drug delivery to tumors. Biomacromolecules 2014; 15(11): 4363-75.
[http://dx.doi.org/10.1021/bm5013822] [PMID: 25310277]
[29]
Byagari K, Shanavas A, Rengan AK, et al. Biocompatible amphiphilic pentablockcopolymeric nanoparticles for anti-cancer drug delivery. J Biomed Nanotechnol 2014; 10(1): 109-19.
[30]
Wang Z, Zeng X, Ma Y, et al. Antitumor efficiency of D-alpha-tocopheryl polyethylene glycol 1000 succinate-b-poly(epsilon-caprolactone-ran-lactide) nanoparticle-based delivery of docetaxel in mice bearing cervical cancer. J Biomed Nanotechnol 2014; 10(8): 1509-19.
[http://dx.doi.org/10.1166/jbn.2014.1844] [PMID: 25016651]
[31]
Zeng X, Tao W, Mei L, Huang L, Tan C, Feng SS. Cholic acid-functionalized nanoparticles of star-shaped PLGA-vitamin E TPGS copolymer for docetaxel delivery to cervical cancer. Biomaterials 2013; 34(25): 6058-67.
[http://dx.doi.org/10.1016/j.biomaterials.2013.04.052] [PMID: 23694904]
[32]
Ekladious I, Colson YL, Grinstaff MW. Polymer-drug conjugate therapeutics: advances, insights and prospects. Nat Rev Drug Discov 2019; 18(4): 273-94.
[http://dx.doi.org/10.1038/s41573-018-0005-0] [PMID: 30542076]
[33]
Ma Y, Huang L, Song C, et al. Nanoparticle formulation of poly(ɛ-caprolactone-co-lactide)-d-α-tocopheryl polyethylene glycol 1000 succinate random copolymer for cervical cancer treatment. Int J Nanomedicine 2010; 51: 5952-9.
[34]
Yang H, Li K, Liu Y, Liu Z, Miyoshi H. Poly(D,L-lactide-co-glycolide) nanoparticles encapsulated fluorescent isothiocyanate and paclitaxol: preparation, release kinetics and anticancer effect. J Nanosci Nanotechnol 2009; 9(1): 282-7.
[http://dx.doi.org/10.1166/jnn.2009.J065] [PMID: 19441308]
[35]
Qiu B, Ji M, Song X, et al. Co-delivery of docetaxel and endostatin by a biodegradable nanoparticle for the synergistic treatment of cervical cancer. Nanoscale Res Lett 2012; 7(1): 666.
[http://dx.doi.org/10.1186/1556-276X-7-666] [PMID: 23216701]
[36]
Vivero-Escoto JL, Slowing II, Lin VS. Tuning the cellular uptake and cytotoxicity properties of oligonucleotide intercalator-functionalized mesoporous silica nanoparticles with human cervical cancer cells HeLa. Biomaterials 2010; 31(6): 1325-33.
[http://dx.doi.org/10.1016/j.biomaterials.2009.11.009] [PMID: 19932923]
[37]
Matthaiou EI, Barar J, Sandaltzopoulos R, Li C, Coukos G, Omidi Y. Shikonin-loaded antibody-armed nanoparticles for targeted therapy of ovarian cancer. Int J Nanomedicine 2014; 9: 1855-70.
[PMID: 24790428]
[38]
Mooguee M, Omidi Y, Davaran S. Synthesis and in vitro release of adriamycin from star-shaped poly(lactide-co-glycolide) nano- and microparticles. J Pharm Sci 2010; 99(8): 3389-97.
[http://dx.doi.org/10.1002/jps.22106] [PMID: 20229603]
[39]
Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. J Control Release 2012; 161(2): 505-22.
[http://dx.doi.org/10.1016/j.jconrel.2012.01.043] [PMID: 22353619]
[40]
Locatelli E, Franchini MC. Biodegradable PLGA-b-PEG polymeric nanoparticles: synthesis, properties, and nanomedical applications as drug delivery system. J Nanopart Res 2012; 14: 1-17.
[http://dx.doi.org/10.1007/s11051-012-1316-4]
[41]
Ohya Y, Takahashi A, Nagahama K. Biodegradable polymeric assemblies for biomedical materialspolymers in nanomedicine advanced in polymer science Springer 2011; 65-114.
[42]
Pandita D, Kumar S, Lather V. Hybrid poly (lactic-co-glycolic acid) nanoparticles: design and delivery prospectives. Drug Discov Today 2015; 20(1): 95-104.
[http://dx.doi.org/10.1016/j.drudis.2014.09.018] [PMID: 25277320]
[43]
Vllasaliu D, Fowler R, Stolnik S. PEGylated nanomedicines: recent progress and remaining concerns. Expert Opin Drug Deliv 2014; 11(1): 139-54.
[http://dx.doi.org/10.1517/17425247.2014.866651] [PMID: 24295065]
[44]
Turecek PL, Bossard MJ, Schoetens F, Ivens IA. PEGylation of biopharmaceuticals: a review of chemistry and nonclinical safety information of approved drugs. J Pharm Sci 2016; 105(2): 460-75.
[http://dx.doi.org/10.1016/j.xphs.2015.11.015] [PMID: 26869412]
[45]
Chauhan N, Maher DM, Hafeez BB, et al. Ormeloxifene nanotherapy for cervical cancer treatment. Int J Nanomedicine 2019; 14: 7107-21.
[http://dx.doi.org/10.2147/IJN.S200944] [PMID: 31564868]
[46]
Hatami E, Nagesh PKB, Chowdhury P, et al. Tannic acid-lung fluid assemblies promote interaction and delivery of drugs to lung cancer cells. Pharmaceutics 2018; 10(3)E111
[http://dx.doi.org/10.3390/pharmaceutics10030111] [PMID: 30071698]
[47]
Kim BS, Kim CS, Lee KM. The intracellular uptake ability of chitosan-coated Poly (D,L-lactide-co-glycolide) nanoparticles. Arch Pharm Res 2008; 31(8): 1050-4.
[http://dx.doi.org/10.1007/s12272-001-1267-5] [PMID: 18787796]
[48]
Zhang C, Zhang Z, Zhao L. Folate-decorated poly(3-hydroxybutyrate-co-3-hydroxyoctanoate) nanoparticles for targeting delivery: optimization and in vivo antitumor activity. Drug Deliv 2016; 23(5): 1830-7.
[http://dx.doi.org/10.3109/10717544.2015.1122675] [PMID: 26652055]
[49]
Fonseca C, Simões S, Gaspar R. Paclitaxel-loaded PLGA nanoparticles: Preparation, physicochemical characterization and in vitro anti-tumoral activity. J Control Release 2002; 83(2): 273-86.
[http://dx.doi.org/10.1016/S0168-3659(02)00212-2] [PMID: 12363453]
[50]
Mu L, Feng SS. PLGA/TPGS nanoparticles for controlled release of paclitaxel: Effects of the emulsifier and drug loading ratio. Pharm Res 2003; 20(11): 1864-72.
[http://dx.doi.org/10.1023/B:PHAM.0000003387.15428.42] [PMID: 14661934]
[51]
Feng SS, Mu L, Win KY, Huang G. Nanoparticles of biodegradable polymers for clinical administration of paclitaxel. Curr Med Chem 2004; 11(4): 413-24.
[http://dx.doi.org/10.2174/0929867043455909] [PMID: 14965222]
[52]
Khin YW, Feng SS. In vitro and in vivo studies on vitamin E TPGS-emulsified poly(D,L-lactic-co-glycolic acid) nanoparticles for clinical administration of paclitaxel. Biomaterials 2006; 27: 2285-91.
[http://dx.doi.org/10.1016/j.biomaterials.2005.11.008] [PMID: 16313953]
[53]
Liggins RT, D’Amours S, Demetrick JS, Machan LS, Burt HM. Paclitaxel loaded poly(L-lactic acid) microspheres for the prevention of intraperitoneal carcinomatosis after a surgical repair and tumor cell spill. Biomaterials 2000; 21(19): 1959-69.
[http://dx.doi.org/10.1016/S0142-9612(00)00080-6] [PMID: 10941917]
[54]
Xu Q, Ensign LM, Boylan NJ, et al. Impact of surface Polyethylene Glycol (PEG) density on biodegradable nanoparticle transport in mucus ex vivo and distribution in vivo. ACS Nano 2015; 9(9): 9217-27.
[http://dx.doi.org/10.1021/acsnano.5b03876] [PMID: 26301576]
[55]
Zaman MS, Chauhan N, Yallapu MM, et al. Curcumin nanoformulation for cervical cancer treatment. Sci Rep 2016; 6: 20051.
[http://dx.doi.org/10.1038/srep20051] [PMID: 26837852]
[56]
Nagapoosanam AL, Ganesan N, Umapathy D, Moorthy RK, Arockiam AJV. Knockdown of human telomerase reverse transcriptase induces apoptosis in cervical cancer cell line. Indian J Med Res 2019; 149(3): 345-53.
[http://dx.doi.org/10.4103/ijmr.IJMR_1676_16] [PMID: 31249199]
[57]
Boondireke S, Léonard M, Durand A, Thanomsub Wongsatayanon B. Encapsulation of monomyristin into polymeric nanoparticles improved its in vitro antiproliferative activity against cervical cancer cells. Colloids Surf B Biointerfaces 2019; 176: 9-17.
[http://dx.doi.org/10.1016/j.colsurfb.2018.12.062] [PMID: 30590347]
[58]
Agnihotri SA, Mallikarjuna NN, Aminabhavi TM. Recent advances on chitosan-based micro- and nanoparticles in drug delivery. J Control Release 2004; 100(1): 5-28.
[http://dx.doi.org/10.1016/j.jconrel.2004.08.010] [PMID: 15491807]
[59]
Illum L. Chitosan and its use as a pharmaceutical excipient pharm res 1998; 15: 1326-31.
[60]
Highton AJ, Girardin A, Bell GM, Hook SM, Kemp RA. Chitosan gel vaccine protects against tumour growth in an intracaecal mouse model of cancer by modulating systemic immune responses. BMC Immunol 2016; 17(1): 39.
[http://dx.doi.org/10.1186/s12865-016-0178-4] [PMID: 27756214]
[61]
Strasser A. OConnor L, Dixit VM. Apoptosis signalling. Annu Rev Biochem 2000; 69: 217-45.
[http://dx.doi.org/10.1146/annurev.biochem.69.1.217] [PMID: 10966458]
[62]
Wu H, Zhang J. Chitosan-based zinc oxide nanoparticle for enhanced anticancer effect in cervical cancer: A physicochemical and biological perspective. Saudi Pharm J 2018; 26(2): 205-10.
[http://dx.doi.org/10.1016/j.jsps.2017.12.010] [PMID: 30166917]
[63]
Yang J, Li S, Guo F, Zhang W, Wang Y, Pan Y. Induction of apoptosis by chitosan/HPV16 E7 siRNA complexes in cervical cancer cells. Mol Med Rep 2013; 7(3): 998-1002.
[http://dx.doi.org/10.3892/mmr.2012.1246] [PMID: 23258711]
[64]
Saengkrit N, Sanitrum P, Woramongkolchai N, et al. The PEI-introduced CS shell/PMMA core nanoparticle for silencing the expression of E6/E7 oncogenes in human cervical cells. Carbohydr Polym 2012; 90(3): 1323-9.
[http://dx.doi.org/10.1016/j.carbpol.2012.06.079] [PMID: 22939347]
[65]
Jin C, Bai L, Wu H, Song W, Guo G, Dou K. Cytotoxicity of paclitaxel incorporated in PLGA nanoparticles on hypoxic human tumor cells. Pharm Res 2009; 26(7): 1776-84.
[http://dx.doi.org/10.1007/s11095-009-9889-z] [PMID: 19384463]
[66]
Umadevi SK, Thiruganesh R, Suresh S, Reddy KB. Formulation and evaluation of chitosan microspheres of aceclofenac for colon-targeted drug delivery. Biopharm Drug Dispos 2010; 31(7): 407-27.
[http://dx.doi.org/10.1002/bdd.722] [PMID: 20848388]
[67]
Wang JY, Wang Y, Meng X. Chitosan nanolayered cisplatin-loaded lipid nanoparticles for enhanced anticancer efficacy in cervical cancer. Nanoscale Res Lett 2016; 11(1): 524.
[http://dx.doi.org/10.1186/s11671-016-1698-9] [PMID: 27888498]
[68]
Ensign LM. Cone, Hanes J. Nanoparticle-based drug delivery to the vagina. J Control Release 2014; 190: 500-14.
[http://dx.doi.org/10.1016/j.jconrel.2014.04.033] [PMID: 24830303]
[69]
Rossi S, Vigani B, Puccio A, Bonferoni MC, Sandri G, Ferrari F. Chitosan ascorbate nanoparticles for the vaginal delivery of antibiotic drugs in atrophic vaginitis. Mar Drugs 2017; 15(10): 319.
[http://dx.doi.org/10.3390/md15100319] [PMID: 29048359]
[70]
Spadari CC, Lopes LB, Ishida K. Potential use of alginate-based carriers as antifungal delivery system. Front Microbiol 2017; 8: 97.
[http://dx.doi.org/10.3389/fmicb.2017.00097] [PMID: 28194145]
[71]
Vani CZ, Skalko Basnet N. Nanoformulations for vaginal therapy Nanotechnology applied to pharmaceutical technology. Biomedical and Life Sciences Springer 2017; pp. 183-221.
[http://dx.doi.org/10.1007/978-3-319-70299-5_8]
[72]
Das RK, Kasoju N, Bora U. Encapsulation of curcumin in alginate-chitosan-pluronic composite nanoparticles for delivery to cancer cells. Nanomedicine (Lond) 2010; 6(1): 153-60.
[http://dx.doi.org/10.1016/j.nano.2009.05.009] [PMID: 19616123]
[73]
Duncan R. Polymer conjugates as anticancer nanomedicines. Nat Rev Cancer 2006; 6(9): 688-701.
[http://dx.doi.org/10.1038/nrc1958] [PMID: 16900224]
[74]
Dario Rafael OH, Luis Fernándo ZG, Abraham PT, Pedro Alberto VL, Guadalupe GS, Pablo PJ. Production of chitosan-oligosaccharides by the chitin-hydrolytic system of Trichoderma harzianum and their antimicrobial and anticancer effects. Carbohydr Res 2019; 486107836
[http://dx.doi.org/10.1016/j.carres.2019.107836] [PMID: 31669568]
[75]
Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clin Pharmacol Ther 2008; 83(5): 761-9.
[http://dx.doi.org/10.1038/sj.clpt.6100400] [PMID: 17957183]
[76]
Li C, Wallace S. Polymer-drug conjugates: recent development in clinical oncology. Adv Drug Deliv Rev 2008; 60(8): 886-98.
[http://dx.doi.org/10.1016/j.addr.2007.11.009] [PMID: 18374448]
[77]
Wang CY, Looney DJ, Li ML, et al. Peptides: Synthesis, Structures, and Applications. Biomacromolecules 1991; 254: 285-8.
[78]
Oyewumi MO, Kumar A, Cui Z. Nano-microparticles as immune adjuvants: correlating particle sizes and the resultant immune responses. Expert Rev Vaccines 2010; 9(9): 1095-107.
[http://dx.doi.org/10.1586/erv.10.89] [PMID: 20822351]
[79]
Bachmann MF, Jennings GT. Therapeutic vaccines for chronic diseases: successes and technical challenges. Nat Rev Immunol 2010; 10: 787-96.
[http://dx.doi.org/10.1038/nri2868] [PMID: 20948547]
[80]
Bothun GD, Lelis A, Chen Y, et al. Multicomponent folate-targeted magnetoliposomes: design, characterization, and cellular uptake Nano med 2011; 1(7): 797-805.
[http://dx.doi.org/10.1016/j.nano.2011.02.007]
[81]
Yu G. Fabrication of a targeted drug delivery system from a Pillar[5]arene-based supramolecular diblock copolymeric amphiphile for effective cancer therapy. Adv Funct Mater 2016; 26: 8999-9008.
[http://dx.doi.org/10.1002/adfm.201601770]
[82]
Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J Control Release 2010; 148(2): 135-46.
[http://dx.doi.org/10.1016/j.jconrel.2010.08.027] [PMID: 20797419]
[83]
Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2007; 2(12): 751-60.
[http://dx.doi.org/10.1038/nnano.2007.387] [PMID: 18654426]
[84]
Kirpotin DB, Drummond DC, Shao Y, et al. Antibody targeting of long-circulating lipidic nanoparticles does not increase tumor localization but does increase internalization in animal models. Cancer Res 2006; 66(13): 6732-40.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-4199] [PMID: 16818648]
[85]
Byrne JD, Betancourt T, Brannon-Peppas L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev 2008; 60(15): 1615-26.
[http://dx.doi.org/10.1016/j.addr.2008.08.005] [PMID: 18840489]
[86]
Li B, Gao MH, Chu XM, Xu YJ, Yang F. Identification of a novel short peptide seal specific to CD59 and its effect on HeLa cell growth and apoptosis. Cell Oncol (Dordr) 2012; 35(5): 355-65.
[http://dx.doi.org/10.1007/s13402-012-0096-2] [PMID: 22945508]
[87]
Leung TH, Tang HW, Siu MK, et al. Human papillomavirus E6 protein enriches the CD55(+) population in cervical cancer cells, promoting radioresistance and cancer aggressiveness. J Pathol 2018; 244(2): 151-63.
[http://dx.doi.org/10.1002/path.4991] [PMID: 28944962]
[88]
Adams GP, Schier R, McCall AM, et al. High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules. Cancer Res 2001; 61(12): 4750-5.
[PMID: 11406547]
[89]
Gu FX, Karnik R, Wang AZ, et al. Targeted nanoparticles for cancer therapy. Nano Today 2007; 2: 14-21.
[http://dx.doi.org/10.1016/S1748-0132(07)70083-X]
[90]
Waldmann TA. Immunotherapy: past, present and future. Nat Med 2003; 9(3): 269-77.
[http://dx.doi.org/10.1038/nm0303-269] [PMID: 12612576]
[91]
Wang J. Combination treatment of cervical cancer using folate-decorated, pH-sensitive, carboplatin and paclitaxel co-loaded lipid-polymer hybrid nanoparticles. Drug Des Devel Ther 2020; 14: 823-32.
[http://dx.doi.org/10.2147/DDDT.S235098] [PMID: 32161442]
[92]
Kazi J, Mukhopadhyay R, Sen R, Jha T, Ganguly S, Debnath MC. Design of 5-fluorouracil (5-FU) loaded, folate conjugated peptide linked nanoparticles, a potential new drug carrier for selective targeting of tumor cells. MedChemComm 2019; 10(4): 559-72.
[http://dx.doi.org/10.1039/C8MD00565F] [PMID: 31057736]
[93]
Cai L, Yu R, Hao X, Ding X. Folate Receptor-targeted bioflavonoid genistein-loaded chitosan nanoparticles for enhanced anticancer effect in cervical cancers. Nanoscale Res Lett 2017; 12(1): 509.
[http://dx.doi.org/10.1186/s11671-017-2253-z] [PMID: 28853026]
[94]
Deshpande P, Jhaveri A, Pattni B, Biswas S, Torchilin V. Transferrin and octaarginine modified dual-functional liposomes with improved cancer cell targeting and enhanced intracellular delivery for the treatment of ovarian cancer. Drug Deliv 2018; 25(1): 517-32.
[http://dx.doi.org/10.1080/10717544.2018.1435747] [PMID: 29433357]
[95]
Saxena M, Delgado Y, Sharma RK, et al. Inducing cell death in vitro in cancer cells by targeted delivery of cytochrome c via a transferrin conjugate. PLoS One 2018; 13(4)e0195542
[http://dx.doi.org/10.1371/journal.pone.0195542] [PMID: 29649293]
[96]
Kuemmel S, Thomas A, Landt S, et al. Circulating vascular endothelial growth factors and their soluble receptors in pre-invasive, invasive and recurrent cervical cancer. Anticancer Res 2009; 29(2): 641-5.
[PMID: 19331214]
[97]
Randall LM, Monk BJ, Darcy KM, et al. Markers of angiogenesis in high-risk, early-stage cervical cancer: A Gynecologic Oncology Group study. Gynecol Oncol 2009; 112(3): 583-9.
[http://dx.doi.org/10.1016/j.ygyno.2008.11.013] [PMID: 19110305]
[98]
Cheng WF, Chen CA, Lee CN, Wei LH, Hsieh FJ, Hsieh CY. Vascular endothelial growth factor and prognosis of cervical carcinoma. Obstet Gynecol 2000; 96(5 Pt 1): 721-6.
[PMID: 11042307]
[99]
Meira DD, de Almeida VH, Mororó JS, et al. Combination of cetuximab with chemoradiation, trastuzumab or MAPK inhibitors: mechanisms of sensitisation of cervical cancer cells. Br J Cancer 2009; 101(5): 782-91.
[http://dx.doi.org/10.1038/sj.bjc.6605216] [PMID: 19654571]
[100]
Jiang Z, Albanese J, Kesterson J, et al. Monoclonal antibodies against human papillomavirus E6 and E7 oncoproteins inhibit tumor growth in experimental cervical cancer. Transl Oncol 2019; 12(10): 1289-95.
[http://dx.doi.org/10.1016/j.tranon.2019.06.003] [PMID: 31325765]
[101]
Goldstein D, Sader O, Benita S. Influence of oil droplet surface charge on the performance of antibody--emulsion conjugates. Biomed Pharmacother 2007; 61(1): 97-103.
[http://dx.doi.org/10.1016/j.biopha.2006.08.005] [PMID: 17011740]
[102]
Lundberg BB, Griffiths G, Hansen HJ. Conjugation of an anti-B-cell lymphoma monoclonal antibody, LL2, to long-circulating drug-carrier lipid emulsions. J Pharm Pharmacol 1999; 51(10): 1099-105.
[http://dx.doi.org/10.1211/0022357991776787] [PMID: 10579680]
[103]
Lundberg BB, Griffiths G, Hansen HJ. Cellular association and cytotoxicity of anti-CD74-targeted lipid drug-carriers in B lymphoma cells. J Control Release 2004; 94(1): 155-61.
[http://dx.doi.org/10.1016/j.jconrel.2003.09.016] [PMID: 14684279]
[104]
Balzar M, Winter MJ, de Boer CJ, Litvinov SV. The biology of the 17-1A antigen (Ep-CAM). J Mol Med (Berl) 1999; 77(10): 699-712.
[http://dx.doi.org/10.1007/s001099900038] [PMID: 10606205]
[105]
Chantima W, Thepthai C, Cheunsuchon P, Dharakul T. EpCAM expression in squamous cell carcinoma of the uterine cervix detected by monoclonal antibody to the membrane-proximal part of EpCAM. BMC Cancer 2017; 17(1): 811.
[http://dx.doi.org/10.1186/s12885-017-3798-z] [PMID: 29202724]
[106]
Lieder A, Khan MK, Lippert BM. Photodynamic therapy for recurrent respiratory papillomatosis. Cochrane Database Syst Rev 2014; 6(6)CD009810
[PMID: 24898010]
[107]
Cheng Y, Cheng H, Jiang C, et al. Perfluorocarbon nanoparticles enhance reactive oxygen levels and tumour growth inhibition in photodynamic therapy. Nat Commun 2015; 6: 8785.
[http://dx.doi.org/10.1038/ncomms9785] [PMID: 26525216]
[108]
Gong H, Chao Y, Xiang J, et al. Hyaluronidase to enhance nanoparticle-based photodynamic tumor therapy. Nano Lett 2016; 16(4): 2512-21.
[http://dx.doi.org/10.1021/acs.nanolett.6b00068] [PMID: 27022664]
[109]
Szpringer E, Lutnicki K, Marciniak A. Photodynamic therapy--mechanism and employment. Ann Univ Mariae Curie Sklodowska Med 2004; 59(2): 498-502.
[PMID: 16146137]
[110]
Inada NM, Buzzá HH, Leite MFM, et al. Long term effectiveness of photodynamic therapy for CIN treatment. Pharmaceuticals (Basel) 2019; 12(3)E107
[http://dx.doi.org/10.3390/ph12030107] [PMID: 31336848]
[111]
Yang D, Yang G, Wang X, et al. Y2O3:Yb,Er@mSiO2-Cu(x)S double-shelled hollow spheres for enhanced chemo-/photothermal anti-cancer therapy and dual-modal imaging. Nanoscale 2015; 7(28): 12180-91.
[http://dx.doi.org/10.1039/C5NR02269J] [PMID: 26132588]
[112]
Sun MM, Peng D, Hao HJ, et al. Thermally triggered in situ assembly of gold nanoparticles for cancer multimodal imaging and photothermal therapy ACS Appl Mater Interfaces 2017; 91:: 0453-10460..
[113]
Sweeney EE, Burga RA, Li C, Zhu Y, Fernandes R. Photothermal therapy improves the efficacy of a MEK inhibitor in neurofibromatosis type 1-associated malignant peripheral nerve sheath tumors. Sci Rep 2016; 6: 37035.
[http://dx.doi.org/10.1038/srep37035] [PMID: 27833160]
[114]
Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release 2000; 65(1-2): 271-84.
[http://dx.doi.org/10.1016/S0168-3659(99)00248-5] [PMID: 10699287]
[115]
Stern PL, van der Burg SH, Hampson IN, et al. Therapy of human papillomavirus-related disease. Vaccine 2012; 30(5)(Suppl. 5): F71-82.
[http://dx.doi.org/10.1016/j.vaccine.2012.05.091] [PMID: 23199967]
[116]
Olivo M, Bhuvaneswari R, Lucky SS, Dendukuri N, Soo-Ping Thong P. Targeted therapy of cancer using photodynamic therapy in combination with multi-faceted anti-tumor modalities. Pharmaceuticals (Basel) 2010; 3(5): 1507-29.
[http://dx.doi.org/10.3390/ph3051507] [PMID: 27713315]
[117]
Feng Q, Zhang Y, Zhang W, et al. Long-term results of intraoperative electron beam radiation therapy for nonmetastatic locally advanced pancreatic cancer: Retrospective cohort study, 7-year experience with 247 patients at the National Cancer Center in China. Acta Biomater 2017; 49: 402-13.
[http://dx.doi.org/10.1016/j.actbio.2016.11.035] [PMID: 27890732]
[118]
Wolinsky JB, Colson YL, Grinstaff MW. Local drug delivery strategies for cancer treatment: Gels, nanoparticles, polymeric films, rods, and wafers. J Control Release 2012; 159(1): 14-26.
[http://dx.doi.org/10.1016/j.jconrel.2011.11.031] [PMID: 22154931]
[119]
Friend DR. Intravaginal rings: controlled release systems for contraception and prevention of transmission of sexually transmitted infections. Drug Deliv Transl Res 2011; 1(3): 185-93.
[http://dx.doi.org/10.1007/s13346-011-0024-4] [PMID: 25788239]
[120]
Malcolm RK, Fetherston SM, McCoy CF, Boyd P, Major I. Vaginal rings for delivery of HIV microbicides. Int J Womens Health 2012; 4: 595-605.
[http://dx.doi.org/10.2147/IJWH.S36282] [PMID: 23204872]
[121]
Garg S, Goldman D, Krumme M, Rohan LC, Smoot S, Friend DR. Advances in development, scale-up and manufacturing of microbicide gels, films, and tablets. Antiviral Res 2010; 88(Suppl. 1): S19-29.
[http://dx.doi.org/10.1016/j.antiviral.2010.09.010] [PMID: 21109064]
[122]
McConville C, Major I, Friend DR, Clark MR, Malcolm RK. Development of a UC781 releasing polyethylene vinyl acetate vaginal ring. Drug Deliv Transl Res 2012; 2(6): 489-97.
[http://dx.doi.org/10.1007/s13346-012-0101-3] [PMID: 25787327]
[123]
Major I, Boyd P, Kilbourne-Brook M, Saxon G, Cohen J, Malcolm RK. A modified SILCS contraceptive diaphragm for long-term controlled release of the HIV microbicide dapivirine. Contraception 2013; 88(1): 58-66.
[http://dx.doi.org/10.1016/j.contraception.2012.10.018] [PMID: 23177261]
[124]
Koeneman MM, Kruse AJ, Kooreman LF, et al. Preliminary stop of the TOPical Imiquimod treatment of high-grade Cervical intraepithelial neoplasia (TOPIC) trial. BMC Cancer 2017; 17(1): 110.
[http://dx.doi.org/10.1186/s12885-017-3108-9] [PMID: 28173776]
[125]
Jaiswal M, Dudhe R, Sharma PK. Nanoemulsion: an advanced mode of drug delivery system. 3 Biotech 2015; 5(2): 123-7.
[http://dx.doi.org/10.1007/s13205-014-0214-0] [PMID: 28324579]
[126]
Alkhatib MH. AlMotwaa1 S.M, Alkreathy H.M. Incorporation of ifosfamide into various essential oils -based nanoemulsions ameliorates its apoptotic efect in the cancers cells. Sci Rep 2019; 9(695): 1-10.
[127]
Thomas N, Holm R, Rades T, Müllertz A. Characterising lipid lipolysis and its implication in lipid-based formulation development. AAPS J 2012; 14(4): 860-71.
[http://dx.doi.org/10.1208/s12248-012-9398-6] [PMID: 22956477]
[128]
Chang TC, Lai CH, Hong JH, et al. Randomized trial of neoadjuvant cisplatin, vincristine, bleomycin, and radical hysterectomy versus radiation therapy for bulky stage IB and IIA cervical cancer. J Clin Oncol 2000; 18(8): 1740-7.
[http://dx.doi.org/10.1200/JCO.2000.18.8.1740] [PMID: 10764435]
[129]
Rahangdale L, Lippmann QK, Garcia K, Budwit D, Smith JS, van Le L. Topical 5-fluorouracil for treatment of cervical intraepithelial neoplasia 2: a randomized controlled trial. Am J Obstet Gynecol 2014; 210(4): 314.e1-8.
[http://dx.doi.org/10.1016/j.ajog.2013.12.042] [PMID: 24384495]
[130]
AlMotwaa SM, Alkhatib MH, Alkreathy HM. Nanoemulsion-based camphor oil carrying Ifosfamide: preparation, characterization, and in-vitro evaluation in cancer cells. Int J Pharm Sci Res 2019; 10(4): 2018-26.
[131]
Pouton CW. Lipid formulations for oral administration of drugs: Nonemulsifying, self-emulsifying and self-microemulsifying. Eur J Pharm Sci 2000; 11: 93-8.
[http://dx.doi.org/10.1016/S0928-0987(00)00167-6]
[132]
Ujhelyi Z, Kalantari A, Vecsernyés M, et al. The enhanced inhibitory effect of different antitumor agents in self-microemulsifying drug delivery systems on human cervical cancer HeLa cells. Molecules 2015; 20(7): 13226-39.
[http://dx.doi.org/10.3390/molecules200713226] [PMID: 26197311]
[133]
Burdick AD, Bility MT, Girroir EE, et al. Ligand activation of peroxisome proliferator-activated receptor-beta/delta(PPARbeta/delta) inhibits cell growth of human N/TERT-1 keratinocytes. Cell Signal 2007; 19(6): 1163-71.
[http://dx.doi.org/10.1016/j.cellsig.2006.12.007] [PMID: 17254750]
[134]
York M, Griffiths HA, Whittle E, Basketter DA. Evaluation of a human patch test for the identification and classification of skin irritation potential. Contact Dermat 1996; 34(3): 204-12.
[http://dx.doi.org/10.1111/j.1600-0536.1996.tb02175.x] [PMID: 8833466]
[135]
Breiden B, Gallala H, Doering T, Sandhoff K. Optimization of submerged keratinocyte cultures for the synthesis of barrier ceramides. Eur J Cell Biol 2007; 86(11-12): 657-73.
[http://dx.doi.org/10.1016/j.ejcb.2007.02.006] [PMID: 17714827]

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